Proceedings of The National Academy of Sciences NAS Colloquium The Neurobiology of Pain
Proceedings of The National Academy of Sciences NAS Colloquium The Neurobiology of Pain
XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
the authoritative version for attribution.
COLLOQUIUM SERIES ii
                                                                                                                                                                                                                                              In 1991, the National Academy of Sciences inaugurated a series of scientific colloquia, five or six of which are scheduled each year
                                                                                                                                                                                                                                         under the guidance of the NAS Councils Committee on Scientific Programs. Each colloquium addresses a scientific topic of broad and
                                                                                                                                                                                                                                         topical interest, cutting across two or more of the traditional disciplines. Typically two days long, colloquia are international in scope and
                                                                                                                                                                                                                                         bring together leading scientists in the field. Papers from colloquia are published in the Proceedings of the National Academy of Sciences
                                                                                                                                                                                                                                         (PNAS).
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                            National Academy of Sciences Colloquium Arnold and Mabel Beckman Center, Irvine
                                                                                                                                                                                                                                            The Neurobiology of Pain
                                                                                                                                                                                                                                            December 1113, 1998
                                                                                                                                                                                                                                            PROGRAM
                                                                                                                                                                                                                                            Friday, December 11, 1998
                                                                                                                                                                                                                                            Introduction
                                                                                                                                                                                                                                            Ronald Dubner, Colloquium Organizer
                                                                                                                                                                                                                                            Session I: Channels
                                                                                                                                                                                                                                            Chair and Discussion Leader John Hunter, Department of Analgesia, Roche Bioscience
                                                                                                                                                                                                                                            Stephen G. Waxman, Department of Neurology, Yale University School of Medicine
                                                                                                                                                                                                                                            Sodium Channels and the Pathophysiology of Pain
                                                                                                                                                                                                                                            Michael Gold, Department of Oral and Craniofacial Biological Sciences, University of Maryland Dental School
                                                                                                                                                                                                                                            TTX-R INa and Inflammatory Hyperalgesia
                                                                                                                                                                                                                                            Daniel Weinreich, Department of Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine
                                                                                                                                                                                                                                            Which Potassium Channels Keep Vagal Afferent Neurons Mellow?
                                                                                                                                                                                                                                            Discussant Peter McNaughton, Department of Physiology, Kings College London
                                                                                                                                                                                                                                            Heat-Activated Ion Currents in NociceptorsTransduction and Sensitization
                                                                                                                                                                                                                                            Session II: Receptors
                                                                                                                                                                                                                                            Chair and Discussion Leader Patrick Mantyh, Department of Preventive Sciences, School of Dentistry, University of Minnesota
                                                                                                                                                                                                                                            Amy B. MacDermott, Department of Physiology and Cellular Biophysics and Center for Neurobiology and Behavior, Columbia
                                                                                                                                                                                                                                            University
                                                                                                                                                                                                                                            AMPA and Kainate Receptor Expression by DRG Neurons in Culture
                                                                                                                                                                                                                                            Edwin W. McCleskey, Vollum Institute, Oregon Health Sciences University
                                                                                                                                                                                                                                            The Role of Sensory Modality-Selective Gene Transcription in Opioid Analgesia
                                                                                                                                                                                                                                            Michael W. Salter, Programmes in Brain and Behavior and Cell Biology, Hospital for Sick Children, and Department of
                                                                                                                                                                                                                                            Physiology, University of Toronto
                                                                                                                                                                                                                                            NMDA Receptors and Src in Synaptic Plasticity
                                                                                                                                                                                                                                            Discussant Edward R. Perl, Department of Physiology, University of North Carolina School of Medicine, Chapel Hill
                                                                                                                                                                                                                                            Receptor Expression and Regulation as Mechanisms Underlying Pain and Pain Pathology
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
COLLOQUIUM SERIES iv
COLLOQUIUM SERIES v
                                                                                                                                                                                                                                             Lisa Aimone
                                                                                                                                                                                                                                                                                          List of Attendees
                                                                                                                                                                                                                                             George S. Aitken, Delta Medical Dental Centre
                                                                                                                                                                                                                                             David J. Anderson, California Institute of Technology
                                                                                                                                                                                                                                             David Andrew, Barrow Neurological Institute
                                                                                                                                                                                                                                             K. Roger Aoki, Allergan
                                                                                                                                                                                                                                             Dietrich Arndts, Boehringer Ingelheim
                                                                                                                                                                                                                                             Ronald K. Ashley
                                                                                                                                                                                                                                             Laura Audell
                                                                                                                                                                                                                                             Victor Babenko, Aalborg University
                                                                                                                                                                                                                                             Leif K. Bakland, Loma Linda University School of Dentistry
                                                                                                                                                                                                                                             Allan Basbaum, University of California, San Francisco
                                                                                                                                                                                                                                             Gary Bennett, MCP Hahnemann University
                                                                                                                                                                                                                                             Jack M. Berger, University of Southern California School of Medicine
                                                                                                                                                                                                                                             Scott Bowersox, Elan Pharmaceuticals
                                                                                                                                                                                                                                             Walter Bowles, University of Minnesota
                                                                                                                                                                                                                                             Robert Brennan, University of California, Los Angeles
                                                                                                                                                                                                                                             Cathy Bushnell, McGill University
                                                                                                                                                                                                                                             James Campbell, John Hopkins University
                                                                                                                                                                                                                                             Yuqing Cao, University of California, San Francisco
                                                                                                                                                                                                                                             Ken Casey, University of Michigan
                                                                                                                                                                                                                                             Lin Chang, University of California, Los Angeles
                                                                                                                                                                                                                                             Jenny Chen, University of California, Los Angeles
                                                                                                                                                                                                                                             Zhou-Feng Chen, California Institute of Technology
                                                                                                                                                                                                                                             Ken Chow, Allergan
                                                                                                                                                                                                                                             Glenn Clark, University of California, Los Angeles
                                                                                                                                                                                                                                             Patricia Claude, University of Texas, Health Science Center at San Antonio
                                                                                                                                                                                                                                             Amy D. Clegg
                                                                                                                                                                                                                                             Joseph R. Cohen, University of California, Los Angeles
                                                                                                                                                                                                                                             Sean P. Cook, Oregon Health Sciences University-Vollum Institute, L-474
                                                                                                                                                                                                                                             Santosh Coutinho, University of California, Los Angeles
                                                                                                                                                                                                                                             Marie Csete, California Institute of Technology
                                                                                                                                                                                                                                             Minglei Cui, Allergan
                                                                                                                                                                                                                                             Bennet Davis
                                                                                                                                                                                                                                             Xinzhong Dong, California Institute of Technology
                                                                                                                                                                                                                                             Emma Dormand, California Institute of Technology
                                                                                                                                                                                                                                             Ronald Dubner, University of Maryland Dental School
                                                                                                                                                                                                                                             Helena Ennes, University of California, Los Angeles
                                                                                                                                                                                                                                             Mark Erlander, RW Johnson Pharmalogical Research Institute
                                                                                                                                                                                                                                             Stephen EspitiaJack L. Feldman, University of California,
                                                                                                                                                                                                                                             Los Angeles
                                                                                                                                                                                                                                             Howard Fields, University of California, San Francisco
                                                                                                                                                                                                                                             Maria Fitzgerald, University College London
                                                                                                                                                                                                                                             Christopher M. Flores, University of Texas
                                                                                                                                                                                                                                             Nicholas Fuller, Cedars-Sinai Medical Center-The Pain Center
                                                                                                                                                                                                                                             Gerald Gebhart, University of Iowa
                                                                                                                                                                                                                                             Daniel W. Gil, Allergan, Inc.
                                                                                                                                                                                                                                             Michael Gold, University of Maryland Dental School
                                                                                                                                                                                                                                             Jay A. Goldstein
                                                                                                                                                                                                                                             Robert Grimm
                                                                                                                                                                                                                                             Paul Haerich, Loma Linda University
                                                                                                                                                                                                                                             Aldric Hama
                                                                                                                                                                                                                                             Donna Hammond, University of Chicago
                                                                                                                                                                                                                                             Michael Hans
                                                                                                                                                                                                                                             Kenneth Hargreaves, University of Texas, Health Science Center at San Antonio
                                                                                                                                                                                                                                             Michael Hayward, Oregon Health Sciences University
                                                                                                                                                                                                                                             Standiford Helm
                                                                                                                                                                                                                                             David Helton, B. Braun Medical Inc.
                                                                                                                                                                                                                                             Michael A. Henry, University of Colorado Health Science Center
                                                                                                                                                                                                                                             H. Haydon Hill, Rehabilitation Medicine Associates
                                                                                                                                                                                                                                             Dianne Hodges, CoCensys, Inc.
                                                                                                                                                                                                                                             Lynda C. Honour
                                                                                                                                                                                                                                             John Hunter, Roche
                                                                                                                                                                                                                                             Victor Ilyin, CoCensys, Inc
                                                                                                                                                                                                                                             Charles Inturrisi, Cornell University
                                                                                                                                                                                                                                             Douglass L. Jackson, University of Washington-School of Dentistry
the authoritative version for attribution.
TABLE OF CONTENTS ix
Table of Contents
                                                                                                                                                                                                                                            A comparison of the potential role of the tetrodotoxin-insensitive sodium channels, PN3/SNS and NaN/SNS2,   76407644
                                                                                                                                                                                                                                               in rat models of chronic pain
                                                                                                                                                                                                                                               Frank Porreca, Josephine Lai, Di Bian, Sandra Wegert, Michael H. Ossipov, Richard M. Eglen, Laura Kas-
                                                                                                                                                                                                                                               sotakis, Sanja Novakovic, Douglas K. Rabert, Lakshmi Sangameswaran, and John C. Hunter
                                                                                                                                                                                                                                            A visceral pain pathway in the dorsal column of the spinal cord                                             76757679
                                                                                                                                                                                                                                               William D. Willis, Elie D. Al-Chaer, Michael J. Quast, and Karin N. Westlund
                                                                                                                                                                                                                                            The spinal biology in humans and animals of pain states generated by persistent small afferent input        76807686
                                                                                                                                                                                                                                               Tony L. Yaksh, Xiao-Ying Hua, Iveta Kalcheva, Natsuko Nozaki-Taguchi, and Martin Marsala
                                                                                                                                                                                                                                            Src, a molecular switch governing gain control of synaptic transmission mediated by N-methyl-D- aspartate   76977704
                                                                                                                                                                                                                                               receptors
                                                                                                                                                                                                                                               Xian-Min Yu and Michael W. Salter
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
TABLE OF CONTENTS x
                                                                                                                                                                                                                                            Brain-derived neurotrophic factor is an endogenous modulator of nociceptive responses in the spinal cord        77147718
                                                                                                                                                                                                                                               S. W. N. Thompson, D. L. H. Bennett, B. J. Kerr, E. J. Bradbury, and S. B. McMahon
                                                                                                                                                                                                                                            Transcriptional and posttranslational plasticity and the generation of inflammatory pain                        77237730
                                                                                                                                                                                                                                               Clifford J. Woolf and Michael Costigan
                                                                                                                                                                                                                                            Cellular mechanisms of neuropathic pain, morphine tolerance, and their interactions                             77317736
                                                                                                                                                                                                                                               David J. Mayer, Jianren Mao, Jason Holt, and Donald D. Price
                                                                                                                                                                                                                                            Does a neuroimmune interaction contribute to the genesis of painful peripheral neuropathies?                    77377738
                                                                                                                                                                                                                                              Gary J. Bennett
                                                                                                                                                                                                                                            The genetic mediation of individual differences in sensitivity to pain and its inhibition                       77447751
                                                                                                                                                                                                                                               Jeffrey S. Mogil
                                                                                                                                                                                                                                            The  opiate receptor as a candidate gene for pain: Polymorphisms, variations in expression, nociception, and   77527755
                                                                                                                                                                                                                                               opiate responses
                                                                                                                                                                                                                                               George R. Uhl, Ichiro Sora, and Zaijie Wang
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper is the introduction to the following papers, which were presented at the National Academy of Sciences colloquium
                                                                                                                                                                                                                                         The Neurobiology of Pain, held December 1113, 1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                               Peter McNaughton ( 7 ) reviewed the data collected in his laboratory demonstrating for the first time the existence of an ion channel
                                                                                                                                                                                                                                         specifically activated by heat. Pursuing observations indicating that bradykinin modulates the heat activated channel, McNaughton
                                                                                                                                                                                                                                         presented evidence implicating activation of the epsilon isoform of protein kinase C in this process. McNaughtons findings are of even
                                                                                                                                                                                                                                         greater interest because of the similarity of this ion channel to the properties of the recently cloned capsaicin/heat receptor by Julius and
                                                                                                                                                                                                                                         colleagues ( 8 ).
                                                                                                                                                                                                                                               Research on receptors involving the transduction, transmission, and modulation of nociceptive information is clearly one of the most
                                                                                                                                                                                                                                         exciting and rapidly advancing areas in the field of pain research today. With the molecular characterization of many of the receptors
                                                                                                                                                                                                                                         involved in the transmission of nociceptive stimuli as well as the cellular elements necessary for synaptic transmission, researchers have
                                                                                                                                                                                                                                         begun to piece together the essential elements necessary for the first steps ultimately leading to the perception of pain. Amy MacDermott
                                                                                                                                                                                                                                         started the session by describing results from recent experiments performed in her laboratory designed to investigate the role of
                                                                                                                                                                                                                                         presynaptic non-N-methyl-D-aspartate receptors at the first synapse in the nociceptive pathway. Utilizing a dorsal root ganglion neuron/
                                                                                                                                                                                                                                         dorsal horn neuron co-culture, MacDermott and her colleagues obtained evidence indicating dorsal root ganglion neurons express
                                                                                                                                                                                                                                         functional -amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid and kainate receptors. Importantly, activation of these receptors
                                                                                                                                                                                                                                         appears to influence glutamate release from the sensory neuron and therefore the activation of the dorsal horn neurons. Edwin McCleskey
                                                                                                                                                                                                                                         described an exciting series of experiments performed in his laboratory utilizing a combination of electrophysiology and single-cell PCR.
                                                                                                                                                                                                                                         Through beautifully controlled reverse transcriptionPCR reactions, McCleskey and his colleagues were able to determine the number of
                                                                                                                                                                                                                                         mRNA copies encoding the -opioid receptor in a single cell in which the presence of functional -opioid receptors had previously been
                                                                                                                                                                                                                                         investigated. Their results provide a mechanistic explanation for some perplexing aspects of opioid analgesia. Michael Salter ( 9 ) provided
                                                                                                                                                                                                                                         evidence supporting a revolutionary hypothesis concerning the cellular events underlying the development of long-term potentiation in the
                                                                                                                                                                                                                                         hippocampus and, by analogy, central sensitization of spinal cord dorsal horn neurons after tissue injury. Salters compelling evidence
                                                                                                                                                                                                                                         indicates that the initial steps underlying these two phenomena may involve an increase in the intracellular concentration of Na+, activation
                                                                                                                                                                                                                                         of the nonreceptor protein tyrosine kinase, Src, and the subsequent phosphorylation of N-methyl-D-aspartate receptors. Edward Perl ( 10 )
                                                                                                                                                                                                                                         pointed out that changes in the expression of receptors involved in the transmission of nociceptive stimuli may contribute to the
                                                                                                                                                                                                                                         pathophysiology of pain. Perl cited evidence obtained in his laboratory supporting the hypothesis that an increase and/or change in the
                                                                                                                                                                                                                                         expression of -adrenergic receptors present in sensory neurons is an underlying mechanism of adrenergic excitation of sensory neurons
                                                                                                                                                                                                                                         often observed after nerve injury.
                                                                                                                                                                                                                                               The session on imaging and systems neuroscience examined some of the most recent exciting findings on pain pathways and their
                                                                                                                                                                                                                                         modulation. William Willis reported on a new visceral pain pathway that ascends in the dorsal column of the spinal cord ( 11 ).
                                                                                                                                                                                                                                         Postsynaptic dorsal column neurons in the rat sacral spinal cord transmit visceral signals to the gracile nucleus, and this information is then
                                                                                                                                                                                                                                         relayed to the ventral posterior lateral thalamic nucleus. Functional MRI studies have revealed that dorsal column lesions eliminate blood
                                                                                                                                                                                                                                         volume changes in the thalamus produced by noxious pelvic visceral stimulation, suggesting the importance of this pathway. More studies
                                                                                                                                                                                                                                         are needed to determine the functional significance of the spinothalamic and spinoparabrachial visceral pathways in comparison with this
                                                                                                                                                                                                                                         newly discovered dorsal column pathway. Tony Yaksh ( 12 ) described studies with different animal models that support the importance of
                                                                                                                                                                                                                                         spinal cord processing in pain states; he emphasized the functional and pharmacological comparability of symptoms across species and
                                                                                                                                                                                                                                         pointed out that these models are an important source of information for the development of novel clinically relevant analgesics. Howard
                                                                                                                                                                                                                                         Fields described his elegant findings on specific brain stem networks involved in potent pain modulation, -opioid receptor agonists
                                                                                                                                                                                                                                         activate neurons in the periaqueductal gray and the rostral ventral medulla by inhibiting GABAergic inhibition. The behavioral
                                                                                                                                                                                                                                         antinociception and inhibition of dorsal horn neurons is mediated by the release in rostral ventral medulla of an endogenous opioid peptide
                                                                                                                                                                                                                                         acting at the -opioid receptor. Fields also presented exciting data indicating that -opioid receptor selective ligands have actions in rostral
                                                                                                                                                                                                                                         ventral medulla that oppose those of the -opioid receptor-selective ligands and block the antinociceptive effect of periaqueductal gray-
                                                                                                                                                                                                                                         administered morphine. Interestingly, this effect was only observed in male rats. Descending modulation systems were summarized by
                                                                                                                                                                                                                                         Gerry Gebhart ( 13 ), who provided evidence that supraspinal structures make a significant contribution to the development and
                                                                                                                                                                                                                                         maintenance of hyperalgesia associated with tissue injury. He suggested that persistent input engages spinobulbospinal facilitatory
                                                                                                                                                                                                                                         mechanisms that contribute to secondary hyperalgesia that occurs outside the site of injury. The findings by others of descending
                                                                                                                                                                                                                                         inhibitory systems contributing to hyperalgesia emphasizes the bimodal nature of these descending systems in the modulation of persistent
                                                                                                                                                                                                                                         pain. Ken Casey ( 14 ) described the role of forebrain mechanisms of pain in imaging studies in humans and reviewed convincing evidence
                                                                                                                                                                                                                                         that the perceived intensity of unilateral pain evoked by different inputs correlates with increases in regional cerebral blood flow in
                                                                                                                                                                                                                                         primarily five structures: bilaterally in the thalamus, the contralateral insula, the bilateral premotor cortex, the contralateral anterior
                                                                                                                                                                                                                                         cingulate, and the cerebellar vermis. In contrast, results on the role of primary somatosensory cortex are somewhat inconsistent. Cathy
                                                                                                                                                                                                                                         Bushnell ( 15 ) reviewed the factors contributing to this inconsistency including cognitive modulation, average-related degradation of
                                                                                                                                                                                                                                         signal due to anatomical variability in sulcal anatomy and differences in methodology. She provided behavioral evidence indicating that
                                                                                                                                                                                                                                         manipulations that altered pain discrimination altered activity in primary somatosensory processing regions of the cerebral cortex. In
                                                                                                                                                                                                                                         contrast, manipulation that preferentially altered the affective or motivational dimension of pain produced changes in the anterior cingulate
                                                                                                                                                                                                                                         cortex. The combined use of psychophysical testing and brain imaging in humans should help reveal the functional role of these different
                                                                                                                                                                                                                                         forebrain structures that have direct corticofugal projections to the thalamus, brain stem, and spinal cord and thereby modulate the pain
                                                                                                                                                                                                                                         experience at those levels.
                                                                                                                                                                                                                                               The role of trophic factors and cytokines in the development and maintenance of pain in response to various forms of tissue injury is
                                                                                                                                                                                                                                         an area of research that has virtually exploded in the last several years. William Snider opened the session by describing recent
                                                                                                                                                                                                                                         experiments performed in his laboratory designed to distinguish trophic influences of nerve growth factor (NGF) from its role in cell
                                                                                                                                                                                                                                         survival. Through the use of knockout mice, Snider and his colleagues obtained striking results suggesting that, although activation of the
                                                                                                                                                                                                                                         high affinity NGF receptor was necessary to establish proper innervation of peripheral targets, activation of this receptor was not necessary
                                                                                                                                                                                                                                         for the growth and guidance of central terminals. In addition to its role in development, NGF and other growth factors and cytokines have
                                                                                                                                                                                                                                         been shown to mediate pain and hyperalgesia associated with tissue injury. Lorne Mendell ( 16 ), the first to describe the
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         link between NGF and pain, presented results obtained from experiments performed in his laboratory identifying the mechanisms
                                                                                                                                                                                                                                         underlying the initial hyperalgesic response to NGF. The initial hyperalgesia in response to systemic or peripherally administered NGF
                                                                                                                                                                                                                                         depends on indirect mechanisms, specifically mast cell degranulation. Mendell presented resent evidence indicating that NGF also is
                                                                                                                                                                                                                                         capable of potentiating capsaicin-evoked currents in isolated sensory neurons. Utilizing this intriguing observation, Mendell presented a
                                                                                                                                                                                                                                         model that would account for the initial NGFinduced thermal hyperalgesia. Focusing on the interaction between the immune system and
                                                                                                                                                                                                                                         the nervous system, Linda Watkins ( 17 ) described additional pathways through which activation of the immune system results in changes
                                                                                                                                                                                                                                         in multiple sites throughout the nervous system. Watkins described the molecules involved in the signaling pathways as well as how
                                                                                                                                                                                                                                         activation of this system results in changes in behavior. Steve McMahon ( 18 ) brought trophic factors back to center stage with his
                                                                                                                                                                                                                                         summary of a growing body of data implicating a critical role for brain derived neurotrophic factor in the altered nociceptive processing
                                                                                                                                                                                                                                         observed in the presence of inflammation. Brain derived neurotrophic factor appears to function as a neurotransmitter/neuromodulator in
                                                                                                                                                                                                                                         the dorsal horn of the spinal cord, where it is released from the central terminals of small-caliber afferents and increases the excitability of
                                                                                                                                                                                                                                         dorsal horn neurons.
                                                                                                                                                                                                                                               The session on development and plasticity explored plasticity that occurs in the central nervous system after tissue and nerve injury.
                                                                                                                                                                                                                                         Maria Fitzgerald ( 19 ) reported on changes in the neonatal spinal cord that are not simply immature or incomplete versions of what occurs
                                                                                                                                                                                                                                         in the adult. Central sensitization occurs in the normal immature spinal cord in response to electrical stimulation of A  fibers whereas
                                                                                                                                                                                                                                         activity-induced plasticity in the adult spinal cord takes place only in response to C fiber strength stimulation, unless the dorsal horn is
                                                                                                                                                                                                                                         primed by previous peripheral injury. N-methyl-D-aspartate and -amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors are
                                                                                                                                                                                                                                         distributed in higher density in the neonatal cord, and the receptor subunit composition in the neonatal spinal cord maximizes non-N-
                                                                                                                                                                                                                                         methyl-D-aspartate calcium influx. Clifford Woolf ( 20 ) provided an outstanding, concise, and up-to-date review of activity-induced and
                                                                                                                                                                                                                                         signal-induced plasticity in sensory neurons after tissue and nerve injury. He showed the interaction of these mechanisms in the role of
                                                                                                                                                                                                                                         brain derived neurotrophic factor in the generation of central sensitization. Jianren Mao ( 21 ) suggested that hyperalgesia and morphine
                                                                                                                                                                                                                                         tolerance may be interrelated by common neural mechanisms involving excitatory amino acid receptor activation and subsequent
                                                                                                                                                                                                                                         intracellular events, such as protein kinase C translocation and nitric oxide production. This hypothesis is supported by experiments
                                                                                                                                                                                                                                         showing that hyperalgesia develops when animals are made tolerant to morphine and that both the hyperalgesia and morphine tolerance
                                                                                                                                                                                                                                         develop as a consequence of peripheral nerve injury. Gary Bennett ( 22 ) reported on a new model of inflammation in which a focal
                                                                                                                                                                                                                                         neuritis is produced in the rat sciatic nerve. The results suggest the presence of a neuroimmune interaction that occurs at the onset of nerve
                                                                                                                                                                                                                                         injury and contributes to the development of neuropathic pain.
                                                                                                                                                                                                                                               While the tools of molecular genetics were employed by many of the researchers who spoke throughout the colloquium, the issue was
                                                                                                                                                                                                                                         addressed formally in the final session. Alan Basbaum ( 23 ) discussed the use of knockout mice to investigate the role of specific
                                                                                                                                                                                                                                         receptors and second messengers in nociceptive processing. Basbaum eloquently illustrated how this powerful approach has shed new
                                                                                                                                                                                                                                         light on our understanding of the mechanisms of action of molecules such as substance P and the  isoform of protein kinase C. For
                                                                                                                                                                                                                                         example, the contribution of substance P and neurokinin A to central sensitization may be considerably less than previously suspected
                                                                                                                                                                                                                                         while, in contrast, activation of the  isoform of protein kinase C appears to be vital to the development of nerve injury-induced
                                                                                                                                                                                                                                         hyperexcitability of dorsal horn neurons. Michael Moskowitz focused the discussion somewhat by describing several approaches that have
                                                                                                                                                                                                                                         been employed in the study of migraine. Moskowitz reviewed the data implicating the involvement of a specific class of serotonin
                                                                                                                                                                                                                                         receptors in migraine headache. Based on an exciting series of functional MRI studies, Moskowitz demonstrated that brain metabolism
                                                                                                                                                                                                                                         and blood flow may be uncoupled before the onset of headache. In contrast to the approach utilized by many researchers attempting to
                                                                                                                                                                                                                                         identify a role for a specific protein in nociception (a bottom-up approach), Jeffrey Mogil ( 24 ) described a top-down approach in which
                                                                                                                                                                                                                                         genetic mapping may be employed to identify genes responsible for specific behavioral phenotypes. Such an approach is readily applied to
                                                                                                                                                                                                                                         pain research, where it provides a mechanism for the identification of unique molecules critically involved in nociceptive processing. To
                                                                                                                                                                                                                                         illustrate a case in point, Mogil described the identification of a specific serotonin receptor subtype involved in the expression of morphine
                                                                                                                                                                                                                                         analgesia. George Uhl ( 25 ) discussed the integration of the top-down and bottom-up approaches through experiments performed with the
                                                                                                                                                                                                                                         -opioid receptor knockout mouse as well as populations of humans. His results suggest that polymorphisms in the gene encoding the -
                                                                                                                                                                                                                                         opioid receptor may explain much of the variability observed among people with respect to their responsiveness to opiate analgesia.
                                                                                                                                                                                                                                         Identification of the underlying mechanisms controlling opioid responsiveness may enable the development of individualized treatment
                                                                                                                                                                                                                                         programs for ongoing pain.
                                                                                                                                                                                                                                               The exciting new advances in pain research emphasize the importance of this field of neuroscience. The neural apparatus responsible
                                                                                                                                                                                                                                         for the perception of pain includes mechanisms that clearly are prototypic components of all mammalian sensory systems. These
                                                                                                                                                                                                                                         mechanisms include specialized receptors, stimulus transduction mechanisms, ion channel modulation, rapid and slow activity involving
                                                                                                                                                                                                                                         excitatory and inhibitory transmitters and their receptors, amplification of relevant signals at peripheral and central nervous system sites
                                                                                                                                                                                                                                         utilizing activity-dependent and signal-dependent mechanisms of neuronal plasticity, and, finally, distributed processing of environmental
                                                                                                                                                                                                                                         signals and their interaction with learned memories at higher centers. The field of pain research has made giant steps in putting together
                                                                                                                                                                                                                                         important segments of this puzzle. It is, of course, the hope of all of us that these advances will lead to an improvement in the quality of
                                                                                                                                                                                                                                         life of acute and chronic pain sufferers.
                                                                                                                                                                                                                                         1. Terman, G. W. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76317634 .
                                                                                                                                                                                                                                         2. Waxman, S. G. , Dib-Hajj, S. , Cummins, T. R. & Black, J. A. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76357639 .
                                                                                                                                                                                                                                         3. Porreca, F. Lai, J. , Bian, D. , Wegert, S. , Ossipov, M. H. , Eglen, R. M. , Kassotakis, L. , Novakovic, S. , Rabert, D. K. , Sangameswaran, L. &
                                                                                                                                                                                                                                              Hunter, J. C. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76407644 .
                                                                                                                                                                                                                                         4. Dib-Hajj, S. D. , Tyrell, L. , Black, J. A. & Waxman, S. G. ( 1998 ) Proc. Natl. Acad. Sci. USA 95 , 89638968 .
                                                                                                                                                                                                                                         5. Gold, M. S. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76457649 .
                                                                                                                                                                                                                                         6. Cordoba-Rodriguez, R. , Moore, K. A , Kao, J. P. Y. & Weinreich, D. , ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76507657 .
                                                                                                                                                                                                                                         7. Cesare, P. , Moriondo, A. , Vellani, V. & McNaughton, P. A. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76587663 .
                                                                                                                                                                                                                                         8. Caterina, M. J. , Schumacher, M. A. , Tominaga, M. , Rosen, T. A. , Levine, J. D. & Julius. D. ( 1997 ) Nature (London) 389 , 816824 .
                                                                                                                                                                                                                                         9. Yu, X.-M. & Salter, M. W. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76977704 .
                                                                                                                                                                                                                                         10. Perl, E. R. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76647667 .
                                                                                                                                                                                                                                         11. Willis, W. D. , Al-Chaer, E. D. , Quast, M. J. & Westlund, K. N. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76757679 .
the authoritative version for attribution.
                                                                                                                                                                                                                                         12. Yaksh, T. L. , Hua, X.-Y. , Kalcheva, I. , Nozaki-Taguchi, N. & Marsala, M. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76807686 .
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         13. Urban, M. O. & Gebhart, G. F. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76877692 .
                                                                                                                                                                                                                                         14. Casey, K. L. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76687674 .
                                                                                                                                                                                                                                         15. Bushnell, M. C. , Duncan, G. H. , Hofbauer, R. K. , Ha, B. , Chen, J.-I. & Carrier, B. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 77057709 .
                                                                                                                                                                                                                                         16. Shu, X.-Q. & Mendell, L. M. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 76937696 .
                                                                                                                                                                                                                                         17. Watkins, L. R. & Maier, S. F. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 77107713 .
                                                                                                                                                                                                                                         18. Thompson, S. W. N. , Bennett, D. L. H. , Kerr, B. J. , Bradbury, E. J. & McMahon, S. B. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 77147718 .
                                                                                                                                                                                                                                         19. Fitzgerald, M. & Jennings, E. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 77197722 .
                                                                                                                                                                                                                                         20. Woolf, C. J. & Costigan, M. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 77237730 .
                                                                                                                                                                                                                                         21. Mayer, D. J. , Mao, J. , Holt, J. & Price, D. D. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 77317736 .
                                                                                                                                                                                                                                         22. Bennett, G. J. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 77377738 .
                                                                                                                                                                                                                                         23. Basbaum, A. I. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 77397743 .
                                                                                                                                                                                                                                         24. Mogil, J. S. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 77447751 .
                                                                                                                                                                                                                                         25. Uhl., G. R. , Sora, I. & Wang, Z. ( 1999 ) Proc. Natl. Acad. Sci. USA 96 , 77527755 .
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                              GREGORY W. TERMAN *
                                                                                                                                                                                                                                              Department of Anesthesiology and the Graduate Program in Neurobiology and Behavior, University of Washington, Seattle, WA
                                                                                                                                                                                                                                         98195
                                                                                                                                                                                                                                              Id like to begin by thanking those who have helped me prepare this tributecontributing pictures, stories, and/or moral support. This
                                                                                                                                                                                                                                         group includes many of Johns students and friends, his family, and especially those at the Louise M. Darling Biomedical Library at
                                                                                                                                                                                                                                         UCLA where the John C. Liebeskind History of Pain Collection is housed. In particular, Marcia Meldrum, who worked closely with John
                                                                                                                                                                                                                                         on the Pain Collection and, in 1995, took an oral history from him concerning his perspectives on his career, will be stolen from frequently
                                                                                                                                                                                                                                         here. Also, Russell Johnson spent days helping me wade through many of Johns papers on a recent visit to the library.
                                                                                                                                                                                                                                              My job is to address those of you who didnt know John Liebeskind; to give you a flavor for the importance of this man, not only for
                                                                                                                                                                                                                                         the study of the neurobiology of painthe topic of this conferencebut also in positively influencing innumerable lives he came in
                                                                                                                                                                                                                                         contact with and, literally, health care worldwidethough he never treated a patient.
                                                                                                                                                                                                                                              I arrived at UCLA for graduate school in the fall of 1980. I had decided to attend UCLA because my psychology teacher, after
                                                                                                                                                                                                                                         hearing John speak at a weekend seminar for undergraduate teachers on the East Coast, persuaded me that there was only one option for
                                                                                                                                                                                                                                         pain research trainingLiebeskind. Fig. 1 is a picture of the John Liebeskind I met and got to know as a student in his laboratorynotice
                                                                                                                                                                                                                                         the phone. John was never too busy for the phone or the people calling him on it. Although I actually enjoyed this characteristic once I left
                                                                                                                                                                                                                                         UCLA, while I was there I hated it. The other imposing inanimate object in his office was the bulletin board. Now, the bulletin board may
                                                                                                                                                                                                                                         have simply been a device John put up to give students he was supposed to be meeting with something to look at while he was on the
                                                                                                                                                                                                                                         phone. Regardless, Johns bulletin board was such a fixture that when Tim Cannon, a former student, constructed his Unofficial
                                                                                                                                                                                                                                         Liebeskind Lab Web Page a few years back, the whole bulletin board was lovingly reproduced. On it were pictures of his students (and
                                                                                                                                                                                                                                         several teachers), friends, and family, as well as favorite sayings and over 100 misspellings of his name collected from various sources
                                                                                                                                                                                                                                         over the years. Essentially, Johns bulletin board was a reflection of his two great loves, words and people.
                                                                                                                                                                                                                                              John was born in Waterbury, Connecticut in 1935, the son of a clothing store owner. His family encouraged his education, sending
                                                                                                                                                                                                                                         him to private school from 6th grade through college. It was in high school that he first developed his love of words, and in his oral history
                                                                                                                                                                                                                                         he recounts specific teachers that he felt molded his academic interests. In my opinion, even more telling were his summers from age 8
                                                                                                                                                                                                                                         until age 20, when he went off for several weeks each year to Camp Kennebec in Maine ( Fig. 2 ), first as a camper and then as a
                                                                                                                                                                                                                                         counselor. Even after he began to attend college at Harvard he would head to Camp Kennebec in the summer. John described himself as a
                                                                                                                                                                                                                                         good camp counselor; a teacher and mentor, helping younger kids; and psychologically aware. I wouldnt have understood in 1980 if
                                                                                                                                                                                                                                         you asked me how I liked Camp Liebeskind, and I am only now beginning to realize, with trainees of my own, the effort it takes to be a
                                                                                                                                                                                                                                         decent mentor. I remember fondly the group gatherings and his practice of inviting students not going home for holidays to his home for
                                                                                                                                                                                                                                         backyard barbecues (the company was great and the food wasuhwell done). He treated his students/campers as if they were his
                                                                                                                                                                                                                                         children, and the counselor made sure that once you visited Camp Liebeskind, you never left.
                                                                                                                                                                                                                                              John majored in social relations at Harvard, taking the minimum science courses required (History of Science by I. B. Cohen and The
                                                                                                                                                                                                                                         Science of Human Behavior by B. F. Skinner) before heading off to the University of Michigan to graduate school in clinical psychology
                                                                                                                                                                                                                                         in 1957. At Michigan, he switched fairly quickly from clinical to physiological psychology, searching for what he called more precise
                                                                                                                                                                                                                                         research. In his final 4 years of graduate school, he struggled with his inadequate science preparation and three different thesis advisors
                                                                                                                                                                                                                                         before obtaining his Ph.D. in the fall of 1962. In his oral history, he says that these experiences helped him to find peace in studying and
                                                                                                                                                                                                                                         to mold his attitudes toward mentoring. He cited his eventual thesis advisor, Steve Fox, as telling him, Whatever is good for you, John,
                                                                                                                                                                                                                                         and your career, is going to reflect back on me and is going to end up being good for me. John learned well. One of his students, Hanan
                                                                                                                                                                                                                                         Frenk, says of John, We were [each] the best student he ever had, until the next one needed a job [recommendation].
                                                                                                                                                                                                                                              After graduation, John stayed on at Michigan, teaching and working with Steve Fox for another year. It was during this time that he
                                                                                                                                                                                                                                         learned the basics of electrophysiology and decided to go to Paris to train with Madame Denise Albe-Fessard. There he studied muscle
                                                                                                                                                                                                                                         spindle afferent inputs to the cortex in monkeys. When he returned to the United States in the summer of 1965, he came back as one of
                                                                                                                                                                                                                                         few psychologists trained in electrophysiology and had several job offers to choose from before deciding to take a job at UCLA. He
                                                                                                                                                                                                                                         arrived to work at UCLA in January of 1966, shortly after the Melzack/Wall Gate Control theory of pain was published in late 1965. John
                                                                                                                                                                                                                                         was fascinated by this paper and began thinking of studying pain processes in his own new laboratory. His initial NIH grant proposal
                                                                                                                                                                                                                                         concerned the modulation of cortical nociceptive responses by learning. This grant was funded and though I am unaware of him ever
                                                                                                                                                                                                                                         having done any of the studies proposed in it, he kept that NIH grant, his only one, for the next 28 years before he closed his laboratory.
                                                                                                                                                                                                                                              As John settled in, in Los Angeles, he was heavily influenced by his collaborators. Selected reviews of his early work include, offers
                                                                                                                                                                                                                                         a valuable insight, a classic of its time, and [this] work blows my mind because it is so simple and so profound, and refer, of course,
                                                                                                                                                                                                                                         to his uncredited walk-on part in Melvin Van Peebles movie Sweet Sweetbacks Baadasssss Song. Having sat through this movie to get a
                                                                                                                                                                                                                                         glimpse of John, Im afraid I have to give it thumbs down. John may have agreed; it was the closest he would get to Hollywood stardom.
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 1. Dr. John Liebeskind at work in his office at UCLAon the phone, with his bulletin board in the background.
                                                                                                                                                                                                                                               Meanwhile, in the laboratory, Dave Mayer and other early students found some interesting effects of lesions in the midbrain of rats on
                                                                                                                                                                                                                                         their escape behaviors and began to investigate this relatively unstudied brain region in earnest. Among Johns papers, I found a 1971
                                                                                                                                                                                                                                         letter from the journal Science rejecting his manuscript demonstrating an analgesic effect of midbrain stimulation in the rat. John and Dave
                                                                                                                                                                                                                                         had seen a report by Reynolds suggesting an analgesic effect of midbrain stimulation, and while originally setting out to disprove the idea,
                                                                                                                                                                                                                                         ended up supporting it. John was close to a tenure decision at UCLA at this time and whether this extra pressure prompted him or not, he
                                                                                                                                                                                                                                         elected to call Science and ask for a second chance. He told the editor, Mr. Ringle, that he would rewrite the paper specifically for him,
                                                                                                                                                                                                                                         emphasizing the high points, and if Ringle didnt agree that this was fascinating science he would not trouble him further. To Ringles
                                                                                                                                                                                                                                         credit, he recognized the importance of the manuscript, now a Citation Classic, and published it. To Johns credit, in the rewrite, he argued
                                                                                                                                                                                                                                         persuasivelywhat he said was Daves ideathat these data demonstrated the existence of endogenous pain inhibitory systems. The
                                                                                                                                                                                                                                         articulation of this concept made this paper sing (as John would say) and almost certainly is what got it published in Science. For the rest
                                                                                                                                                                                                                                         of his career, John would save his special magic for the discussion (his favorite part of any paper); taking methods and results and turning
                                                                                                                                                                                                                                         out meaningadmittedly, sometimes, going just a bit beyond what the data actually demonstrated. Marcia Meldrum claims that, even as
                                                                                                                                                                                                                                         [John] pursued his long and creative career in science, he was a historian because he could see ideas and events in context and knew how
                                                                                                                                                                                                                                         to tell a story ). More cynically, one student joked that as a scientist, John would have made a wonderful used car salesman. Indeed, John
                                                                                                                                                                                                                                         was always quick to deflect credit for his accomplishments to his excellent students and, truthfully, he did only rarely enter the lab the
                                                                                                                                                                                                                                         last 25 years of his career. Nonetheless, his excellent students might only have been average elsewhere and, in fact, I am unaware of his
                                                                                                                                                                                                                                         ever having turned away any student. He published nine times in Science with seven different first authors. Among his personal papers I
                                                                                                                                                                                                                                         came upon a scribbled quotation attributed to T. S. Eliot, Where is the wisdom we have lost in knowledge? Where is the knowledge we
                                                                                                                                                                                                                                         have lost in information? John found wisdom in the information we brought to him from the lab. Table 1 outlines his scientific findings
                                                                                                                                                                                                                                         and the larger concepts they addressed. His early studies, for example, of stimulation-produced analgesia and his finding that naloxone
                                                                                                                                                                                                                                         blocked this phenomenon probably facilitated the discovery of endogenous opioid peptides. His work on natural ways of activating these
                                                                                                                                                                                                                                         pain-inhibitory systems, the influence of gender and other genetic factors on analgesic mechanisms, and pains inhibition of the immune
                                                                                                                                                                                                                                         system, as well as much of his other work, at their core, all suggested that pain is not just unpleasant but dangerous in some circumstances.
                                                                                                                                                                                                                                         His findings, themselves, were not always first in the literature, but the concepts were routinely ahead of their time, sometimes spawning
                                                                                                                                                                                                                                         entire fields of study.
                                                                                                                                                                                                                                               John loved the word heuristic. I believe his research was heuristic. John was always sure of the right words. As a result, he was a
                                                                                                                                                                                                                                         terrible pain to write with. An almost finished manuscript would come back marked beyond recognition. It was comforting then, looking
                                                                                                                                                                                                                                         through his papers, to uncover a heavily edited version of his Pain Can Kill editorial on which he was the sole author. He couldnt even
                                                                                                                                                                                                                                         write with himself, much less with others. He sure could schmooze with others, however. Doing important science and publishing it in
                                                                                                                                                                                                                                         highprofile journals was only a start for John. He would hit the road, spreading the gospel, and from those first publications in the early
                                                                                                                                                                                                                                         1970s he used his considerable political talents to encourage not just more pain research but better clinical pain management as well. He
                                                                                                                                                                                                                                         was present in 1973 at the famous Issaquah meeting where the International Association for the Study of Pain (IASP) was formed and the
                                                                                                                                                                                                                                         journal Pain was
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         proposed. He started the Western Pain Society virtually by himself and then was instrumental in the formation of the American Pain
                                                                                                                                                                                                                                         Society (APS). The pain-related organizational activities listed in Table 2 are selected from his C.V. and demonstrate the incredible time
                                                                                                                                                                                                                                         and energy John must have spent putting forward the cause of pain research at the national and international levels. The foundation of the
                                                                                                                                                                                                                                         pain societies by John, his good friend John Bonica, and others have fostered dialogue between basic and clinical scientists, setting the
                                                                                                                                                                                                                                         standard for medical research. Moreover, working with the World Health Organization, these societies have succeeded in advancing the
                                                                                                                                                                                                                                         cause of optimum pain management throughout the world. Such international educational and patient care successes have, of course,
                                                                                                                                                                                                                                         raised the bar here at home for more and better pain care and research, benefiting industry, academia, and clinical practice alike. John
                                                                                                                                                                                                                                         Liebeskind will not be able, as planned, to become president of the IASP next year. John Bonica is also dead. For those not already doing
                                                                                                                                                                                                                                         so, I would encourage all who profit from the work of the pain societies every time a significance section of a grant proposal is written, to
                                                                                                                                                                                                                                         consider contributing a portion of your efforts to these societies at other times of the year.
IASP, International Association for the Study of Pain; APS, American Pain Society.
                                                                                                                                                                                                                                               In the late spring of 1997, John again sought medical attention, this time for pain in his chest, and the tumor was found to have
                                                                                                                                                                                                                                         metastasized to his lungs and beyond. Shortly after this diagnosis, he sent an e-mail to his studentsapologizing that, because he was
                                                                                                                                                                                                                                         dying, he would not be able to attend a party we had scheduled for him in honor of his National Academy election. He went on to tell us of
                                                                                                                                                                                                                                         our importance to him in a love letter I have no plans to ever delete.
                                                                                                                                                                                                                                               John died on September 8th, 1997 at home with his family, in little painfortunately, he was spared that irony. As a student and
                                                                                                                                                                                                                                         friend, it has been heartening to see the outpouring of remorse from around the world over the past year at the loss of this honorable
                                                                                                                                                                                                                                         scholar, scientist, and statesman. It is fitting that we pay tribute to him at this conference today. Not long after learning of his poor
                                                                                                                                                                                                                                         prognosis, John e-mailed the Academy, writing in part: Dear Edward. Ive had some bad news about my health . . . . This news calls into
                                                                                                                                                                                                                                         serious question my ability to participate in the [National Academy of Sciences] colloquium Ron Dubner and I have been planning. I
                                                                                                                                                                                                                                         havent said anything to Ron yet, but Im telling you now to see if you can help me in one matter. Ron really and truly is the brains behind
                                                                                                                                                                                                                                         this colloquiumbelieve me. I initiated matters as an Academy member, but the first thing I did was bring Ron on board. My objective all
                                                                                                                                                                                                                                         along has been to do what I could as an Academy member to help promote the field of pain. So I hope that if I am unable to participate
                                                                                                                                                                                                                                         much longer in the planning and am not around for the colloquium itself that Ron . . . will be allowed to continue and actually hold it.
                                                                                                                                                                                                                                         Sorry for the bad news. John. Before he died, John received reassurances that this conference would go on without him.
                                                                                                                                                                                                                                               John Coleman Liebeskind, bringing together again, today, his two loves, words and people, for the benefit of the field of pain. Enjoy
                                                                                                                                                                                                                                         the meeting.
                                                                                                                                                                                                                                               I would like to thank Timothy Cannon, Deborah Colbern, Darryl Dearmore, Ronald Dubner, Michael Gold, Russell Johnson, James
                                                                                                                                                                                                                                         Lewis, Julia Liebeskind, Marcia Meldrum, Michael Morgan, Wendy Sternberg, and the Louise M. Darling Biomedical Library at UCLA
                                                                                                                                                                                                                                         (where the John C. Liebeskind History of Pain collection is housed) for their help in preparing this manuscript.
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                         izations and a persistent current elicited by slow depolarizations close to resting membrane potential ( 25 ); (ii) SNS/PN3, expressed
                                                                                                                                                                                                                                         preferentially in small DRG and trigeminal neurons, encodes a TTX-resistent sodium current ( 26 , 27 ); (iii) NaN, expressed preferentially
                                                                                                                                                                                                                                         in small and trigeminal neurons, exhibits an amino acid sequence that, although only 47% similar to SNS-PN3, predicts that it encodes a
                                                                                                                                                                                                                                         TTX-resistant sodium channel ( 28 ); and (iv) NaG, another putative sodium channel that was originally cloned from astrocytes and at first
                                                                                                                                                                                                                                         thought to be glial specific ( 29 ), is also preferentially expressed at high levels within DRG neurons ( 23 ) and at low levels within other
                                                                                                                                                                                                                                         neurons of neural crest origin but not within other neuronal types ( 30 ).
                                                                                                                                                                                                                                           FIG. 1. Sodium channel -subunit mRNAs visualized in sections from adult rat DRG by in situ hybridization with subtype-specific
                                                                                                                                                                                                                                           antisense riboprobes. mRNAs for -I, Na6, hNE/PN1, SNS, NaN, and NaG are present at moderate to high levels in DRG neurons.
                                                                                                                                                                                                                                           Hybridization signal is not present with sense riboprobes, e.g., for NaG (S). (Bar indicates 100 m.)
                                                                                                                                                                                                                                           FIG. 2. Restriction enzyme profile analysis of Na channel domain 1 reverse transcriptionPCR products from DRG. M lanes contain
                                                                                                                                                                                                                                           100-bp ladder marker. Lane 1 contains the amplification product from DRG cDNA. Lanes 29 show the result of cutting this DNA
                                                                                                                                                                                                                                           with EcoRV, EcoN1, AvaI, SphI, BamHI, AflII, XbaI, and EcoRI, which are specific to subunits -I, -II, -III, Na6, PN1, SNS, NaG,
                                                                                                                                                                                                                                           and NaN, respectively. Reproduced with permission from ref. 28 . (Copyright 1998, National Academy of Sciences, USA).
                                                                                                                                                                                                                                              Preferential expression of SNS/PN3 and NaN within small DRG neurons provides a molecular correlate for the observation ( 19 
                                                                                                                                                                                                                                         22 , 32 , 33 ) that these cells express several distinct sodium currents, including TTX-resistant sodium currents. A role for TTX-resistant
                                                                                                                                                                                                                                         sodium channels in action potential conduction along small diameter afferent fibers has been postulated ( 34 ), and TTX-resistant sodium
                                                                                                                                                                                                                                         potentials have, in fact, been recorded from unmyelinated C-fibers ( 35 ).
                                                                                                                                                                                                                                              Preferential expression of SNS/PN3 and NaN in small DRG neurons, which include nociceptive cells, and the demonstration of a role
                                                                                                                                                                                                                                         of TTX-resistant sodium currents in conduction along their axons, have suggested that these channels may represent unique targets for the
                                                                                                                                                                                                                                         pharmacologic treatment of pain. PN1 and NaG also may represent useful molecular targets for the pharmacologic manipulation of DRG
                                                                                                                                                                                                                                         neurons because of their preferential expression in these cells.
                                                                                                                                                                                                                                                           SODIUM CHANNEL GENE EXPRESSION IS ALTERED AFTER INJURY TO DRG NEURONS
                                                                                                                                                                                                                                               The first observations indicating that, in addition to production of excess channels, there is a switch in the type of channels produced
                                                                                                                                                                                                                                         after axonal injury were provided by Waxman et al. ( 36 ), who found a significant up-regulation of expression of the previously silent -
                                                                                                                                                                                                                                         III sodium channel gene in DRG neurons
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 3. Transcripts for sodium channel -III (A) are up-regulated, neurons after transection of their axons within the sciatic nerve.
                                                                                                                                                                                                                                           The and transcripts for SNS (B) and NaN (C) are down-regulated, in DRG micrographs (Right) show in situ hybridizations in control
                                                                                                                                                                                                                                           DRG, and at 57 days postaxotomy. Reverse transcriptionPCR (Left) shows products of coamplification of -III (A) and SNS (B)
                                                                                                                                                                                                                                           together with -actin transcripts in control (C) and axotomized (A) DRG (days postaxotomy indicated above gels in A and B), with
                                                                                                                                                                                                                                           computerenhanced images of amplification products shown below gels. Coamplification of NaN (392 bp) and glyceraldehyde-3-
                                                                                                                                                                                                                                           phosphate dehydrogenase (GAPDH) (606 bp) (C) shows decreased expression of NaN mRNA at 7 days postaxotomy (lanes 2, 4, and
                                                                                                                                                                                                                                           6) compared with controls (lanes 1, 3, and 5). A and B modified from ref. 37 ; C modified from ref. 28 . (Copyright 1998, National
                                                                                                                                                                                                                                           Academy of Sciences, USA).
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         after axotomy. This finding was followed by demonstration of down-regulation of the SNS/PN3 gene expression, which can persist as long
                                                                                                                                                                                                                                         as 210 days after axotomy ( 37 ), and of down-regulation of the NaN gene ( 28 ). These changes are illustrated in Fig. 3 .
                                                                                                                                                                                                                                           FIG. 4. TTX-resistant sodium currents in small DRG neurons are down-regulated after axotomy. (A and B, Left) Whole-cell patch-
                                                                                                                                                                                                                                           clamp recordings from representative control (A) and axotomized (B, 6 days postaxotomy) DRG neurons. Note the loss of the TTX-
                                                                                                                                                                                                                                           resistant slowly inactivating component of sodium current after axotomy. Steady-state inactivation curves (A and B, Right) show loss
                                                                                                                                                                                                                                           of a component characteristic of TTX-resistant currents. (C) Attenuation of TTX-resistant current persists for at least 60 days
                                                                                                                                                                                                                                           postaxotomy. (D) Cell capacitance, which provides a measure of cell size, does not change significantly after axotomy (modified
                                                                                                                                                                                                                                           from ref. 39 ).
                                                                                                                                                                                                                                                  PHYSIOLOGIC CHANGES ACCOMPANY ALTERED SODIUM GENE EXPRESSION AFTER DRG NEURON
                                                                                                                                                                                                                                                                                                             INJURY
                                                                                                                                                                                                                                              On the basis of the down-regulation of SNS/PN3 and NaN genes in DRG neurons after axonal transection, it would be expected that
                                                                                                                                                                                                                                         TTX-resistant sodium currents should be reduced in these cells after axotomy. Patch-clamp studies have demonstrated that, indeed, there is
                                                                                                                                                                                                                                         a loss of TTX-resistant sodium currents in DRG neurons after axonal transection ( 38 ); this down-regulation persists in small DRG
                                                                                                                                                                                                                                         neurons for at least 60 days ( 39 ), consistent with the long-lasting changes in gene expression that have been described ( 37 ) in these cells
                                                                                                                                                                                                                                         ( Fig. 4 ). In addition, as shown in Fig. 5 , there is a switch in the properties of the TTX-sensitive sodium currents in these cells after
                                                                                                                                                                                                                                         axotomy, with the emergence of a rapidly repriming current (i.e., a current that recovers rapidly from inactivation) ( 39 ). Cummins and
                                                                                                                                                                                                                                         Waxman ( 39 ) have suggested that the type III sodium channel is responsible for the rapidly repriming sodium current, but this conjecture
                                                                                                                                                                                                                                         remains to be proven.
                                                                                                                                                                                                                                              These changes may poise DRG neurons to fire spontaneously, or at inappropriately high frequencies, after injury. Increased sodium
                                                                                                                                                                                                                                         channel densities, in themselves, will tend to lower threshold ( 12 ). In addition, Rizzo et al. ( 40 ) have pointed out that the overlap
                                                                                                                                                                                                                                         between steady-state activation and inactivation curves, together with weak voltage dependence of TTX-resistant sodium channels may
                                                                                                                                                                                                                                         confer instability on the neuronal membrane. Coexpression of abnormal combinations of several types of channels, whose window
                                                                                                                                                                                                                                         currents can bracket each other, would be expected to permit subthreshold ocillations in voltage, supported by TTX-resistant channels, to
                                                                                                                                                                                                                                         cross-activate other sodium channels, thereby producing spontaneous activity ( 40 ). Cummins and Waxman ( 39 ) noted that, because the
                                                                                                                                                                                                                                         TTX-sensitive sodium current in DRG neurons after axotomy reprimes relatively rapidly, injured neurons would be expected to sustain
                                                                                                                                                                                                                                         higher firing frequencies. Moreover, if persistent currents participate in setting the resting potential, as demonstrated in optic nerve axons (
                                                                                                                                                                                                                                         41 ), loss of TTX-resistant currents in DRG neurons after axotomy could produce a hyperpolarizing shift in resting potential, which, by
                                                                                                                                                                                                                                         relieving resting inactivation, might increase the amount of TTX-sensitive sodium current available for electrogenesis.
                                                                                                                                                                                                                                                            NEUROTROPHINS MODULATE SODIUM CHANNEL EXPRESSION IN DRG NEURONS
                                                                                                                                                                                                                                              A number of studies have suggested that, in response to nerve or tissue injury, there are changes in synthesis or delivery of various
                                                                                                                                                                                                                                         neurotrophins to neurons. Early studies in culture demonstrated that nerve growth factor (NGF) can affect sodium channel expression in
                                                                                                                                                                                                                                         DRG neurons ( 42 , 43 ). Black et al. ( 44 ) showed that NGF, delivered directly to DRG cell bodies, acts to down-regulate -III mRNA
                                                                                                                                                                                                                                         and maintain high levels of SNS/PN3 mRNA expression in small DRG neurons in an in vitro model that mimics axotomy. Following up
                                                                                                                                                                                                                                         on these observations, Dib-Hajj et al. ( 45 ) studied small DRG neurons in vivo after axotomy and demonstrated that administration of
                                                                                                                                                                                                                                         exogenous NGF to the proximal nerve stump results in an up-regulation of TTX-resistant sodium current and of SNS/PN3 mRNA levels in
                                                                                                                                                                                                                                         small DRG neurons ( Fig. 6 ). These observations suggest that at least some of the changes
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 5. The kinetics of recovery from inactivation in TTX-sensitive sodium currents are different in axotomized DRG neurons. The
                                                                                                                                                                                                                                           graph shows recovery of TTX-sensitive sodium current from inactivation as a function of time in DRG neurons after axonal
                                                                                                                                                                                                                                           transection (6 and 22 days postaxotomy, results pooled) compared with uninjured controls. Note the leftward shift in the recovery
                                                                                                                                                                                                                                           curve. Modified from ref. 39 .
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
observed in DRG neurons after axotomy reflect loss of access to peripheral pools of neurotrophic factors.
                                                                                                                                                                                                                                           FIG. 6. Reverse transcriptionPCR (A), in situ hybridization (B), and patch-clamp recordings (C), showing partial rescue of SNS
                                                                                                                                                                                                                                           mRNA and TTX-resistant sodium currents in axotomized DRG neurons after delivery of NGF to the proximal nerve stump. (A)
                                                                                                                                                                                                                                           Coamplification of SNS (479 bp) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH) (666 bp) products in Ringers solution-
                                                                                                                                                                                                                                           treated axotomized DRG (lanes 1, 2, 5, and 6) and NGF-treated axotomized DRG (lanes 3, 4, 7, and 8). The graph shows the increase
                                                                                                                                                                                                                                           in SNS amplification product in NGF-treated DRG. (B) In situ hybridization showing down-regulation of SNS mRNA in DRG after
                                                                                                                                                                                                                                           axotomy (axotomy + Ringers solution compared with control), and the partial rescue of SNS mRNA by NGF. (C) Representative
                                                                                                                                                                                                                                           patch-clamp recordings showing partial rescue of slowly inactivating TTX-resistant sodium currents in axotomized DRG neurons
                                                                                                                                                                                                                                           after exposure to NGF. Corresponding steady-state inactivation curves are shown below the recordings. Modified from ref. 45 .
                                                                                                                                                                                                                                              Brain-derived growth factor has been studied and has been found not to alter sodium currents in DRG neurons, although it affects the
                                                                                                                                                                                                                                         expression of -aminobutyric acid receptormediated currents in these cells ( 46 ). Glial-derived growth factor has been found to modulate
                                                                                                                                                                                                                                         the expression of NaN in a subpopulation of small DRG neurons, which are known to express the ret receptor ( 53 ). Multiple
                                                                                                                                                                                                                                         neurotrophins and growth factors have effects on DRG neurons, and it is likely that sodium channel expression in these cells reflects
                                                                                                                                                                                                                                         combinatorial effects of multiple factors.
                                                                                                                                                                                                                                                                      SODIUM CHANNEL EXPRESSION IN INFLAMMATORY PAIN MODELS
                                                                                                                                                                                                                                               Several studies have demonstrated that inflammatory molecules such as prostaglandins and serotonin can modulate TTX-resistant
                                                                                                                                                                                                                                         sodium currents in DRG neurons ( 47 ), possibly acting through a cyclic AMP-protein kinase A cascade ( 48 ). However, the question, of
                                                                                                                                                                                                                                         whether sodium channel gene expression is affected in inflammatory models of pain had not been addressed. To understand the role of
                                                                                                                                                                                                                                         sodium channels in inflammatory pain, we have carried out studies in the carageenan inflammatory pain model in the rat ( 49 ). In these
                                                                                                                                                                                                                                         studies, carried out before our cloning of NaN, we focused on SNS/PN3 because its expression was known to be labile. Based on our
                                                                                                                                                                                                                                         previous observation in which we detected peak changes in SNS/PN3 mRNA 5 days after axotomy ( 37 ), we studied rats in the subacute
                                                                                                                                                                                                                                         phase, 4 days after injection of carageenan into the hind paw. As shown in Fig. 7 , these experiments demonstrated significantly increased
                                                                                                                                                                                                                                         SNS/PN3 mRNA expression in DRG neurons projecting to the inflamed limb, compared with DRG neurons from the contralateral side or
                                                                                                                                                                                                                                         naive (uninjected) controls. Moreover, our patch-clamp recordings demonstrated that the amplitude of the TTXresistant sodium current in
                                                                                                                                                                                                                                         small DRG neurons projecting to the inflamed limb was significantly larger than on the contralateral side 4 days postinjection (31.7  3.3
                                                                                                                                                                                                                                         vs. 20.0  2.1 nA). The TTX-resistant current density was also significantly increased in the carageenan-challenged DRG neurons.
                                                                                                                                                                                                                                         Consistent with these results, a persistent increase in sodium channel immunoreactivity is observed in DRG neurons within 24 hr of
                                                                                                                                                                                                                                         injection of complete Freunds adjuvant into their projection field and persists for at least 2 months ( 50 ). The mechanism responsible for
                                                                                                                                                                                                                                         this inflammation-associated change in sodium channel expression is not known. Interestingly, NGF normally is produced in peripheral
                                                                                                                                                                                                                                         target tissues by supporting cells that include fibroblasts, Schwann cells, and keratinocytes; NGF production is stimulated in immune cells,
                                                                                                                                                                                                                                         and increased NGF levels have been observed in the local area after treatment with inflammatory agents such as carageenan and Freunds
                                                                                                                                                                                                                                         adjuvant ( 51 , 52 ), raising the possibility that inflammation may indirectly trigger changes in sodium channel gene expression via
                                                                                                                                                                                                                                         changes in neurotrophin levels.
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 7. SNS mRNA levels and TTX-resistant sodium currents are increased 4 days after injection of carrageenan into the projection
                                                                                                                                                                                                                                           fields of DRG neurons. (Upper) In situ hybridization showing SNS mRNA in carrageenan-injected (A), contralateral control (B), and
                                                                                                                                                                                                                                           naive (C) DRG. Patch-clamp recordings (DF) do not reveal any change in voltage dependence of activation or steady-state
                                                                                                                                                                                                                                           inactivation of TTX-resistant sodium currents after carrageenan injection, but demonstrate an increase in TTX-resistant current
                                                                                                                                                                                                                                           amplitude (D) and density. Modified from ref. 49 .
                                                                                                                                                                                                                                                                      SODIUM CHANNELS AS MOLECULAR TARGETS IN PAIN RESEARCH
                                                                                                                                                                                                                                             Given what we have learned about sodium channels, where do we go next in the search for better treatments for pain syndromes? The
                                                                                                                                                                                                                                         answer to this question is not entirely clear at this time. We can, however, come to a number of conclusions. First, sodium channels are
                                                                                                                                                                                                                                         important participants in electro
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         genesis within primary sensory neurons, including DRG neurons. Second, a multiplicity of sodium channels are present within DRG
                                                                                                                                                                                                                                         neurons, where they probably subserve multiple functions (transduction, signal amplification, action potential electrogenesis, etc.) and
                                                                                                                                                                                                                                         interact in a complex manner. Third, DRG neurons express a number of sodium channel genes (SNS/PN3, NaN, PN1, and NaG) in a
                                                                                                                                                                                                                                         preferential manner, at levels much higher than in any other neuronal cell type. This observation may present a therapeutic opportunity for
                                                                                                                                                                                                                                         the selective manipulation of primary sensory neurons in general, or nociceptive neurons in particular. Fourth, sodium channel expression
                                                                                                                                                                                                                                         in DRG neurons is highly dynamic, with multiple sodium channel genes (including -III, SNS/PN3, and NaN) exhibiting up- or down-
                                                                                                                                                                                                                                         regulation after various injuries to these cells. Importantly, different injuries may trigger opposing changes of certain sodium channel
                                                                                                                                                                                                                                         genes (e.g., down-regulation of SNS/PN3 after axotomy vs. up-regulation in the carageenan inflammation model) in DRG neurons, so that
                                                                                                                                                                                                                                         it may be difficult to extrapolate from one model system to another. Nevertheless, we have learned, at a minimum, that sodium channel
                                                                                                                                                                                                                                         expression in DRG neurons is dynamic and can change significantly after injury, and that changes in sodium channel expression can
                                                                                                                                                                                                                                         substantially alter excitability in these cells.
                                                                                                                                                                                                                                              Delineation of the precise role(s) of each sodium channel subtype in the physiology of DRG neurons and the pathophysiology of pain
                                                                                                                                                                                                                                         remains to be established, and the utility of selective blockade of each channel subtype as an approach to the treatment of pain will require
                                                                                                                                                                                                                                         further careful study. However, the stage has been set for these investigations. It is quite likely, in our opinion, that sodium channel
                                                                                                                                                                                                                                         blockade will emerge as a viable strategy for pharmacologic treatment of pain.
                                                                                                                                                                                                                                              This work has been supported in part by grants from the National Multiple Sclerosis Society and the Paralyzed Veterans of America/
                                                                                                                                                                                                                                         Eastern Paralyzed Veterans Association, and by the Medical Research Service, Department of Veterans Affairs. T.R.C. was supported in
                                                                                                                                                                                                                                         part by a fellowship from the Spinal Cord Research Foundation.
                                                                                                                                                                                                                                         1. Ochoa, J. & Torebjork, H. E. ( 1980 ) Brain 103 , 835854 .
                                                                                                                                                                                                                                         2. Nordin, M. , Nystrom, B. , Wallin, U. & Hagbarth, K.-E. ( 1984 ) Pain 20 , 231245 .
                                                                                                                                                                                                                                         3. Devor, M. ( 1994 ) in Textbook of Pain , eds. Wall, P. D. & Melzack, R. ( Churchill Livingstone , Edinburgh ), 2nd Ed. , pp. 79101 .
                                                                                                                                                                                                                                         4. Eccles, J. C , Libet, B. & Young, R. R. ( 1958 ) J. Physiol. (London) 143 , 1140 .
                                                                                                                                                                                                                                         5. Kuno, M. & Llinas, R. ( 1970 ) J. Physiol. (London) 210 , 807821 .
                                                                                                                                                                                                                                         6. Gallego, R. , Ivorra, I. & Morales, A. ( 1987 ) J. Physiol. (London) 391 , 3956 .
                                                                                                                                                                                                                                         7. Gurtu, S. & Smith, P. A. ( 1988 ) J. Neurophysiol. 59 , 408423 .
                                                                                                                                                                                                                                         8. Devor, M. , Keller, C. H. , Deerinck, T. J. & Ellisman, M. H. ( 1989 ) Neurosci. Lett. 102 , 149154 .
                                                                                                                                                                                                                                         9. England, J. D. , Gamboni, F. , Ferguson, M. A. & Levinson, S. R. ( 1994 ) Muscle Nerve 17 , 593598 .
                                                                                                                                                                                                                                         10. England, J. D. , Happel, L. T. , Kline, D. G. , Gamboni, F. , Thouron, C. L. , Liu, Z. P. & Levinson, S. R. ( 1996 ) Neurology 47 , 272276 .
                                                                                                                                                                                                                                         11. Waxman, S. G. & Brill, M. H. ( 1978 ) J. Neurol . Neurosurg. Psychiatr . 41 , 408417 .
                                                                                                                                                                                                                                         12. Matzner, O. & Devor, M. ( 1992 ) Brain Res. 597 , 9298 .
                                                                                                                                                                                                                                         13. Matzner, O. & Devor, M. ( 1994 ) J. Neurophysiol. 72 , 349359 .
                                                                                                                                                                                                                                         14. Zhang, J.-M. , Donnelly, D. F. , Song, X.-J. & LaMotte, R. H. ( 1997 ) J. Neurophysiol. 78 , 27902794 .
                                                                                                                                                                                                                                         15. Chabal, C. , Russell, L. C. & Burchiel, K. J. ( 1989 ) Pain 38 , 333338 .
                                                                                                                                                                                                                                         16. Devor, M. , Wall, P. D. & Catalan, N. ( 1992 ) Pain 48 , 261268 .
                                                                                                                                                                                                                                         17. Omana-Zapata, I. , Khabbaz, M. A. , Hunter, J. C. & Bley, K. R. ( 1997 ) Brain Res. 771 , 228237 .
                                                                                                                                                                                                                                         18. Rizzo, M. A. ( 1997 ) J. Neurol. Sci. 152 , 103106 .
                                                                                                                                                                                                                                         19. Kostyuk, P. G. , Veselovsky, N. S. & Tsyandryenko, A. Y. ( 1981 ) Neuroscience 6 , 24232430 .
                                                                                                                                                                                                                                         20. Roy, M. L. & Narahashi, T. ( 1992 ) J. Neurosci. 12 , 21042111 .
                                                                                                                                                                                                                                         21. Caffrey, J. M. , Eng, D. L. , Black, J. A. , Waxman, S. G. & Kocsis, J. D. ( 1992 ) Brain Res. 592 , 283297 .
                                                                                                                                                                                                                                         22. Elliott, A. A. & Elliott, J. R. ( 1993 ) J. Physiol. (London) 463 , 3956 .
                                                                                                                                                                                                                                         23. Black, J. A. , Dib-Hajj, S. , McNabola, K. , Jeste, S. , Rizzo, M. A. , Kocsis, J. D. & Waxman, S. G. ( 1996 ) Mol. Brain Res. 43 , 117132 .
                                                                                                                                                                                                                                         24. Toledo-Aral, J. J. , Moss, B. L. , He, Z.J. , Koszowski, A. G. , Whisenand, T. , Levinson, S. R. , Wolff, J. J. , Silos-Santiago, I. , Halegoua, S. &
                                                                                                                                                                                                                                              Mandel, G. ( 1997 ) Proc. Natl. Acad. Sci. USA 94 , 15271532 .
                                                                                                                                                                                                                                         25. Cummins, T. R. , Howe, J. R. & Waxman, S. G. ( 1999 ) J. Neurosci. 18 , 96079619 .
                                                                                                                                                                                                                                         26. Akopian, A. N. , Sivilotti, L. & Wood, J. N. ( 1996 ) J. Biol. Chem. 271 , 59535956 .
                                                                                                                                                                                                                                         27. Sangameswaran, L. , Delgado, S. G. , Fish, L. M. , Koch, B. D. , Jakeman, L. B. , Stewart, G. R. , Sze, P. , Hunter, J. C. , Eglen, R. M. & Herman, R.
                                                                                                                                                                                                                                              C. ( 1996 ) J. Biol. Chem. 271 , 59535956 .
                                                                                                                                                                                                                                         28. Dib-Hajj, S. D. , Tyrrell, L. , Black, J. A. & Waxman, S. G. ( 1998 ) Proc. Natl. Acad. Sci. USA 95 , 89638969 .
                                                                                                                                                                                                                                         29. Gautron, S. , Dos Santos, G. , Pinto-Henrique, D. , Koulkoff, A. , Gros, F. & Berwald-Netter, Y. ( 1992 ) Proc. Natl. Acad. Sci. USA 89 , 72727276 .
                                                                                                                                                                                                                                         30. Felts, P. A. , Black, J. A. , Dib-Hajj, S. D. & Waxman, S. G. ( 1997 ) Glia 21 , 269277 .
                                                                                                                                                                                                                                         31. Rizzo, M. A. , Kocsis, J. D. & Waxman, S. G. ( 1995 ) J. Neurophysiol. 72 , 27962816 .
                                                                                                                                                                                                                                         32. Rush, A. M. , Brau, M. E. , Elliott, A. A. & Elliott, J. R. ( 1998 ) J. Physiol. (London) 511 , 771789 .
                                                                                                                                                                                                                                         33. Scholz, A. , Appel, N. & Vogel, W. ( 1998 ) Eur. J. Neurosci. 10 , 25472556 .
                                                                                                                                                                                                                                         34. Jeftinija, S. ( 1994 ) Brain Res. 639 , 125134 .
                                                                                                                                                                                                                                         35. Quasthoff, S. , Grosskreutz, J. , Schroder, J. M. , Schneider, U. & Grafe, P. ( 1995 ) Neuroscience 69 , 955965 .
                                                                                                                                                                                                                                         36. Waxman, S. G. , Kocsis, J. K. & Black, J. A. ( 1994 ) J. Neurophysiol. 72 , 466471 .
                                                                                                                                                                                                                                         37. Dib-Hajj, S. , Black, J. A. , Felts, P. & Waxman, S. G. ( 1996 ) Proc. Natl. Acad. Sci. USA 93 , 1495014954 .
                                                                                                                                                                                                                                         38. Rizzo, M. A. , Kocsis, J. D. & Waxman, S. G. ( 1995 ) Neurobiol. Dis. 2 , 8796 .
                                                                                                                                                                                                                                         39. Cummins, T. R. & Waxman, S. G. ( 1997 ) J. Neurosci. 17 , 35033514 .
                                                                                                                                                                                                                                         40. Rizzo, M. A. , Kocsis, J. D. & Waxman, S. G. ( 1996 ) Eur. Neurol. 36 , 312 .
                                                                                                                                                                                                                                         41. Stys, P. K. , Ransom, B. R. & Waxman, S. G. ( 1993 ) Proc. Natl. Acad. Sci. USA 90 , 69766980 .
                                                                                                                                                                                                                                         42. Aguayo, L. G. & White, G. ( 1992 ) Brain Res. 570 , 6167 .
                                                                                                                                                                                                                                         43. Zur, K. B. , Oh, Y. , Waxman, S. G. & Black, J. A. ( 1995 ) Mol. Brain Res. 30 , 97103 .
                                                                                                                                                                                                                                         44. Black, J. A. , Langworthy, K. , Hinson, A. W. , Dib-Hajj, S. D. & Waxman, S. G. ( 1997 ) NeuroReport 8 , 23312335 .
                                                                                                                                                                                                                                         45. Dib-Hajj, S. D. , Black, J. A. , Cummins, T. R. , Kenney, A. M. , Kocsis, J. D. & Waxman, S. G. ( 1998 ) J. Neurophysiol. 79 , 26682678 .
                                                                                                                                                                                                                                         46. Oyelese, A. A. , Rizzo, M. A. , Waxman, S. G. & Kocsis, J. D. ( 1997 ) J. Neurophysiol. 78 , 3142 .
                                                                                                                                                                                                                                         47. Gold, M. S. , Reichling, D. B. , Shuster, M. J. & Levine, J. D. ( 1996 ) Proc. Natl. Acad. Sci. USA 93 , 11081112 .
                                                                                                                                                                                                                                         48. England, S. , Bevan, S. & Docherty, R. J. ( 1996 ) J. Physiol. (London) 495 , 429440 .
                                                                                                                                                                                                                                         49. Tanaka, M. , Cummins, T. R. , Ishikawa, K. , Dib-Hajj, S. D. , Black, J. A. & Waxman, S. G. ( 1998 ) NeuroReport 9 , 967972 .
                                                                                                                                                                                                                                         50. Gould, H. J. III , England, J. D. , Liu, Z. P. & Levinson, S. R. ( 1998 ) Brain Res. 802 , 6974 .
the authoritative version for attribution.
                                                                                                                                                                                                                                         51. Woolf, C. J. , Safieh-Garabedian, B. , Ma, Q.-P., Crilly, P. & Winter, J. ( 1994 ) Neuroscience 62 , 327331 .
                                                                                                                                                                                                                                         52. Weskamp, G. & Otten, U. ( 1987 ) J. Neurochem. 48 , 17791786 .
                                                                                                                                                                                                                                         53. Fzell, J. , Cummins, P. R. , Dib-Hajj, S. D. , Fried, K. , Black, J. A. & Waxman, S. G. ( 1999 ) Mol. Brain Res. 67 , 267282 .
                                                                                                                                                                                                                                         A COMPARISON OF THE POTENTIAL ROLE OF THE TETRODOTOXIN-INSENSITIVE SODIUM CHANNELS, PN3/SNS AND NAN/                                        7640
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                         repetitive firing at the depolarized potentials characteristic of an injured peripheral nerve; that is, they will be slowly adapting in response
                                                                                                                                                                                                                                         to a persistent, depolarizing stimulus ( 10 , 21 , 24 ).
                                                                                                                                                                                                                                           FIG. 1. Immunohistochemical (peroxidase-diaminobenzidine) analysis of PN3 antibody labeling of L4 DRG cells from normal and
                                                                                                                                                                                                                                           SNL animals, 7 days post-surgery. (A) Naive animals show a predominant labeling of the small diameter cells. In contrast, after SNL
                                                                                                                                                                                                                                           injury (B), in animals that had received saline for 48 hr, a marked increase was observed in the number of large diameter cells
                                                                                                                                                                                                                                           expressing PN3 in the L4 DRG ipsilateral to the side of injury. Animals treated with mismatch (MM) ODN (C) for 48 hr (day 7 post-
                                                                                                                                                                                                                                           surgery) exhibited a similar pattern of labeling to the saline controls. In contrast, animals that had received antisense (AS) ODN (D)
                                                                                                                                                                                                                                           for a similar time period to the MM ODN demonstrated a marked loss of PN3 immunolabeling in both small and large cells of the L4
                                                                                                                                                                                                                                           DRG ipsilateral to the side of injury. (Bar = 50 m.)
                                                                                                                                                                                                                                               Immunohistochemical Studies. Such a potentially specialized, pathophysiological role for TTX-R INa/PN3 has been supported by
                                                                                                                                                                                                                                         alterations in channel distribution observed after sustained injury to the peripheral nerve. The effect, however, is complex and appears
                                                                                                                                                                                                                                         dependent on the nature and degree of the injury. After axotomy, TTX-R INa is substantially reduced in the small cells of the DRG ( 25 ,
                                                                                                                                                                                                                                         26 ) with the level of PN3/SNS mRNA ( 27 ) and protein ( 20 ) expression reduced in parallel. In contrast, in the chronic constriction
                                                                                                                                                                                                                                         injury model of neuropathic pain in the rat, there was no significant change in the amplitude or IV relationship of either TTX-R or TTX-S
                                                                                                                                                                                                                                         INa recorded from small-diameter DRG neurons at the maximum time of injury, i.e., 14 days post-surgery ( 20 ). This would suggest that
                                                                                                                                                                                                                                         the chronic constriction injury type of injury has had little impact on the number of sodium channels present in the somal membrane at any
                                                                                                                                                                                                                                         given time. However, an initial loss of PN3/SNS immunolabeling was observed from the DRG at all levels of the lumbar enlargement (L4-
                                                                                                                                                                                                                                         L6), presumably from the predominantly intracellular pool, and correlated closely with a subsequent redistribution and accumulation of
                                                                                                                                                                                                                                         channel protein within the peripheral nerve just proximal to the site of injury ( 20 ). The onset and subsequent reversal of PN3/SNS
                                                                                                                                                                                                                                         channel redistribution appeared to correlate closely with temporal changes in behavioral thermal hyperalgesia and, morphologically, with
                                                                                                                                                                                                                                         the damage and recovery of primary afferent fibers after this type of nerve ligation ( 28 ). A similar observation has been made in another
                                                                                                                                                                                                                                         rat model of peripheral nerve injury in which the L5 and L6 dorsal spinal roots are tightly ligated, evoking behavioral signs of hyperalgesia
                                                                                                                                                                                                                                         and allodynia ( 29 ). A loss of PN3/SNS protein was observed in the DRG at the level of L5 and L6 with a subsequent accumulation at the
                                                                                                                                                                                                                                         injury site proximal to the ligatures (P. Mantyh, personal communication). However, although the expression of PN3 protein in the L5 and
                                                                                                                                                                                                                                         L6 DRG decreases after spinal nerve ligation (SNL) injury, PN3 protein levels in the uninjured L4 DRG were preserved in small diameter
                                                                                                                                                                                                                                         cells and significantly increased in the large diameter cells ( Fig. 1 ). It is possible to speculate that the increased number of large diameter
                                                                                                                                                                                                                                         cells expressing PN3 protein after SNL may account, in part, for the observed tactile allodynia. Such observations provide additional
                                                                                                                                                                                                                                         support for the involvement of uninjured primary afferents in adjacent segments of the sciatic nerve in mediating certain types of
                                                                                                                                                                                                                                         neuropathic pain behaviors ( 30 ). The reasons for increased PN3 expression in large diameter cells in the non-injured L4 DRG after SNL
                                                                                                                                                                                                                                         and decreases in PN3 expression in cell bodies of the injured L5/L6 DRG are unclear but may reflect the differential availability of factors
                                                                                                                                                                                                                                         such as nerve growth factor, which is known to be released from peripheral tissues and to be retrogradely transported to the ganglion,
                                                                                                                                                                                                                                         where it regulates mRNA expression of PN3 ( 31 ) as well as other sodium channels ( 32 , 33 ). However, it should be emphasized that the
                                                                                                                                                                                                                                         extent to which the immunolabeling pattern observed in the L4 ganglion translates into axonal accumulation of the channel protein and/or
                                                                                                                                                                                                                                           FIG. 2. Antisense (AS), but not mismatch (MM), ODN to PN3 prevents and reverses tactile allodynia (A) and thermal hyperalgesia
                                                                                                                                                                                                                                           (B) after SNL. After determination of baseline (BL) responses, the rats received twice daily injections (45 g, intrathecal in a 5-l
                                                                                                                                                                                                                                           volume) of either MM (open symbols) or AS (filled symbols) (arrow a). After 5 days of ODN pretreatment, rats were subjected to
                                                                                                                                                                                                                                           either SNL (squares) or sham surgery (circles) (arrow b). The ODN injections were terminated after the afternoon injection on day
                                                                                                                                                                                                                                           5 (arrow c), recommenced on the morning of day 12 (arrow d), and were terminated again after the afternoon injection on day 15
                                                                                                                                                                                                                                           (arrow e). Tactile allodynia was indicated by a significant (P  0.05) reduction in paw withdrawal threshold to application of a
                                                                                                                                                                                                                                           series of calibrated (0.415 g) von Frey filaments to the plantar surface of the hindpaw. Thermal hyperalgesia was indicated by a
                                                                                                                                                                                                                                           significant (P  0.05) reduction in paw withdrawal latency to application of noxious radiant heat to the plantar surface of the affected
the authoritative version for attribution.
                                                                                                                                                                                                                                           hindpaw of the nerve or sham-operated rats. A maximal cut-off of 40 sec was used to prevent tissue damage. n = 6 rats per group.
                                                                                                                                                                                                                                         A COMPARISON OF THE POTENTIAL ROLE OF THE TETRODOTOXIN-INSENSITIVE SODIUM CHANNELS, PN3/SNS AND NAN/                                      7642
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         changes in the response thresholds to non-noxious sensory stimulation after nerve injury is unknown. In this regard, the possible
                                                                                                                                                                                                                                         accumulation of the PN3 channel at the site of the injured nerve in the L5 or L6 fibers may be sufficient to generate sustained ectopic
                                                                                                                                                                                                                                         discharge necessary to maintain the spinal cord in a sensitized state and to produce the well known changes in expression of central
                                                                                                                                                                                                                                         proteins characteristic of the nerve-injured state. The importance of the contribution of the input from the injured fibers is well
                                                                                                                                                                                                                                         documented as rhizotomy of the L5 and L6 dorsal roots blocks established SNL induced allodynia/ hyperalgesia ( 34 ).
                                                                                                                                                                                                                                           FIG. 3. Immunohistochemical (peroxidase-diaminobenzidine) analysis of PN1 (AD) and PN4 (EH) antibody labeling of L4 DRG
                                                                                                                                                                                                                                           cells from normal and SNL animals, 7 days post-surgery. (A) Naive animals show labeling of all cell types but with an increased
                                                                                                                                                                                                                                           intensity observed in small diameter cells. (B) The pattern of PN1 immunolabeling remains unchanged by SNL injury in animals that
                                                                                                                                                                                                                                           also received saline for 48 hr. SNL animals treated with either PN3 MM (C) or AS (D) ODNs exhibited a similar pattern of PN1
                                                                                                                                                                                                                                           immunolabeling to the saline treated controls. Immunolabeling of L4 DRG cells with PN4 antibody followed a similar pattern to that
                                                                                                                                                                                                                                           observed with the PN1 antibody. (E) PN4 immunolabeling of all cell types, in L4 DRGs taken from nave animals, but with an
                                                                                                                                                                                                                                           increased intensity observed in small diameter cells. (F) Animals receiving a SNL injury and treated with saline showed a similar
                                                                                                                                                                                                                                           PN4 immunolabeling pattern to the nave animals. SNL animals treated with either PN3 MM (G) or AS (H) ODNs exhibited a similar
                                                                                                                                                                                                                                           pattern of PN4 immunolabeling to the saline treated controls. (Bar = 50 m.)
                                                                                                                                                                                                                                               PN3 also has been implicated in the hyperesthesias that result from tissue injury. Thus, modulation of TTX-R INa, by a number of
                                                                                                                                                                                                                                         naturally occurring hyperalgesic substances, e.g., prostaglandin E2 (PGE2), adenosine and serotonin, has been suggested to be a
                                                                                                                                                                                                                                         mechanism that could underlie the subsequent increase in excitability and sensitization of sensory neurons mediated by these agents after a
                                                                                                                                                                                                                                         peripheral nerve or tissue injury ( 35 , 36 , 37 ). Immunohistochemical studies have found that PN3 protein expression appears to increase
                                                                                                                                                                                                                                         in the small-diameter DRG cells of the lumbar enlargement (L4-L6) after chronic inflammation induced by complete Freunds adjuvant
                                                                                                                                                                                                                                         (CFA). In contrast, little evidence has been found for changes in PN3 protein levels after acute inflammation induced by either
                                                                                                                                                                                                                                         carrageenan or formalin (P. Mantyh, personal communication).
                                                                                                                                                                                                                                               Antisense Studies. In support of the immunohistochemical observations, evidence shows that spinal administration (45 g,
                                                                                                                                                                                                                                         intrathecal, twice a day) of an antisense (5-TCC-TCT-GTG-CTT-GGT-TCT-GGC-CT-3), but not mismatch (5-TCC-TTC-GTG-CTG-
                                                                                                                                                                                                                                         TGT-TCG-TGC-CT-3), oligodeoxynucleotide (ODN) to a unique sequence of PN3/SNS produces a selective and reversible block of
                                                                                                                                                                                                                                         channel protein expression ( Fig. 1 ) in rats with spinal nerve (L5/L6) ligation and can prevent the behavioral thermal hyperalgesia and
                                                                                                                                                                                                                                         tactile allodynia ( Fig. 2 ) evoked by this type of injury. Cessation of the antisense ODN treatment at any time after the SNL injury results
                                                                                                                                                                                                                                         in the reoccurrence of tactile allodynia and thermal hyperalgesia within 48 hr, demonstrating the reversibility of the ODN effect. The
                                                                                                                                                                                                                                         equivalent time course for the onset and reversibility of the antisense ODN effect is consistent with the reported 26-hr half life for the rate
                                                                                                                                                                                                                                         of sodium channel turnover and biosynthesis ( 38 ). The lack of effect of the corresponding PN3 mismatch ODN, together with the lack of
                                                                                                                                                                                                                                         effect on baseline (i.e., uninjured) responses to non-noxious or noxious stimuli, also suggests that this was not a nonspecific artifact caused
                                                                                                                                                                                                                                         by repeated injections of an ODN. Intrathecal administration of ODNs previously have been demonstrated to alter the expression of
                                                                                                                                                                                                                                         proteins in the spinal cord, the DRG, and peripheral nerve terminals ( 39 , 40 ). Further, the PN3 antisense, but not mismatch, ODN
                                                                                                                                                                                                                                         affected the expression level of the PN3 channel protein in the DRG, but neither ODN had an effect ( Fig.3 ) on the TTX-S sodium
                                                                                                                                                                                                                                         channels PN1 ( 12 , 13 ) and NaCh6/ SCN8A/PN4 ( 14 , 15 , 16 ). Pretreatment with PN3 antisense also appears to block the development
                                                                                                                                                                                                                                         of both tactile allodynia and thermal hyperalgesia in rats treated with CFA but has no effect on the hyperesthesias evoked acutely by
                                                                                                                                                                                                                                         carrageenan ( Fig. 4 ). In each case, the corresponding mismatch ODN was completely ineffective.
                                                                                                                                                                                                                                           FIG. 4. Prevention of the development of tactile allodynia (A) or thermal hyperalgesia (B) after CFA, but not carrageenan (CAR)-
                                                                                                                                                                                                                                           induced, inflammation by PN3 antisense (AS) ODN. Rats received twice-daily injections (45 g, intrathecal) of either mismatch
                                                                                                                                                                                                                                           (MM) or AS for 2 days and again on the morning of day 3. On the afternoon of day 3, the rats received an injection of CFA (150 l)
                                                                                                                                                                                                                                           or of CAR (200 l of 2%) to the hindpaw. Animals receiving CFA were tested 4 days afterward while receiving twice-daily
the authoritative version for attribution.
                                                                                                                                                                                                                                           administration of AS and MM ODNs throughout this period. Rats receiving CAR were tested on the afternoon of day 3, 3 hr after
                                                                                                                                                                                                                                           CAR injection. Tactile allodynia was indicated by a significant (*, P  0.05) reduction in paw withdrawal threshold in rats receiving
                                                                                                                                                                                                                                           MM, but not AS, on the afternoon of the fourth day after CFA injection. Thermal hyperalgesia was indicated by a significant (*, P 
                                                                                                                                                                                                                                           0.05) reduction in paw withdrawal latency in rats receiving MM, but not AS, on the afternoon of the fourth day after CFA. All
                                                                                                                                                                                                                                           treatment groups receiving CAR demonstrated significant (*, P  0.05) reductions in response thresholds to tactile stimuli (A) or
                                                                                                                                                                                                                                           response latencies to thermal stimuli (B), and these responses were unaffected by either AS or MM ODNs. n = 6 rats per group.
                                                                                                                                                                                                                                         A COMPARISON OF THE POTENTIAL ROLE OF THE TETRODOTOXIN-INSENSITIVE SODIUM CHANNELS, PN3/SNS AND NAN/                                   7643
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                           FIG. 5. Antisense (AS) to NaN/SNS2 administered to rats with SNL injury produced no effect on either tactile allodynia (A) or
                                                                                                                                                                                                                                           thermal hyperalgesia (B). Groups of six rats were used for each of the ODN or saline treatments and were monitored daily for tactile
                                                                                                                                                                                                                                           (von Frey) and thermal nociceptive (radiant heat) responses of the ipsilateral hindpaw. Mismatch (MM) or AS ODN (45 g,
                                                                                                                                                                                                                                           intrathecal) were given twice daily to sham-operated rats and rats with SNL injury. Neither MM nor AS reversed tactile allodynia or
                                                                                                                                                                                                                                           thermal hyperalgesia in the ligated groups, and, likewise, neither treatment altered baseline values in the sham-operated groups. (),
                                                                                                                                                                                                                                           sham-operated, AS; (), SNL, AS; () , sham-operated, MM; () , SNL, MM.
                                                                                                                                                                                                                                               These data suggest that the behavioral consequences of SNL injury and chronic inflammation require de novo PN3 protein synthesis.
                                                                                                                                                                                                                                         This view is borne out by noting that inhibition of PN3 protein expression by the antisense ODN does not affect normal noxious and non-
                                                                                                                                                                                                                                         noxious sensory thresholds on the contralateral side to the nerve injury and fails to affect the allodynia and hyperalgesia seen after
                                                                                                                                                                                                                                         carrageenan, an acute inflammation with a time-course that would not likely be associated with new channel protein synthesis. However,
                                                                                                                                                                                                                                         prevention of protein synthesis by pretreatment with antisense ODN before, or during, chronic injuries, such as SNL or CFA, results in a
                                                                                                                                                                                                                                         complete inhibition of the development of the behavioral consequences of the injury. It should be noted that the lack of effect of PN3
                                                                                                                                                                                                                                         antisense ODN on the behavioral consequences of carrageenaninduced inflammation was studied only at the 3-hr time point after the
                                                                                                                                                                                                                                         initiation of the inflammation. This was most probably insufficient time for PN3 gene transcription and translation to occur to a level that
                                                                                                                                                                                                                                         would have had an impact on the inflammatory process. It is entirely possible that expression of PN3 might be regulated at a later time
                                                                                                                                                                                                                                         point and that PN3 antisense ODN might be active under such circumstances, in a similar manner to the effect observed after CFA
                                                                                                                                                                                                                                         treatment. In apparent contrast to these effects observed in vivo, it has been shown that the TTX-R current recorded from small diameter
                                                                                                                                                                                                                                         cells of the DRG appears to be modulated rapidly in cell culture by inflammatory agents such as PGE2 ( 36 ). This may simply be related
                                                                                                                                                                                                                                         to the much more rapid and sustainable local concentrations of the prostanoid achieved in vitro; that is, the time course remains to be
                                                                                                                                                                                                                                         determined for such inflammatory-mediated increases in TTX-R current in vivo. Nevertheless, the PGE2 effect on the TTX-R INa in vitro
                                                                                                                                                                                                                                         was partially reversed by application of an antisense ODN 21-mer, synthesized against a unique sequence of the PN3/SNS cDNA.
                                                                                                                                                                                                                                         Moreover, the PN3 antisense ODN also reversed the behavioral mechanical hyperalgesia evoked by PGE2 intradermal administration into
                                                                                                                                                                                                                                         the rat hindpaw ( 41 ).
                                                                                                                                                                                                                                           FIG. 6. Immunohistochemical (peroxidase-diaminobenzidine) analysis of NaN/SNS2 antibody labeling of L4 DRG cells from normal
                                                                                                                                                                                                                                           and SNL animals. (A) Naive animals show labeling of small diameter neurons with NaN/SNS2 antibody. No labeling is seen in the
                                                                                                                                                                                                                                           preabsorbed control group (B). In contrast, sham-operated (C) or SNL (D) animals that had received AS ODN to NaN/SNS2 for 48 hr
                                                                                                                                                                                                                                           (a total of four injections) demonstrated a marked loss of NaN/SNS2 immunolabeling in small cells of the L4 DRG ipsilateral to the
                                                                                                                                                                                                                                           side of surgery. Labeling for NaN/SNS2 returned in both sham-operated (E) or SNL rats (F) that were perfused 4 days after the last
                                                                                                                                                                                                                                           AS ODN to NaN/SNS2 injection. (Bar = 50 m.)
                                                                                                                                                                                                                                                       THE PATHOPHYSIOLOGICAL CONTRIBUTION OF NAN/SNS2 IN PERIPHERAL NERVE INJURY?
                                                                                                                                                                                                                                              The novel sodium channel NaN/SNS2, recently cloned from rat DRG ( 22 , 23 ), has an even more restricted distribution within the
                                                                                                                                                                                                                                         small diameter cell population of the DRG. This has led to its consideration as a second potential candidate for the TTX-R INa found in
                                                                                                                                                                                                                                         these cells. However, in contrast to PN3, the low threshold for activation, fast rate of inactivation, and intermediate TTX sensitivity of
                                                                                                                                                                                                                                         NaN/SNS2 resemble most closely the previously described properties of the cardiac channel and, more specifically, the TTX-R3 subtype
the authoritative version for attribution.
                                                                                                                                                                                                                                         of INa ( 21 ). In chronic pain states, a potential role for TTX-R3 INa and, for that matter, NaN/SNS2 has not yet been elucidated, but, like
                                                                                                                                                                                                                                         PN3/SNS, NaN/SNS2 mRNA levels in the DRG are markedly reduced after a peripheral nerve (sciatic) axotomy ( 23 ) and are elevated in
                                                                                                                                                                                                                                         the small diameter DRG cells after persistent inflammation evoked by CFA ( 23 ).
                                                                                                                                                                                                                                              In contrast to the findings with PN3 antisense, spinal administration of antisense (5 GCC TTG TCT TTG GAC TTC TTC 3) and
                                                                                                                                                                                                                                         mismatch (5 GCT CTG TTC TTG AGC TTT CTC 3) ODN to NaN/SNS2 failed to produce any change in sensory thresholds after spinal
                                                                                                                                                                                                                                         nerve (L5/L6) ligation injury ( Fig. 5 ) and did not alter gross behavior, as demonstrated by normal food intake, weight gain, and/or motor
                                                                                                                                                                                                                                         performance, in spite of a significant knock-down of the NaN/SNS2 protein ( Fig. 6 ). This observation suggests, therefore, that the NaN/
                                                                                                                                                                                                                                         SNS2 subtype of TTX-insensitive sodium channel appears unlikely to play a prominent role in the alterations in the sensory phenotype
                                                                                                                                                                                                                                         that have been
                                                                                                                                                                                                                                         A COMPARISON OF THE POTENTIAL ROLE OF THE TETRODOTOXIN-INSENSITIVE SODIUM CHANNELS, PN3/SNS AND NAN/                                                  7644
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         proposed to contribute to the ongoing paresthesias and pain after peripheral nerve injury. NaN/SNS2 channels, like TTX-R3 INa, appear to
                                                                                                                                                                                                                                         be activated, and possibly also inactivated, at much more hyperpolarized potentials than, for example, TTX-R1 INa/PN3. It is therefore
                                                                                                                                                                                                                                         possible that the ineffectiveness of the NaN/SNS2 antisense may reflect the unavailability of NaN/SNS2 channels to contribute to
                                                                                                                                                                                                                                         repetitive firing at the sustained level of membrane depolarization associated with injury to a peripheral nerve; that is, most will be in the
                                                                                                                                                                                                                                         inactivated state ( 21 ).
                                                                                                                                                                                                                                               The physiological or, indeed, pathophysiological role of NaN/ SNS2 therefore remains to be elucidated, particularly in persistent
                                                                                                                                                                                                                                         inflammatory conditions, where an increased expression level of NaN/SNS2 protein in the small diameter cells of the DRG has been
                                                                                                                                                                                                                                         recently reported ( 23 ). The lack of behavioral effects of NaN/SNS2 knock-down serves to validate that ODN administration does not
                                                                                                                                                                                                                                         elicit changes in behaviorally determined thresholds to noxious and non-noxious sensory stimuli in normal or nerve-injured animals non-
                                                                                                                                                                                                                                         specifically but, rather, that knockdown of a functionally important protein (i.e., PN3) is required for these effects to occur.
                                                                                                                                                                                                                                                                                                        CONCLUSIONS
                                                                                                                                                                                                                                               Collectively, therefore, the data suggest that the primary symptoms of neuropathic pain may be significantly attenuated by interfering
                                                                                                                                                                                                                                         with the expression and, consequently, the function of PN3 but not of other sodium channels, which are mainly distributed within the DRG
                                                                                                                                                                                                                                         such as NaN/SNS2. The PN3 antisense-mediated prevention of thermal hyperalgesia induced by CFA, but not by carrageenan, treatment
                                                                                                                                                                                                                                         indicates that the clinical potential for a selective inhibitor of PN3 may extend to pain resulting from chronic tissue, as well as nerve,
                                                                                                                                                                                                                                         injury. Such an effect is also consistent with recent evidence indicating that hyperalgesic substances associated with tissue injury can alter
                                                                                                                                                                                                                                         the function of PN3 ( 35 , 36 ). The lack of an effect of the PN3 antisense on the noxious/non-noxious response thresholds of the
                                                                                                                                                                                                                                         contralateral side to nerve and tissue injury and on the consequences of an acute inflammation (i.e., carrageenan), where the time-course of
                                                                                                                                                                                                                                         the response makes new expression of the PN3 protein unlikely, suggests that this channel does not play a role in normal nociceptive
                                                                                                                                                                                                                                         function. Consequently, these data strongly suggest that selective inhibition of PN3 will not result in changes in normal, nociceptive
                                                                                                                                                                                                                                         function, although this clearly needs to be confirmed. The collective profile of the antisense ODNs against PN3 therefore implicates this
                                                                                                                                                                                                                                         channel in the pathophysiology of pain after nerve and tissue injury. However, it will be important to determine whether PN3 antisense
                                                                                                                                                                                                                                         administration will reverse an established injury in the same way that it has been found to prevent the injury. Moreover, although a role for
                                                                                                                                                                                                                                         PN3 appears to be emerging in peripheral nerve injury, it will be intriguing to see whether this profile might expand to encompass some of
                                                                                                                                                                                                                                         the other types of common neuropathies, e.g., metabolic and chemotherapy. The lack of any overt, particularly central nervous system,
                                                                                                                                                                                                                                         adverse events with the antisense ODN was consistent with the previously described discrete localization of the channel to sensory nerve
                                                                                                                                                                                                                                         fibers with an absence of staining in the central nervous system and cardiac tissue ( 20 ). It also reaffirms the potential that a selective
                                                                                                                                                                                                                                         inhibitor of PN3 may be clinically analgesic, providing not only an improved therapeutic window over existing therapies, but offering
                                                                                                                                                                                                                                         relief from neuropathic pain that is normally resistant to current therapies.
                                                                                                                                                                                                                                               We thank Elda Tzoumaka, Dan Waligora, and Kimberly Wong for their expert technical assistance.
                                                                                                                                                                                                                                         1. Devor, M. ( 1994 ) in Textbook of Pain , eds. Wall, P. D. & Melzack, R. ( Churchill Livingstone , Edinburgh ), pp. 79100 .
                                                                                                                                                                                                                                         2. Woolf, C. J. ( 1994 ) in Textbook of Pain , eds. Wall, P. D. & Melzack, R. ( Churchill Livingstone , Edinburgh ), pp. 101112 .
                                                                                                                                                                                                                                         3. Devor, M. , Govrin-Lippmann, R. & Angelides, K. ( 1993 ) J. Neurosci. 13 , 19761992 .
                                                                                                                                                                                                                                         4. England, J. D. , Happel, L. T. , Kline, D. G. , Gamboni, F. Thouron, C. L. , Liu, Z. P. & Levinson, S. R. ( 1996 ) Neurology 47 , 272276 .
                                                                                                                                                                                                                                         5. Wall, P. D. & Gutnick, M. ( 1974 ) Nature (London) 248 , 740743 .
                                                                                                                                                                                                                                         6. Matzner, O. & Devor, M. J. ( 1994 ) J. Neurophysiol. 72 , 349359 .
                                                                                                                                                                                                                                         7. Devor, M. , Wall, P. D. & Catalan, N. ( 1992 ) Pain 48 , 261268 .
                                                                                                                                                                                                                                         8. Ogata, N. & Tatebayashi, H. ( 1993 ) J. Physiol. 466 , 937 .
                                                                                                                                                                                                                                         9. Roy, M. L. & Narahashi, T. ( 1992 ) J. Neurosci. 12 , 21042111 .
                                                                                                                                                                                                                                         10. Elliott, A. A. & Elliott, J. R. ( 1993 ) J. Physiol. 463 , 3956 .
                                                                                                                                                                                                                                         11. Black, J. A. Dib-Hajj, S. , McNabola, K. , Jeste, S. , Rizzo, M. A. , Kocsis, J. D. & Waxman, S. G. ( 1996 ) Mol. Brain Res. 43 , 117132 .
                                                                                                                                                                                                                                         12. Toledo-Aral, J. J. , Moss, B. L. , He, Z. J. , Koszowski, A. G. , Whisenand, T. , Levinson, S. R. , Wolf, J. J. , Silos-Santiago, I. , Halegoua, S. &
                                                                                                                                                                                                                                              Mandel, G. ( 1997 ) Proc. Natl. Acad. Sci. USA 94 , 15271532 .
                                                                                                                                                                                                                                         13. Sangameswaran, L. , Fish, L. M. , Koch, B. D. , Rabert, D. K. , Delgado, S. G. , Ilnicka, M. , Jakeman, L. B. , Novakovic, S. , Wong, K. , Sze, P. , et
                                                                                                                                                                                                                                              al. ( 1997 ) J. Biol. Chem. 272 , 1480514809 .
                                                                                                                                                                                                                                         14. Schaller, K. L. , Krzemien, D. M. , Yarowsky, P. J. , Krueger, B. K. & Caldwell, J. H. ( 1995 ) J. Neurosci. 15 , 32313242 .
                                                                                                                                                                                                                                         15. Burgess, D. L. , Kohrman, D. C. , Galt, J. , Plummer, N. W. , Jones, J. M. , Spear, B. & Meisler, M. H. ( 1995 ) Nat. Genet . 10 , 461465 .
                                                                                                                                                                                                                                         16. Dietrich, P. S. , McGivern, J. G. , Delgado, S. G. , Koch, B. D. , Eglen, R. M. , Hunter J. C. & Sangameswaran L. ( 1998 ) J. Neurochem. 70 , 2262
                                                                                                                                                                                                                                              2272 .
                                                                                                                                                                                                                                         17. Arbuckle, J. B. & Docherty, R. J. ( 1995 ) Neurosci. Lett. 185 , 7073 .
                                                                                                                                                                                                                                         18. Sangameswaran, L. , Delgado, S. G. , Fish, L. M. , Koch, B. D. , Jakeman, L. B. , Stewart, G. R. , Sze, P. , Hunter, J. C. , Eglen, R. M. & Herman, R.
                                                                                                                                                                                                                                              C. ( 1996 ) J. Biol. Chem. 271 , 59535956 .
                                                                                                                                                                                                                                         19. Akopian, A. N. , Sivilotti, L. & Wood, J. N. ( 1996 ) Nature (London) 379 , 257262 .
                                                                                                                                                                                                                                         20. Novakovic, S. D. , Tzoumaka, E. , McGivern, J. G. , Haraguchi, M. , Sangameswaran, L. , Gogas, K. R. , Eglen, R. M. & Hunter, J. C. ( 1998 ) J.
                                                                                                                                                                                                                                              Neurosci. 18 , 21742187 .
                                                                                                                                                                                                                                         21. Rush, A. M. , Brau, M. E. , Elliott, A. A. & Elliott, J. R. ( 1998 ) J. Physiol. 551 , 771789 .
                                                                                                                                                                                                                                         22. Dib-Hajj, S. D. , Tyrrell, L. , Black, J. A. & Waxman, S. G. ( 1998 ) Proc. Natl. Acad. Sci. USA 95 , 89638968 .
                                                                                                                                                                                                                                         23. Tate, S. , Benn, S. , Hick, C. , Trezise, D. , John, V. , Mannion, R. J. , Costigan, M. , Plumpton, C. , Grose, D. , Gladwell, Z. , et al. ( 1998 ) Nat.
                                                                                                                                                                                                                                              Neurosci. 1 , 653655 .
                                                                                                                                                                                                                                         24. Elliott, J. R. ( 1997 ) Brain Res 754 , 221226 .
                                                                                                                                                                                                                                         25. Rizzo, M. A. , Kocsis, J. D. & Waxman, S. G. ( 1995 ) Neurobiol. Disease 2 , 8796 .
                                                                                                                                                                                                                                         26. Cummins, T. R. & Waxman, S. G. ( 1997 ) J. Neurosci. 17 , 35033514 .
                                                                                                                                                                                                                                         27. Dib-Hajj, S. , Black, J. A. , Felts, P. & Waxman, S. G. ( 1996 ) Proc. Natl. Acad. Sci. USA 93 , 1495014954 .
                                                                                                                                                                                                                                         28. Coggeshall, R. E. , Dougherty, P. M. , Pover, C. M. & Carlton, S. M. ( 1993 ) Pain 52 , 233242 .
                                                                                                                                                                                                                                         29. Kim, S. H. & Chung, J. M. ( 1992 ) Pain 50 , 355363 .
                                                                                                                                                                                                                                         30. Yoon, Y. W. , Na, H. S. & Chung, J. M. ( 1996 ) Pain 64 , 2736 .
                                                                                                                                                                                                                                         31. Black, J. A. , Langworthy, K. , Hinson, A. W. , Dib-Hajj, S. D. & Waxman, S. G. ( 1997 ) NeuroReport 8 , 23312335 .
                                                                                                                                                                                                                                         32. DArcangelo, G. , Paradiso, K. , Shepherd, D. , Brehm, P. , Halegoua, S. & Mandel, G. ( 1993 ) J. Cell Biol. 122 , 915921 .
the authoritative version for attribution.
                                                                                                                                                                                                                                         33. Zur, K. B. , Oh, Y. , Waxman, S. G. & Black, J. A. ( 1995 ) Mol. Brain Res. 30 , 97103 .
                                                                                                                                                                                                                                         34. Sheen, K. & Chung, J. M. ( 1993 ) Brain Res. 610 , 6268 .
                                                                                                                                                                                                                                         35. England, S. , Bevan, S. & Docherty, R. J. ( 1996 ) J. Physiol. 495 , 429440 .
                                                                                                                                                                                                                                         36. Gold, M. S. , Reichling, D. B. , Shuster, M. J. & Levine, J. D. ( 1996 ) Proc. Natl. Acad. Sci. USA 93 , 11081112 .
                                                                                                                                                                                                                                         37. Cardenas, C. G. , Del Mar, L. P. & Scroggs, R. S. ( 1997 ) J. Neurosci. 17 , 71817189 .
                                                                                                                                                                                                                                         38. Waechter, C. J. , Schmidt, J. W. & Catterall, W. A. ( 1983 ) J. Biol. Chem. 258 , 51175123 .
                                                                                                                                                                                                                                         39. Bilsky, E. J. , Wang, T. , Lai, J. & Porreca, F. ( 1996 ) Neurosci. Lett. 220 , 155158 .
                                                                                                                                                                                                                                         40. Khasar, S. G. , Gold, M. S. , Dastmalchi, S. & Levine, J. D. ( 1996 ) Neurosci. Lett. 218 , 1720 .
                                                                                                                                                                                                                                         41. Khasar, S. G. , Gold, M. S. & Levine, J. D. ( 1998 ) Neurosci. Lett. 256 , 1720 .
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                              MICHAEL S. GOLD *
                                                                                                                                                                                                                                              Department of Oral and Craniofacial Biological Sciences, University of Maryland Dental School, 666 West Baltimore Street,
                                                                                                                                                                                                                                         Baltimore, MD 21201
                                                                                                                                                                                                                                              ABSTRACT Several mechanisms have been identified that may underlie inflammation-induced sensitization of high-
                                                                                                                                                                                                                                         threshold primary afferent neurons, including the modulation of voltage- and Ca2+-dependent ion channels and ion channels
                                                                                                                                                                                                                                         responsible for the production of generator potentials. One such mechanism that has recently received a lot of attention is the
                                                                                                                                                                                                                                         modulation of a tetrodotoxin (TTX)-resistant voltage-gated Na+ current. Evidence supporting a role for TTX-resistant Na+
                                                                                                                                                                                                                                         currents in the sensitization of primary afferent neurons and inflammatory hyperalgesia is reviewed. Such evidence is derived
                                                                                                                                                                                                                                         from studies on the distribution of TTX-resistant Na+ currents among primary afferent neurons and other tissues of the body that
                                                                                                                                                                                                                                         suggest that these currents are expressed only in a subpopulation of primary afferent neurons that are likely to be involved in
                                                                                                                                                                                                                                         nociception. Data from studies on the biophysical properties of these currents suggest that they are ideally suited to mediate the
                                                                                                                                                                                                                                         repetitive discharge associated with prolonged membrane depolarizations. Data from studies on the effects of inflammatory
                                                                                                                                                                                                                                         mediators and antinociceptive agents on TTX-resistant Na+ currents suggest that modulation of these currents is an underlying
                                                                                                                                                                                                                                         mechanism of primary afferent neuron sensitization. In addition, the second-messenger pathways underlying inflammatory
                                                                                                                                                                                                                                         mediator-induced modulation of these currents appear to underlie inflammatory mediator-induced hyperalgesia. Finally, recent
                                                                                                                                                                                                                                         antisense studies have also yielded data supporting a role for TTX-resistant Na+ currents in inflammatory hyperalgesia. Although
                                                                                                                                                                                                                                         data from these studies are compelling, data presented at the Neurobiology of Pain colloquium raised a number of interesting
                                                                                                                                                                                                                                         questions regarding the role of TTX-resistant Na+ currents in inflammatory hyperalgesia; implications of three of these questions
                                                                                                                                                                                                                                         are discussed.
                                                                                                                                                                                                                                              Hyperalgesia that develops in the presence of tissue injury or inflammation reflects, at least in part, an increase in the excitability of
                                                                                                                                                                                                                                         high-threshold primary afferent neurons innervating the site of injury. The increase in afferent excitability, or sensitization, develops
                                                                                                                                                                                                                                         within minutes of an inflammatory stimulus and involves a leftward shift in neuronal stimulus response function and/or an increase in
                                                                                                                                                                                                                                         spontaneous activity. The relatively rapid development of sensitization in response to inflammatory stimuli is likely to reflect the
                                                                                                                                                                                                                                         modulation of proteins within or around the afferent terminal. In contrast, a change in the expression of protein(s) appears to be involved
                                                                                                                                                                                                                                         in afferent sensitization observed in the presence of ongoing inflammation or nerve injury (see accompanying papers). At least three
                                                                                                                                                                                                                                         underlying mechanisms have been identified that may contribute to the initial phase of inflammation-induced afferent sensitization: (i) a
                                                                                                                                                                                                                                         change in the compliance of the tissue surrounding the afferent terminal ( 1 ); (ii) a change in efficacy of a transducer(s) within the afferent
                                                                                                                                                                                                                                         terminal ( 2  4 ); and (iii) a change in a voltage- or Ca2+-dependent current within the afferent terminal ( 5  8 ). Because inflammatory
                                                                                                                                                                                                                                         mediators may sensitize sensory neurons in vitro to stimuli that presumably bypass the afferent transduction apparati [i.e., high
                                                                                                                                                                                                                                         extracellular potassium or current injection through a recording electrode ( 9 , 10 )], with no consistent changes either in resting membrane
                                                                                                                                                                                                                                         conductance ( 7 ) or potential ( 10 ), modulation of a voltage- or Ca2+-dependent current must contribute to the sensitization of primary
                                                                                                                                                                                                                                         afferent neurons. We have focused this review on the contribution of a particular class of voltage-gated Na+ currents (VGSCs), namely
                                                                                                                                                                                                                                         tetrodotoxin (TTX)-resistant voltage-gated Na+ currents, to changes in afferent excitability.
                                                                                                                                                                                                                                                                                            WHY FOCUS ON NA+ CHANNELS?
                                                                                                                                                                                                                                              Although a number of distinct voltage- and Ca2+-dependent currents have been identified that may underlie inflammationinduced
                                                                                                                                                                                                                                         changes in afferent excitability (for review, see ref. 11 ), we have focused on the role of VGSCs for several reasons. First, VGSC
                                                                                                                                                                                                                                         activation is critical for the generation and propagation of neuronal action potentials. Second, there is a growing body of evidence
                                                                                                                                                                                                                                         indicating that modulation of these currents is an endogenous mechanism used to control neuronal excitability ( 8 , 12  15 ). Third,
                                                                                                                                                                                                                                         evidence from injury in experimental animals ( 16  19 ) and humans ( 20 , 21 ) suggests that therapeutic interventions with compounds
                                                                                                                                                                                                                                         known to block Na+ channels may be effective for the treatment of hyperalgesia and pain.
                                                                                                                                                                                                                                                                                          WHY TTX-RESISTANT CHANNELS?
                                                                                                                                                                                                                                              Distribution. Evidence for the selective distribution of unique VGSC(s) among sensory neurons has been obtained in vivo and in
                                                                                                                                                                                                                                         vitro. Intracellular recording from the cell bodies of sensory neurons in vivo indicated that the somal action potential of high-threshold
                                                                                                                                                                                                                                         receptors is resistant to tetrodotoxin (TTX) at concentrations as high as 200 M applied to the surface of the ganglion ( 22 ). Similar
                                                                                                                                                                                                                                         results were obtained with intracellular recording from intact ganglia in vitro, where it was observed that TTX-resistant action potentials
                                                                                                                                                                                                                                         were present in neurons with slow-conducting axons (i.e., neurons likely to be associated with high-threshold receptors) ( 23 ).
                                                                                                                                                                                                                                         Electrophysiological studies on dissociated sensory neurons have demonstrated that while TTX-sensitive Na+ currents are distributed
                                                                                                                                                                                                                                         throughout the population of spinal sensory neurons, TTX-resistant Na+ currents are primarily restricted to a subpopulation of sensory
                                                                                                                                                                                                                                         neurons likely to be involved in nociception ( 8 , 24  28 ). Specifically, TTX-resistant Na+ currents are present primarily in neurons that
                                                                                                                                                                                                                                         have a small cell-body diameter (these are the neurons that tend to give rise to
the authoritative version for attribution.
                                                                                                                                                                                                                                         small-diameter slow-conducting axons) and that are responsive to the algogenic compound capsaicin. Of note, a TTX-insensitive Na+
                                                                                                                                                                                                                                         current (i.e., a current blocked by TTX at concentrations between 500 nM and 1 M) had been described in other tissues ( 29  31 ).
                                                                                                                                                                                                                                         However, the biophysical properties of TTX-insensitive currents appears to differ from those of TTX-resistant currents ( 29 , 31 ).
                                                                                                                                                                                                                                               Identification of a gene encoding a TTX-resistant Na+ channel confirmed the electrophysiological data indicating the existence of
                                                                                                                                                                                                                                         unique Na+ currents in a subpopulation of primary afferent neurons. The first TTX-resistant Na+ channel cloned, referred to as SNS ( 32 ),
                                                                                                                                                                                                                                         PN3 ( 33 ), and subsequently ScN10 ( 34 ), is only present in primary afferent neurons, in particular, a subpopulation of primary afferent
                                                                                                                                                                                                                                         neurons with small-diameter cell bodies. Heterologous expression of SNS/ PN3 indicated that this clone encodes a voltage-gated Na+
                                                                                                                                                                                                                                         channel with biophysical properties similar to those of the TTX-resistant channels present in sensory neurons ( 32 , 33 ). A second TTX-
                                                                                                                                                                                                                                         resistant Na+ channel, referred to as NaN ( 35 ) or SNS2 ( 36 ), recently cloned from sensory neurons is also present in a subpopulation of
                                                                                                                                                                                                                                         sensory neurons with a small-diameter cell body.
                                                                                                                                                                                                                                               Biophysical Properties. At least three distinct TTX-resistant Na+ currents have been electrophysiologically isolated in rat primary
                                                                                                                                                                                                                                         afferent neurons ( 37 , 38 ). The first TTX-resistant currents to be described ( 25  28 , 39 , 40 ) had several unique features. First, these
                                                                                                                                                                                                                                         TTX-resistant currents have high thresholds for activation relative to TTX-sensitive currents ( 26 , 27 , 38 ). If the channels underlying
                                                                                                                                                                                                                                         these currents are present in the peripheral terminals of primary afferent neurons and if spike initiation involves activation of these
                                                                                                                                                                                                                                         channels, then the activation properties of these channels may explain why afferent neurons responsive to noxious stimuli have high
                                                                                                                                                                                                                                         thresholds for activation. Second, TTX-resistant currents have high thresholds for steady-state inactivation relative to TTX-sensitive
                                                                                                                                                                                                                                         currents ( 26 , 27 , 38 ). As a result, the majority of TTX-resistant channels are available for activation at membrane potentials as high as
                                                                                                                                                                                                                                         40 mV ( 27 ). Consequently, it has been suggested that activation of these channels contributes to ongoing activity observed in the
                                                                                                                                                                                                                                         presence of a sustained depolarization of primary afferent neurons ( 38 ). Third, TTX-resistant currents recover from inactivation rapidly
                                                                                                                                                                                                                                         relative to TTX-sensitive currents (refs. 26 and 38 , but see ref. 27 ). Rapid recovery from inactivation is another factor that would enable
                                                                                                                                                                                                                                         TTX-resistant currents to underlie sustained spiking in response to prolonged depolarizations ( 26 , 41 ). Fourth, the inactivation rates for
                                                                                                                                                                                                                                         TTX-resistant Na+ currents are considerably slower than those of TTX-sensitive currents ( 38 ). This is particularly true at membrane
                                                                                                                                                                                                                                         potentials close to the activation potential for TTX-resistant currents. Consequently, membrane depolarization may be facilitated after the
                                                                                                                                                                                                                                         activation of a sustained inward current carried by TTX-resistant Na+ channels. The slow inactivation rate of TTX-resistant Na+ currents
                                                                                                                                                                                                                                         also contributes to the broad action potential typically observed in high-threshold primary afferent neurons ( 41 ). Thus, the biophysical
                                                                                                                                                                                                                                         properties of the first and most widely studied TTX-resistant Na+ currents are such that these currents may play a critical role in the
                                                                                                                                                                                                                                         determination of the excitability of the afferent neurons in which they are present.
                                                                                                                                                                                                                                               The biophysical properties of recently described TTX-resistant Na+ currents ( 37 , 38 ) more closely resemble TTX-sensitive Na+
                                                                                                                                                                                                                                         currents than the TTX-resistant Na+ currents described above. For example, a second and third TTX-resistant Na+ current characterized by
                                                                                                                                                                                                                                         Rush et al. ( 38 ) activated and inactivated at relatively hyperpolarized membrane potentials; the third current inactivated at membrane
                                                                                                                                                                                                                                         potentials even more negative than those observed for TTX-sensitive Na+ currents. Of note, the inactivation rate of all three TTX-resistant
                                                                                                                                                                                                                                         Na+ currents described by Rush et al. was relatively slow compared with that of TTX-sensitive Na+ currents. In contrast, a second TTX-
                                                                                                                                                                                                                                         resistant Na+ current described by Sholz et al. ( 37 ) displayed both rapid kinetics for activation and inactivation and a voltage dependence
                                                                                                                                                                                                                                         of activation and inactivation that occurred over relatively hyperpolarized membrane potentials. The role these additional TTX-resistant Na
                                                                                                                                                                                                                                         + currents play in regulating the excitability of high-threshold primary afferent neurons has yet to be determined.
                                                                                                                                                                                                                                               Effects of Inflammatory Mediators. The distribution and biophysical properties of the classically described TTX-resistant Na+
                                                                                                                                                                                                                                         currents suggests that these currents are involved in the control of the excitability of primary afferent neurons. Furthermore, several
                                                                                                                                                                                                                                         inflammatory mediators released in response to injury are capable of directly sensitizing subpopulations of primary afferent neurons ( 9 ,
                                                                                                                                                                                                                                         10 , 14 , 42 , 43 ). Therefore, we hypothesized that an inflammatory mediator-induced modulation of TTX-resistant Na+ currents is a
                                                                                                                                                                                                                                         mechanism underlying the sensitization of primary afferent neurons. In support of this hypothesis, we observed that directly acting
                                                                                                                                                                                                                                         hyperalgesic inflammatory mediators such as prostaglandin E2 (PGE2), serotonin, and adenosine decrease the activation threshold,
                                                                                                                                                                                                                                         increase the rates of activation and inactivation, and increase the magnitude of TTX-resistant Na+ current ( 8 ). These changes could
                                                                                                                                                                                                                                         contribute to both the decrease in threshold and increase in the number of action potentials evoked from a sensitized neuron. Further
                                                                                                                                                                                                                                         support of our hypothesis is provided by the observation that the time course of inflammatory mediator-induced modulation of TTX-
                                                                                                                                                                                                                                         resistant Na+ currents [developing within seconds and attaining a maximum within minutes ( 8 )] mirrors the time course of the
                                                                                                                                                                                                                                         development of hyperalgesia in response to a peripheral injection of directly acting inflammatory mediators ( 44 ). Similar observations
                                                                                                                                                                                                                                         have subsequently been reported by other investigators ( 14 , 45 ).
                                                                                                                                                                                                                                               Effects of Antinociceptive Agents. Levine and Taiwo ( 46 ) previously observed that the peripheral administration of -opioid
                                                                                                                                                                                                                                         receptor agonists blocked inflammatory mediatorinduced hyperalgesia. Therefore, we hypothesized that if inflammatory mediator-induced
                                                                                                                                                                                                                                         modulation of TTX-resistant Na+ currents is an underlying mechanism of inflammatory hyperalgesia, then -opioid receptor agonists
                                                                                                                                                                                                                                         should block inflammatory mediator-induced modulation of current. Consistent with this hypothesis, we observed that a -opioid receptor
                                                                                                                                                                                                                                         agonist blocked PGE2-induced modulation of the current ( 47 ).
                                                                                                                                                                                                                                               Second-Messenger Pathways. In a final series of experiments designed to test the hypothesis that inflammatory mediator-induced
                                                                                                                                                                                                                                         modulation of TTX-resistant Na+ currents is an underlying mechanism of inflammatory hyperalgesia, we attempted to determine the role
                                                                                                                                                                                                                                         of protein kinase A (PKA) in PGE2-induced modulation of the current. Previous studies performed in vivo suggested that direct acting
                                                                                                                                                                                                                                         inflammatory mediators, including PGE2, serotonin, and adenosine, produced hyperalgesia ( 48  52 ) and afferent sensitization ( 53 ) via
                                                                                                                                                                                                                                         the activation of a cAMP/PKA second-messenger cascade. The effects of these mediators were mimicked by compounds that increase the
                                                                                                                                                                                                                                         intracellular concentration of cAMP, prolonged by agents that blocked the breakdown of cAMP, and blocked by agents that inhibit
                                                                                                                                                                                                                                         adenylate cyclase and/or PKA.
                                                                                                                                                                                                                                               Consistent with our hypothesis, England et al. ( 14 ) reported that PGE2-induced modulation of the TTX-resistant Na+ currents
                                                                                                                                                                                                                                         involved activation of a cAMP/PKA second-messenger pathway. However, these experiments were performed on primary afferent
                                                                                                                                                                                                                                         neurons from neonatal rats, and there are several lines of evidence suggesting primary afferent neurons from neonates may be qualitatively
                                                                                                                                                                                                                                         and quantitatively different than neurons from adults ( 39 , 54  56 ). Furthermore, after failing to detect an effect with a membrane-
                                                                                                                                                                                                                                         permeable analog of cAMP on TTX-resistant Na+ currents in primary afferent neurons from adult rats, Cardenas et al. ( 45 ) were
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         forced to conclude that modulation of the current must involve activation of another second-messenger pathway. We have recently
                                                                                                                                                                                                                                         reported ( 57 ) that although an increase in the intracellular concentration of cAMP may result in the modulation of TTX-resistant Na+
                                                                                                                                                                                                                                         current, the doseresponse relationship for such manipulations is bell-shaped. This observation may explain, at least in part, differences
                                                                                                                                                                                                                                         between the observations of England et al. and those of Cardenas et al. It should be noted that a recent study involving heterologous
                                                                                                                                                                                                                                         expression and site directed mutagenesis of the cloned TTX-resistant Na+ channel, SNS/PN3, indicates that the channel is phosphorylated
                                                                                                                                                                                                                                         after activation of PKA ( 58 ). Furthermore, PKA-induced phosphorylation of the channel results in changes in gating properties similar to
                                                                                                                                                                                                                                         those induced by inflammatory mediators ( 58 ), suggesting that inflammatory mediator-induced modulation of TTX-resistant currents
                                                                                                                                                                                                                                         reflects a direct phosphorylation of the underlying channel(s). Importantly, and more to the point, our recent results ( 57 ) strongly support
                                                                                                                                                                                                                                         the suggestion that PGE2-induced modulation of TTX-resistant Na+ currents in primary afferent neurons involves PKA activation.
                                                                                                                                                                                                                                               The Function of TTX-Resistant Na+ Currents in Peripheral Terminals. Results obtained through the study of primary afferent
                                                                                                                                                                                                                                         neuron cell bodies in vitro has provided compelling evidence in support of the hypothesis that modulation of TTX-resistant Na+ currents is
                                                                                                                                                                                                                                         an underlying mechanism of inflammatory hyperalgesia. However, it is critical to determine whether these currents contribute to
                                                                                                                                                                                                                                         inflammatory hyperalgesia in vivo. TTX-resistant Na+ currents are clearly present in the DRG cell body in vivo ( 22 ). Furthermore, results
                                                                                                                                                                                                                                         from at least two studies suggest that these currents are present in the central terminals of primary afferent neurons ( 59 , 60 ). There also is
                                                                                                                                                                                                                                         evidence that TTX-resistant Na+ currents are present in peripheral axons ( 61 ), but given that axonal conduction is blocked with TTX (
                                                                                                                                                                                                                                         22 , 23 ), the function of TTX-resistant Na+ current in the axon has yet to be determined. Importantly, Brock et al. ( 62 ) have recently
                                                                                                                                                                                                                                         obtained evidence suggesting that TTX-resistant Na+ channels play a role in action potential generation in the peripheral terminals of
                                                                                                                                                                                                                                         corneal afferent neurons. Consistent with observations made while recording from cell bodies ( 22 , 23 ), recording from the peripheral
                                                                                                                                                                                                                                         terminals revealed that electrical stimulation of the nerve trunk evoked action potentials that were blocked by TTX whereas spontaneous
                                                                                                                                                                                                                                         or naturally evoked (with pressure or capsaicin) action potential persisted in the presence of TTX ( 62 ).
                                                                                                                                                                                                                                               Although these recent results support the suggestion that TTX-resistant currents are present and functional in the peripheral terminals
                                                                                                                                                                                                                                         of primary afferent neurons, they do not address the question of whether modulation of these currents contributes to inflammatory
                                                                                                                                                                                                                                         hyperalgesia. Given the lack of specific pharmacological agents with which to manipulate TTX-resistant Na+ currents, it is not possible to
                                                                                                                                                                                                                                         address this issue with traditional pharmacological approaches. However, through the use of antisense oligodeoxynucleotides (ODNs) to
                                                                                                                                                                                                                                         selectively knock down expression of protein encoded by targeted mRNA ( 63 ), it has become possible to study the function of specific
                                                                                                                                                                                                                                         proteins. Furthermore, we ( 64 ), and others ( 65 ) had previously demonstrated that the intrathecal administration of ODNs could be used
                                                                                                                                                                                                                                         to knock down expression of proteins present in the peripheral terminals of primary afferent neurons. Therefore, we generated antisense
                                                                                                                                                                                                                                         ODNs to a unique region of the cloned TTX-resistant Na+ channel, PN3/SNS, and assessed the effects of intrathecal ODN administration
                                                                                                                                                                                                                                         on PGE2-induced hyperalgesia ( 66 ). Our results indicated that antisense, but not control, ODN sequences produced a small but significant
                                                                                                                                                                                                                                         increase in baseline threshold to mechanical nociceptive stimuli, suggesting that activity in a TTX-resistant current contributes to the
                                                                                                                                                                                                                                         determination of mechanical threshold. More importantly, antisense, but not control ODN sequences, resulted in a significant reduction in
                                                                                                                                                                                                                                         PGE2-induced hyperalgesia. This observation is consistent with the electrophysiological data indicating that functional TTX-resistant Na+
                                                                                                                                                                                                                                         channels are present in the peripheral terminals of primary afferent neurons. Furthermore, it supports the hypothesis that modulation of a
                                                                                                                                                                                                                                         TTX-resistant Na+ current is an underlying mechanism of inflammatory hyperalgesia.
                                                                                                                                                                                                                                               Questions Concerning the Role of TTX-Resistant Na+ Currents in Inflammatory Hyperalgesia. There are a number of questions
                                                                                                                                                                                                                                         that remain to be answered concerning the role of TTX-resistant Na+ currents in inflammatory pain. At least three of these deserve
                                                                                                                                                                                                                                         comment in light of data presented at the Neurobiology of Pain colloquium. First, what is the function of the second TTX-resistant Na+
                                                                                                                                                                                                                                         current (NAN/ SNS2) cloned from spinal sensory neurons? Expression of the channel appears restricted to primary afferent neurons with
                                                                                                                                                                                                                                         the smallest cell-body diameter in a subpopulation of neurons expressing PN3/SNS ( 36 ). The biophysical properties of the NAN/SNS2
                                                                                                                                                                                                                                         expressed in HEK292 cells appear to more closely resemble TTX-sensitive Na+ currents (i.e., with faster activation and inactivation
                                                                                                                                                                                                                                         kinetics), although these properties may not be reflective of the properties of the channel expressed in native tissue. Because there is no
                                                                                                                                                                                                                                         homology between PN3/SNS and NaN/SNS2 in the region we targeted with our antisense ODN, it is unlikely that our results with
                                                                                                                                                                                                                                         antisense ODNs reflect knock-down of both channels. Consequently, the residual PGE2-induced hyperalgesia we observed after PN3/SNS
                                                                                                                                                                                                                                         antisense ODN administration may reflect an effect of PGE2 on NaN/SNS2. Porreca et al. ( 67 ) have recently obtained data suggesting
                                                                                                                                                                                                                                         that NaN/SNS2 is not involved in either the establishment of nociceptive thresholds in control animals or in the maintenance of
                                                                                                                                                                                                                                         hyperalgesia and allodynia in a neuropathy model. However, these investigators did not investigate the role of this channel in
                                                                                                                                                                                                                                         inflammatory hyperalgesia. Thus, a role of NaN/SNS2 has yet to be determined.
                                                                                                                                                                                                                                               The contribution of TTX-resistant Na+ currents to nociceptive thresholds in uninjured tissue is a second question concerning the role
                                                                                                                                                                                                                                         of TTX-resistant Na+ currents in inflammatory pain. Results from our antisense study suggest that these currents do contribute, to a limited
                                                                                                                                                                                                                                         extent, to the determination of mechanical nociceptive threshold ( 66 ). That the contribution of these currents to the determination of
                                                                                                                                                                                                                                         nociceptive threshold is small is supported by observations made by Porreca et al. ( 67 ). These investigators were able to clearly
                                                                                                                                                                                                                                         demonstrate a decrease in PN3/SNS protein in the cell bodies of primary afferent neurons and an attenuation of both inflammatory and
                                                                                                                                                                                                                                         neuropathic hyperalgesia, by using an antisense strategy similar to the one we used. However, antisense ODNs had no effect on baseline
                                                                                                                                                                                                                                         mechanical or thermal nociceptive thresholds. The small effect of antisense ODN treatment on baseline nociceptive threshold is striking in
                                                                                                                                                                                                                                         light of the observation that in the nociceptor cell body, TTX-resistant Na+ current is the Na+ current primarily responsible for action
                                                                                                                                                                                                                                         potential generation ( 14 ). The apparent difference between the role of TTX-resistant Na+ current in the cell body and in the peripheral
                                                                                                                                                                                                                                         terminal suggests that either the current contributes little to the determination of baseline nociceptive threshold or activity in the population
                                                                                                                                                                                                                                         of TTX-resistant Na+ current-containing afferent neurons contributes little to baseline nociceptive threshold. Although a single-unit
                                                                                                                                                                                                                                         electrophysiological study is necessary to distinguish between these possibilities, observations obtained with the neurotoxin capsaicin
                                                                                                                                                                                                                                         would suggest the latter. That is, rats treated neonatally with capsaicin to eliminate a vast majority of c-fiber afferent neurons have
                                                                                                                                                                                                                                         baseline nociceptive thresholds that are only slightly elevated. However, PGE2-induced hyperalgesia is completely eliminated in these
                                                                                                                                                                                                                                         animals ( 68 ).
                                                                                                                                                                                                                                               Third, there is the question as to why the administration of antisense ODNs directed against SNS/PN3 had no effect on carrageenan-
                                                                                                                                                                                                                                         induced hyperalgesia as observed by Porreca et al.
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         ( 67 ). The peripheral administration of carrageenan is used as a model of acute inflammation associated with hyperalgesia that develops
                                                                                                                                                                                                                                         within tens of minutes. In their carefully controlled study, Porreca et al. were able to demonstrate profound effects with antisense ODN
                                                                                                                                                                                                                                         treatment on hyperalgesia resulting from the peripheral administration of Freunds adjuvant. Like carrageenan, the peripheral
                                                                                                                                                                                                                                         administration of Freunds adjuvant is used as a model of inflammation, except the hyperalgesia associated with this model develops more
                                                                                                                                                                                                                                         slowly, over many hours. Our results suggest that modulation of SNS/PN3 or a highly homologous species, contributes to inflammatory
                                                                                                                                                                                                                                         mediator-induced hyperalgesia ( 66 ). Furthermore, carrageenan-induced hyperalgesia appears to involve the production and release of
                                                                                                                                                                                                                                         hyperalgesic inflammatory mediators (i.e., see ref. 69 ). Consequently, we would predict that carrageenan-induced hyperalgesia should be
                                                                                                                                                                                                                                         attenuated after SNS/PN3 antisense ODN administration. Identification of the reason(s) for the discrepancy between our predicted results
                                                                                                                                                                                                                                         and the observations of Porreca et al. appears to require further experimentation.
                                                                                                                                                                                                                                                                                                       CONCLUSIONS
                                                                                                                                                                                                                                               Pain is clearly a complex process involving considerably more than the modulation of a single class of ion channels resulting in
                                                                                                                                                                                                                                         changes in the excitability of a subpopulation of neurons. That the most effective analgesics available tend to have a wide spectrum of
                                                                                                                                                                                                                                         action at a number of sites throughout the nervous system is largely reflective of this fact. Nevertheless, the study of a single class of ion
                                                                                                                                                                                                                                         channels, TTX-resistant Na+ channels, has increased our understanding of the neurobiology of pain. Furthermore, because of the restricted
                                                                                                                                                                                                                                         distribution of TTX-resistant Na+ currents and the observation that a decrease in the expression of these currents has little impact on low-
                                                                                                                                                                                                                                         threshold mechanical transduction, targeting these currents may lead to the development of a therapeutic modality for the treatment of
                                                                                                                                                                                                                                         hyperalgesia with fewer side effects than currently available modalities.
                                                                                                                                                                                                                                               I would like to thank Normal Capra for helpful comments regarding the manuscript. Some of the work described in this article was
                                                                                                                                                                                                                                         supported by grants from the National Institute of Health (Grant 1RO1NS3692901A1).
                                                                                                                                                                                                                                         1. Cooper, B. ( 1993 ) J. Neurophysiol. 70 , 512521 .
                                                                                                                                                                                                                                         2. Cesare, P. & McNaughton, P. ( 1996 ) Proc. Natl. Acad. Sci. USA 93 , 1543515439 .
                                                                                                                                                                                                                                         3. Reichling, D. B. & Levine, J. D. ( 1997 ) Proc. Natl. Acad. Sci. USA 94 , 70067011 .
                                                                                                                                                                                                                                         4. Lopshire, J. C. & Nicol, G. D. ( 1997 ) J. Neurophysiol. 78 , 31543164 .
                                                                                                                                                                                                                                         5. Nicol, G. D. , Vasko, M. R. & Evans, A. R. ( 1997 ) J. Neurophysiol. 77 , 167176 .
                                                                                                                                                                                                                                         6. Weinreich, D. ( 1995 ) Pulm. Pharmacol. 8 , 173179 .
                                                                                                                                                                                                                                         7. Gold, M. S. , Shuster, M. J. & Levine, J. D. ( 1996 ) Neurosci. Lett. 205 , 161164 .
                                                                                                                                                                                                                                         8. Gold, M. S. , Reichling, D. B. , Shuster, M. J. & Levine, J. D. ( 1996 ) Proc. Natl. Acad. Sci. USA 93 , 11081112 .
                                                                                                                                                                                                                                         9. Baccaglini, P. I. & Hogan, P. G. ( 1983 ) Proc. Natl. Acad. Sci. USA 80 , 594598 .
                                                                                                                                                                                                                                         10. Gold, M. S. , Dastmalchi, S. & Levine, J. D. ( 1996 ) Neuroscience 71 , 265275 .
                                                                                                                                                                                                                                         11. Tanner, K. D. , Gold, M. S. , Reichling, D. B. & Levine, J. D. ( 1997 ) in Molecular Neurobiology of Pain , ed. Borsook, D. ( Int. Assoc. Study of
                                                                                                                                                                                                                                              Pain , Seattle ), Vol. 9 , pp. 79105 .
                                                                                                                                                                                                                                         12. Cantrell, A. R. , Ma, J. Y. , Scheuer, T. & Catterall, W. A. ( 1996 ) Neuron 16 , 10191026 .
                                                                                                                                                                                                                                         13. Cantrell, A. R. , Smith, R. D. , Goldin, A. L , Scheuer, T. & Catterall, W. A. ( 1997 ) J. Neurosci. 17 , 73307338 .
                                                                                                                                                                                                                                         14. England, S. , Bevan, S. & Docherty, R. J. ( 1996 ) J. Physiol. (London) 495 , 429440 .
                                                                                                                                                                                                                                         15. Ono, K. , Fozzard, H. A. & Hanck, D. A. ( 1993 ) Circ. Res. 72 , 807815 .
                                                                                                                                                                                                                                         16. Abram, S. E. & Yaksh, T. L. ( 1994 ) Anesthesiology 80 , 383391 .
                                                                                                                                                                                                                                         17. Chabal, C. , Russell, L. C. & Burchiel, K. J. ( 1989 ) Pain 38 , 333338 .
                                                                                                                                                                                                                                         18. Puig, S. & Sorkin, L. S. ( 1996 ) Pain 64 , 345355 .
                                                                                                                                                                                                                                         19. Devor, M. , Wall, P. D. & Catalan, N. ( 1992 ) Pain 48 , 261268 .
                                                                                                                                                                                                                                         20. Chabal, C. , Jacobson, L , Russell, L. C. & Burchiel, K. J. ( 1992 ) Pain 49 , 912 .
                                                                                                                                                                                                                                         21. Rizzo, M. A. ( 1997 ) J. Neurol. Sci. 152 , 103106 .
                                                                                                                                                                                                                                         22. Ritter, A. M. & Mendell, L. M. ( 1992 ) J. Neurophysiol. 68 , 20332041 .
                                                                                                                                                                                                                                         23. Villiere, V. & McLachlan, E. M. ( 1996 ) J. Neurophysiol. 76 , 19241941 .
                                                                                                                                                                                                                                         24. Arbuckle, J. B. & Docherty, R. J. ( 1995 ) Neurosci. Lett. 185 , 7073 .
                                                                                                                                                                                                                                         25. Caffrey, J. M. , Eng, D. L. , Black, J. A. , Waxman, S. G. & Kocsis, J. D. ( 1992 ) Brain Res. 592 , 283297 .
                                                                                                                                                                                                                                         26. Elliott, A. A. & Elliott, J. R. ( 1993 ) J. Physiol. (London) 463 , 3956 .
                                                                                                                                                                                                                                         27. Ogata, N. & Tatebayashi, H. ( 1993 ) J. Physiol. (London) 466 , 937 .
                                                                                                                                                                                                                                         28. Roy, M. L. & Narahashi, T. ( 1992 ) J. Neurosci. 12 , 21042111 .
                                                                                                                                                                                                                                         29. Frohnwieser, B. , Weigl, L. & Schreibmayer, W. ( 1995 ) Pflugers Arch. 430 , 751753 .
                                                                                                                                                                                                                                         30. Donahue, L. M. ( 1995 ) Neurochem. Res. 20 , 713717 .
                                                                                                                                                                                                                                         31. Thio, C. L. & Sontheimer, H. ( 1993 ) J. Neurosci. 13 , 48894897 .
                                                                                                                                                                                                                                         32. Akopian, A. N. , Sivilotti, L. & Wood, J. N. ( 1996 ) Nature (London) 379 , 257262 .
                                                                                                                                                                                                                                         33. Sangameswaran, L , Delgado, S. G. , Fish, L. M. , Koch, B. D. , Jakeman, L. B. , Stewart, G. R. , Sze, P. , Hunter, J. C. , Eglen, R. M. & Herman, R.
                                                                                                                                                                                                                                              C. ( 1996 ) J. Biol. Chem. 271 , 59535956 .
                                                                                                                                                                                                                                         34. Oaklander, A. L. & Belzberg, A. J. ( 1997 ) Brain Res. Mol. Brain Res. 52 , 162165 .
                                                                                                                                                                                                                                         35. Dib-Hajj, S. D. , Tyrrell, L , Black, J. A. & Waxman, S. G. ( 1998 ) Proc. Natl. Acad. Sci. USA 95 , 89638968 .
                                                                                                                                                                                                                                         36. Tate, S. , Benn, S. , Hick, C. , Trezise, D. , John, V. , Mannion, R. J. , Costigan, M. , Plumpton, C. , Grose, D. , Gladwell, Z. , et al. ( 1998 ) Nat.
                                                                                                                                                                                                                                              Neurosci. 1 , 653655 .
                                                                                                                                                                                                                                         37. Scholz, A. , Appel, N. & Vogel, W. ( 1998 ) Eur. J. Neurosci. 10 Suppl. , 25472556 .
                                                                                                                                                                                                                                         38. Rush, A. M. , Brau, M. E. , Elliott, A. A. & Elliott, J. R. ( 1998 ) J. Physiol. (London) 511 , 771789 .
                                                                                                                                                                                                                                         39. Schwartz, A. , Palti, Y. & Meiri, H. ( 1990 ) J. Membr. Biol. 116 , 117128 .
                                                                                                                                                                                                                                         40. Kostyuk, P. G. , Veselovsky, N. S. , Fedulova, S. A. & Tsyndrenko, A. Y. ( 1981 ) Neuroscience 6 , 24242430 .
                                                                                                                                                                                                                                         41. Schild, J. H. & Kunze, D. L. ( 1997 ) J. Neurophysiol. 78 , 3198 3209 .
                                                                                                                                                                                                                                         42. Levine, J. D. , Basbaum, A. I. & Fields, H. L. ( 1993 ) J. Neurosci. 13 , 22732286 .
                                                                                                                                                                                                                                         43. Cui, M. & Nicol, G. D. ( 1995 ) Neuroscience 66 , 459466 .
                                                                                                                                                                                                                                         44. Taiwo, Y. O. , Goetzl, E. J. & Levine, J. D. ( 1987 ) Brain Res. 423 , 333337 .
                                                                                                                                                                                                                                         45. Cardenas, C. G. , Del Mar, L. P. , Cooper, B. Y. & Scroggs, R. S. ( 1997 ) J. Neurosci. 17 , 71817189 .
                                                                                                                                                                                                                                         46. Levine, J. D. & Taiwo, Y. O. ( 1989 ) Neuroscience 32 , 571575 .
                                                                                                                                                                                                                                         47. Gold, M. S. & Levine, J. D. ( 1996 ) Neurosci. Lett. 212 , 8386 .
the authoritative version for attribution.
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
of the AHPslow. An analogous slow AHP has also been recently described in 25% of C type dorsal root ganglion neurons of the rat (9, 10).
                                                                                                                                                                                                                                           FIG. 1. A single AP can evoke three types of AHP in nodose neurons. (Top) A neuron with a single-component afterpotential lasting
                                                                                                                                                                                                                                           30 ms. This AHP is designated AHPfast. All neurons have this short duration afterpotential. (Middle) Example of a neuron with two
                                                                                                                                                                                                                                           afterpotentials, an AHPfast followed by a longer lasting afterpotential (300 ms), the AHPmedium. In approximately half of the neurons,
                                                                                                                                                                                                                                           the AHPmedium is Ca2+-dependent. (Bottom) In a subset of C fiber type nodose neurons, a slowly developing (hundreds of ms) and
                                                                                                                                                                                                                                           long-lasting (215 s) afterpotential is observed. This slow afterpotential (AHPslow) is always Ca2+-dependent. Intracellular recordings
                                                                                                                                                                                                                                           were obtained at room temperature from adult neurons isolated from rabbit nodose ganglia. The values near the horizontal lines are
                                                                                                                                                                                                                                           resting membrane potentials. The calibration in the Top also applies to the Middle. Similar results have been recorded in guinea pig
                                                                                                                                                                                                                                           and ferret nodose neurons.
                                                                                                                                                                                                                                              The AHPslow in vagal afferent neurons influences cellular excitability and controls AP frequency over the physiological range from
                                                                                                                                                                                                                                         0.1 Hz to 10 Hz ( 11 , 12 ). One interesting property of the AHPslow is that its amplitude is tuned to both AP number and frequency. Over
                                                                                                                                                                                                                                         the range of 1100 Hz, the amplitude of the AHPslow increases with the number of APs until it plateaus after 15 APs ( Fig. 2 ); similar
                                                                                                                                                                                                                                         results were observed when the current underlying the AHPslow was monitored. For reasons still unresolved, 10 Hz (100-msec interspike
                                                                                                                                                                                                                                         intervals) consistently evokes the largest responses.
                                                                                                                                                                                                                                         Table 1. Inflammatory mediators that block AHPslow in vagal afferent neurons
                                                                                                                                                                                                                                         Mediator                                          Receptor type                                         EC50, nM
                                                                                                                                                                                                                                         Bradykinin                                        B2                                                    72
                                                                                                                                                                                                                                         Histamine                                         H1                                                    2,000
                                                                                                                                                                                                                                         Serotonin                                         nd                                                    300
                                                                                                                                                                                                                                         PGD2, PGE2                                        nd                                                    20
                                                                                                                                                                                                                                         Leukotriene C4                                    nd                                                    100
                                                                                                                                                                                                                                         Bradykinin ( 26 ), histamine ( 27 ), serotonin ( 28 ), PGD2 and PGE2 ( 12 ), and leukotriene C4 ( 3 ) block the AHPslow. nd, not determined;
                                                                                                                                                                                                                                         PG prostaglandin.
                                                                                                                                                                                                                                           FIG. 2. Effects of varying numbers of APs and frequency on the amplitude of the AHPslow. All data points were recorded from a
                                                                                                                                                                                                                                           single acutely dissociated adult rabbit nodose neuron at room temperature. Resting potential and membrane input resistance were 55
                                                                                                                                                                                                                                           mV and 53 M, respectively. APs were evoked by transmembrane depolarizing current pulses (2 nA, 3 ms) at the frequencies
                                                                                                                                                                                                                                           indicated. Similar results were obtained when measuring IAHP by using the hybrid voltage-clamp technique in rabbit, guinea pig, and
                                                                                                                                                                                                                                           ferret nodose neurons.
                                                                                                                                                                                                                                              The current generating the AHPslow (IAHP) is a voltage-insensitive Ca2+-dependent K+ current ( 13 , 14 ) that is unaffected by a wide
                                                                                                                                                                                                                                         range of K+ channel antagonists: 100 nM apamin, 10 M d-tubocurarine, 5 mM Cs+, 30 mM tetraethylammonium, 10 mM Ba2+, 4 mM 4-
                                                                                                                                                                                                                                         aminopyridine, and 10 nM charybdotoxin. The magnitude of the AHPslow (or the IAHP) is linearly related to the concentration of
                                                                                                                                                                                                                                         extracellular Ca2+ ( Fig. 3 ) and requires a rise in cytosolic free Ca2+ ([Ca2+]i) for activation. Buffering intracellular Ca2+ with 1,2-bis(2-
                                                                                                                                                                                                                                         aminophenoxy)ethane-N, N, N, N-tetraacetic acid (BAPTA) abolishes the AHPslow ( Fig. 4 ). Noise analysis of the IAHP suggests a single-
                                                                                                                                                                                                                                         channel conductance of 10 pS (unpublished observations). These features are consistent with the properties of a small-conductance Ca2+-
                                                                                                                                                                                                                                         activated K+ channel (SK channel; ref. 8 ). Of the several SK channels recently cloned from mammalian brain ( 15 ), the hSK1 channel
                                                                                                                                                                                                                                         has a pharmacological and biophysical profile compatible with the K+ current underlying the AHPslow in nodose neurons.
the authoritative version for attribution.
                                                                                                                                                                                                                                              Ca2+ Injection Evokes Two Temporally Distinct Outward Currents. To test whether the K+ channels associated with the
                                                                                                                                                                                                                                         AHPmedium and the AHPslow are directly activated by Ca2+, we iontophoretically injected Ca2+ into nodose neurons. Independent of the
                                                                                                                                                                                                                                         AHPslow, a large outward current with rapid activation and decay kinetics was elicited by Ca2+ injection. This current (IK-medium) was
                                                                                                                                                                                                                                         evoked at holding potentials between 2 mV and 45 mV. It was completely blocked by 5 mM tetraethylammonium but unaffected by
                                                                                                                                                                                                                                         inhibitors of the AHPslow (100 nM prostaglandins D2 or E2 or 1 M forskolin). IK-medium was strongly voltage-dependent, requiring
                                                                                                                                                                                                                                         membrane holding potentials more positive than 55 mV. Assuming a reversal potential of 80 mV, IK-medium had an e-fold increase in
                                                                                                                                                                                                                                         peak conductance for each 8.0 1.0 mV (mean  SEM; n = 8) depolarization, as calculated from semilogarithmic plots of peak chord
                                                                                                                                                                                                                                         conductance versus voltage-clamp holding potential. These properties are similar to those of large-conductance BK (AHPmedium) channels.
                                                                                                                                                                                                                                              In neurons that exhibited AHPslow, Ca2+ injection provoked a slowly developing and protracted outward current (IK-slow).
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         Fig. 5 shows an overlay of the outward current responses evoked by Ca2+ injection in a single nodose C type neuron at holding potentials
                                                                                                                                                                                                                                         of 20 mV and 50 mV. The kinetic differences between IK-medium and IK-slow after Ca2+ injection are dramatic. In contrast to the rapid
                                                                                                                                                                                                                                         activation of IK-medium, the onset of IK-slow is delayed, and the decay of IK-medium is nearly complete before the peak amplitude of the IK-
                                                                                                                                                                                                                                         slow is reached. These two outward currents mirror the temporal and pharmacological differences between AHPmedium and AHPslow IK-slow,
                                                                                                                                                                                                                                         like the AHPslow, was blocked by 100 nM prostaglandin D2. The data shown in Table 2 summarize quantitative differences between these
                                                                                                                                                                                                                                         two Ca2+-induced outward currents.
                                                                                                                                                                                                                                           FIG. 3. Effects of varying [Ca2+]o on the amplitude of the AHPslow recorded in isolated nodose neurons. (A) Sample traces of
                                                                                                                                                                                                                                           AHPslow evoked by a train of four APs in the presence of different [Ca2+]o. APs are evoked by transmembrane depolarizing current
                                                                                                                                                                                                                                           pulses (2 nA, 3 ms, 10 Hz) and are truncated. [Ca2+]o was varied from 2.0 to 0.0 mM in 0.5 mM decrements. The AHPslow is
                                                                                                                                                                                                                                           completely blocked when [Ca2+]o is reduced to nominally zero. On returning to 2.0 mM [Ca2+]o, the AHPslow recovers to its original
                                                                                                                                                                                                                                           amplitude. (B) Relation between [Ca2+]o and AHPslow amplitude recorded in several neurons. Values are means  SEM of the number
                                                                                                                                                                                                                                           of observations indicated near each data point. Data are normalized to the maximum response recorded in a given neuron. Linear
                                                                                                                                                                                                                                           regression analysis yields the solid line (r = 0.993).
                                                                                                                                                                                                                                               It is possible that the delayed onset of IK-slow compared with IK-medium results from unequal Ca2+ diffusion distances from the injection
                                                                                                                                                                                                                                         site to the two types of K+ channels. This cause seems unlikely because the orientation of impalement was random, and the plasma
                                                                                                                                                                                                                                         membranes of dissociated nodose neurons appear devoid of processes that would provide semiisolated regions where IK-slow might be
                                                                                                                                                                                                                                         generated. An alternative possibility is that additional intermediate steps, such as the synthesis or release of a second messenger, are
                                                                                                                                                                                                                                         required to activate IK-slow. The large Q10 (>3.0; ref. 14 ) supports the latter alternative. One candidate is mobilization of intracellularly
                                                                                                                                                                                                                                         stored Ca2+.
                                                                                                                                                                                                                                               Ca2+ Released by the CICR Pool Is Essential for the Generation of the AHPslow. Single APs produce transient increases in [Ca2+]i
                                                                                                                                                                                                                                         (Cat) as measured by the fluorescent indicator fura-2. The magnitude of the Cat depends on both [Ca2+]o and the number of APs. Over
                                                                                                                                                                                                                                         the range of one to eight APs, there is an approximately linear relation between the magnitude of the Cat and the number of APs (
                                                                                                                                                                                                                                         Fig. 6 ). In the presence of drugs that block CICR but do not significantly affect AP-induced Ca2+ influx [(RY, 10 M), 2,5,-di(t-butyl)
                                                                                                                                                                                                                                         hydroquinone (DBHQ, 10 M), or thapsigargin (TG, 100 nM)], we found that at least eight APs were required to evoke a detectable Cat (
                                                                                                                                                                                                                                         Fig. 6 ). In the presence of RY, DBHQ, and TG, the CatAP relation exhibits slopes of 0.5, 1.1, and 0.8 nM per AP, respectively. When
                                                                                                                                                                                                                                         compared with the slope of 9.6 nM per AP in control neurons, Ca2+ influx produced by a single nodose AP is amplified by 5- to 10-fold by
                                                                                                                                                                                                                                         CICR ( 16 ). Nodose neurons demonstrate a relatively low stimulus threshold for eliciting CICR. For instance, a robust CICR response can
                                                                                                                                                                                                                                         be observed after a single AP stimulus in nodose neurons, whereas many tens of APs are required in dorsal root ganglion neurons ( 17 ).
                                                                                                                                                                                                                                         The greater CICR response in nodose neurons is not due to greater Ca2+ influx through voltage-dependent Ca2+ channels (VDCCs); a
                                                                                                                                                                                                                                         single AP produces comparable Ca2+ influx in nodose and dorsal root ganglion neurons (39 vs. 49 pC, respectively; refs. 16 and 18 ).
                                                                                                                                                                                                                                         Rather, the more responsive CICR pool in nodose neurons
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 4. Effects of BAPTA on the AHPslow and on the excitability of an acutely dissociated rabbit nodose neuron. (A) Bath-applied
                                                                                                                                                                                                                                           BAPTA/acetomethylester (10 M) blocks the AHPslow within 5 min without changing the resting membrane potential or membrane
                                                                                                                                                                                                                                           input resistance. APs were evoked by transmembrane depolarizing current pulses (4 nA, 1.5 ms, 10 Hz) and are truncated. (B)
                                                                                                                                                                                                                                           Responses recorded at a faster sweep speed to illustrate the kinetics of the AHPfast and AHPmedium, which precede the AHPslow. The
                                                                                                                                                                                                                                           AHPfast is unaffected by 10 M BAPTA/acetomethylester (compare a with b). The Ca2+ dependence of the AHPmedium is illustrated in
                                                                                                                                                                                                                                           c, where the neuron is superfused with 100 M CdCl2 for 30 s, which blocks most of the AHPmedium. The residual component of the
                                                                                                                                                                                                                                           AHP recorded in CdCl2 is the AHPfast, which is mediated by delayed rectifier K+ channels. (C) Depression of the AHPslow markedly
                                                                                                                                                                                                                                           increases neuronal excitability. The average AP firing frequency induced by a current ramp protocol (1 nA, 2 s) increased from 1 to
                                                                                                                                                                                                                                           5.5 Hz when the AHPslow was blocked. Similar loss of spike-frequency adaptation was observed with bradykinin, prostaglandin D2,
                                                                                                                                                                                                                                           histamine, and other inflammatory autacoids (see Table 2 ). The scale bar represents 3 mV, 2 s in A; 15 mV, 0.25 s in B; and 15 mV,
                                                                                                                                                                                                                                           0.5 s in C. The dashed line represents the resting membrane potential (60 mV). Resting membrane input resistance was 70 M.
                                                                                                                                                                                                                                           Data is from ref. 19 with permission from the American Physiological Society.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         may reflect either a closer proximity between plasma membrane Ca2+ influx channels and endoplasmic reticulum RY receptors or a more
                                                                                                                                                                                                                                         sensitive RY receptor.
                                                                                                                                                                                                                                           FIG. 5. Comparison of two outward K+ currents evoked by intracellular Ca2+ injection. Recordings were made in a single acutely
                                                                                                                                                                                                                                           isolated adult rabbit nodose neuron. A slow outward current (IK-slow) was activated by a 5-nA, 1-s iontophoretic Ca2+ injection at a
                                                                                                                                                                                                                                           holding potential of 50 mV. A second outward current (IK-medium) was activated at 20 mV (5 nA, 0.5 sec). IK-medium activates and
                                                                                                                                                                                                                                           decays blocked by 10 mM tetraethylammonium; IK-slow was blocked by 100 nM completely before IK-slow reaches peak amplitude. IK-
                                                                                                                                                                                                                                           medium was prostaglandin D2. The iontophoretic pipette was filled with a 0.2 M CaCl2 solution. Voltage-clamp currents were recorded
                                                                                                                                                                                                                                           with a second intracellular pipette. The discontinuous (switched) current injection mode of an Axoclamp II amplifier was used for
                                                                                                                                                                                                                                           both currentand voltage-clamp applications. The larger calibration value is for IK-medium. Population data is shown in Table 2 .
                                                                                                                                                                                                                                              By using physiological stimuli (APs) in conjunction with pharmacological manipulations of CICR, we have demonstrated that CICR
                                                                                                                                                                                                                                         is essential for the development of the AHPslow. Over the range of 116 APs, the magnitudes of the AP-induced AHPslow and the Cat (a
                                                                                                                                                                                                                                         monitor of CICR in these neurons) were highly correlated (r = 0.985). Simultaneous recordings of Cat and AHPslow before and during
                                                                                                                                                                                                                                         bath application of CICR inhibitors (RY, TG, DBHQ, or 10 M cyclopiazonic acid) revealed that both responses were blocked in a
                                                                                                                                                                                                                                         parallel fashion ( Fig. 7 ; see also Table 1 in ref. 19 ). These data indicate that a CICR pool is essential for the generation of the AHPslow.
                                                                                                                                                                                                                                         They also provide a potential explanation for the slow kinetics of the AHPslow, namely Ca2+ mobilization from CICR.
                                                                                                                                                                                                                                              Effects of Changing [Ca2+]o on the AHPslow, Cat, and Ca2+ influx. If the AHPslow depends on Ca2+ released from the CICR pool
                                                                                                                                                                                                                                         triggered by AP-induced Ca2+ influx, it would follow that changes in [Ca2+]o should produce corresponding effects on both the AHPslow
                                                                                                                                                                                                                                         and the Cat. The data shown in Fig. 3 A illustrate the effects of progressively lowering [Ca2+]o from 2.0 mM to nominally zero on the
                                                                                                                                                                                                                                         amplitude of the AHPslow recorded in a single nodose neuron. As [Ca2+]o was decreased, the amplitude of the AHPslow was reduced
                                                                                                                                                                                                                                         proportionally. When the results from this and five additional neurons were plotted ( Fig. 3 B), the relation between [Ca2+]o and the
                                                                                                                                                                                                                                         amplitude of the AHPslow was linear (r = 0.993; n = 6, pooled data from three current-clamp and three hybrid voltage-clamp experiments).
                                                                                                                                                                                                                                         Table 2. Comparison of IK-slow and IK-medium
                                                                                                                                                                                                                                         Current    Peak conductance,      n    Holding                      n      Time-to-           n      Decay time           n     Duration, s     n
                                                                                                                                                                                                                                                    nS                          potential, mV                       peak, ms                  constant, ms
                                                                                                                                                                                                                                         IK-slow    27.9  6.5             14   55.4  2.7                  14     6,570  1085       12     6,735  789          5     23  3.4        14
                                                                                                                                                                                                                                         IK-medium  53.2  16.5            6    20  3.7                    6      958  56           6      818  97             6     2.5  0.16      6
                                                                                                                                                                                                                                                                                                                             2+ into acutely isolated nodose neurons of the rabbit. The peak
                                                                                                                                                                                                                                         K-slow and IK- medium are outward currents elicited by iontophoretic injection Ca
                                                                                                                                                                                                                                         conductance is the largest conductance elicited, independent of membrane potential. The holding potential is the potential at which the peak conductance
                                                                                                                                                                                                                                         was measured. The decay time constant was measured by fitting a line, by eye, to the log transform of the decay of the current. The duration was
                                                                                                                                                                                                                                         calculated from the onset of Ca2+ injection to the time at which the current had decayed to 20% of its peak value. Data are summarized as the mean 
                                                                                                                                                                                                                                         SEM.
                                                                                                                                                                                                                                           FIG. 6. (Upper) Effect of RY on AP-induced Ca2+ transients. Traces are Ca2+ transients evoked by varying numbers of APs, as
                                                                                                                                                                                                                                           indicated below each trace. In control neurons, distinct Ca2+ transients can be elicited by very few APs. In contrast, in the presence of
                                                                                                                                                                                                                                           10 M RY, a CICR inhibitor, at least eight APs are required to generate a discernible change in [Ca2+]i. Suppression of the Ca2+
                                                                                                                                                                                                                                           transient by RY is due to its effect on CICR and not the result of nonspecific effects on Ca2+ channels; the kinetics and amplitude of
                                                                                                                                                                                                                                           ICa elicited by APs are completely unaffected by RY. (Lower) Effect of RY on the relation between the amplitude of Ca2+ transients
the authoritative version for attribution.
                                                                                                                                                                                                                                           and number of APs.  and  are mean amplitudes of Ca2+ transients evoked by varying numbers of action potentials for control (n =
                                                                                                                                                                                                                                           10) and for RY-treated nodose neurons (n = 3), respectively. Linear regression of data from control (4 action potentials) and RY-
                                                                                                                                                                                                                                           treated cells yielded slopes of 9.6  0.01 and 0.5  0.23 nM per AP, respectively. Comparison of the slopes illustrates that CICR is
                                                                                                                                                                                                                                           capable of amplifying the trigger Ca2+ resulting from AP-induced Ca2+ influx by 20-fold. Data is modified from ref. 16 with
                                                                                                                                                                                                                                           permission from Journal of Physiology (London).
                                                                                                                                                                                                                                              Next, we examined the relation between [Ca2+]o and the magnitude of the AP-induced Cat. Fig. 8 A illustrates Cats elicited by
                                                                                                                                                                                                                                         varying numbers of APs recorded from a single neuron in Locke solution containing 2.2 or 1.1 mM Ca2+. The population results relating
                                                                                                                                                                                                                                         the normalized amplitude of the Cats recorded in four neurons to the number of APs is shown in Fig. 8 B. In 1.1 mM [Ca2+]o, the first
                                                                                                                                                                                                                                         few APs did not elicit a measurable Cat. For the neuron shown in Fig. 8 A, at least eight APs were necessary to evoke a detectable Cat.
                                                                                                                                                                                                                                         In three additional neurons, the minimum number of APs necessary to
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         elicit a detectable Cat ranged from 4 to 32. The CatAP relation recorded in 1.1 mM [Ca2+]o, as in Locke solution containing normal
                                                                                                                                                                                                                                         [Ca2+]o, followed a hyperbolic relation (2 = 6.75 and 0.31; r = 0.988 and 0.999 for 2.2 and 1.1 mM Ca2+,respectively; Fig. 8 B and see
                                                                                                                                                                                                                                         also Fig. 1 in ref. 16 ). Given the hyperbolic nature of the CatAP relation, deducing the effects of altered [Ca2+]o on the magnitude of the
                                                                                                                                                                                                                                         Cat clearly depends on where along this relation the comparison is made. At one extreme, there is a 2-fold change when comparing the
                                                                                                                                                                                                                                         plateau phases of the curves in normal and one-half normal [Ca2+]o. It is also possible to calculate the limiting initial slopes for the rising
                                                                                                                                                                                                                                         phase of the curves (dashed lines in Fig. 8 B). The limiting slopes, which represent the full Ca2+ release potential of the CICR pool before
                                                                                                                                                                                                                                         any release has actually occurred, were 15  3.8 and 2  0.7 nM per AP in 2.2 and 1.1 mM [Ca2+]o, respectively. Thus, reducing [Ca2+]o by
                                                                                                                                                                                                                                         a factor of 2 results in a reduction of the Cat by a factor of 7  2.8 when the rising phases of the two curves are compared. The 7-fold
                                                                                                                                                                                                                                         reduction of the Cat associated with halving [Ca2+]o is much larger than the 2-fold reduction in the AHPslow amplitude ( Fig. 3 ),
                                                                                                                                                                                                                                         suggesting that some, but not all, of the Ca2+ released from the CICR pool is required for the generation of the AHPslow.
                                                                                                                                                                                                                                           FIG. 7. Effect of DBHQ, a functional CICR inhibitor, on the AP-induced Ca2+ transient and on the AHPslow recorded simultaneously
                                                                                                                                                                                                                                           in an acutely isolated rabbit nodose neuron. Upper traces represent superimposed Ca2+ transients evoked by a train of four APs (10
                                                                                                                                                                                                                                           Hz) recorded in control Locke solution and 7 min after switching to Locke solution containing 10 M DBHQ. The lower pair of
                                                                                                                                                                                                                                           traces shows AHPslow. DBHQ treatment completely blocked both the Ca2+ transient and the AHPslow. Resting [Ca2+]i was 91 nM.
                                                                                                                                                                                                                                           Fluorescence data were acquired at 10 Hz. Resting membrane potential was 67 mV. AP amplitudes are truncated. Data are from ref.
                                                                                                                                                                                                                                           19 with permission from the American Physiological Society.
                                                                                                                                                                                                                                              The disproportionate effect of reduced [Ca2+]o on the Cat versus the AHPslow could arise from a nonlinear reduction of Ca2+ influx
                                                                                                                                                                                                                                         per AP and/or from a decreased Ca2+ release from CICR pool per unit Ca2+ influx. To study these possibilities, we examined the effect of
                                                                                                                                                                                                                                         lowering [Ca2+]o on APinduced Ca2+ influx. The amount of Ca2+ entering a neuron with each AP in normal and in reduced [Ca2+]o was
                                                                                                                                                                                                                                         determined by using a prerecorded AP as whole-cell voltage-clamp command under experimental conditions where the major inward
                                                                                                                                                                                                                                         charge carrier is Ca2+ (for details, see Fig. 2 in ref. 16 ). When [Ca2+]o was decrementally reduced from 2 mM to nominally zero, the
                                                                                                                                                                                                                                         magnitude of the ICa decreased proportionally. The charge movement caused by Ca2+ influx, normalized to cell membrane capacitance (pC/
                                                                                                                                                                                                                                         pF), was plotted against varying [Ca2+]o for 12 neurons. Over the range of 02.0 mM [Ca2+]o, Ca2+ influx varied linearly with [Ca2+]o (r =
                                                                                                                                                                                                                                         0.974). These results indicate that changes in Ca2+ influx alone cannot account for the disproportionate reduction in the Cat relative to the
                                                                                                                                                                                                                                         AHPslow that is observed when [Ca2+]o is reduced.
                                                                                                                                                                                                                                           FIG. 8. Effect of varying [Ca2+]o on the amplitude of AP-induced Ca2+ transients. (A) Representative traces of Ca2+ transients evoked
                                                                                                                                                                                                                                           by varying numbers of APs in normal (2.2 mM) and reduced (1.1 mM) [Ca2+]o. APs were elicited by transmembrane depolarizing
                                                                                                                                                                                                                                           current pulses (2 nA, 1.5 ms, 10 Hz). The number of APs is indicated below each trace. (B) The normalized (mean  SEM) amplitude
                                                                                                                                                                                                                                           of Ca2+ transients recorded in four neurons is plotted against varying numbers of APs. Data are normalized to the maximal response
                                                                                                                                                                                                                                           recorded in a given neuron. represents Ca2+ transients recorded in 2.2 mM [Ca2+] o;  represents Ca2+ transients recorded in the
                                                                                                                                                                                                                                           same neurons in 1.1 mM [Ca2+]o. Continuous curves are rectangular hyperbolas fit to the data (2 = 6.75 and 0.31, r = 0.988 and 0.999
                                                                                                                                                                                                                                           for 2.2 and 1.1 mM [Ca2+]o, respectively). The dashed lines represent the limiting initial slopes (15  3.8 and 2  0.7 nM per AP for
                                                                                                                                                                                                                                           2.2 and 1.1 mM [Ca2+]o, respectively).
                                                                                                                                                                                                                                               The disproportionate effect of reduced [Ca2+]o on the CatAHPslow relation could arise from a diminution in the amount of Ca2+
                                                                                                                                                                                                                                         released from the CICR pool. Caffeine, a known agonist of CICR, is traditionally used to assess the releasable content of the CICR pool.
                                                                                                                                                                                                                                         In 8 of the 13 neurons studied, halving [Ca2+]o reduced the caffeine-induced Cat by 2079% (100% vs. 47  7.2% in 2.2 and 1.1 mM [Ca2+]
                                                                                                                                                                                                                                                                                                       2+
                                                                                                                                                                                                                                         o, respectively; P = 0.0002). In other words, decreasing [Ca ]o by a factor of 2 caused a 1.25- to 5-fold reduction in the caffeine response.
the authoritative version for attribution.
                                                                                                                                                                                                                                         On returning to normal Locke solution, the caffeine response was restored to near control values. In the remaining five neurons, the
                                                                                                                                                                                                                                         caffeine-induced Cat was unaffected by reducing [Ca2+]o (100% vs. 112  8.4% in 2.2 and 1.1 mM [Ca2+]o, respectively; P = 0.690).
                                                                                                                                                                                                                                         There was no significant difference in resting levels of [Ca2+]i between these two groups of neurons (93  29.5 nM vs. 111  29.7 nM; P =
                                                                                                                                                                                                                                         0.530). Unfortunately, the wide variability in the effects of reduced [Ca2+]o on the caffeine responses prevents a meaningful interpretation
                                                                                                                                                                                                                                         of the effect of [Ca2+]o on the releasable content of the CICR pool.
                                                                                                                                                                                                                                               Ca2+ Influx Through N Type Calcium Channels Selectively Elicits AHPslow. Six types of VDCCs have been described in neurons:
                                                                                                                                                                                                                                         L, N, P, Q, R, and T ( 20 ). Nodose neurons express several types of VDCCs. By using a panel of pharmacologic reagents that are selective
                                                                                                                                                                                                                                         for different types of VDCCs, we tested the contribution of each to the total AP-induced Ca2+ current. Our results, summarized in Table 3 ,
                                                                                                                                                                                                                                         reveal that 85%
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         of the AP-induced inward Ca2+ current is shared by L and N type Ca2+ channels ( Fig. 9 ). P, Q, and T type Ca2+ channel antagonists were
                                                                                                                                                                                                                                         ineffective, suggesting that the remaining Ca2+ current is associated with Ca2+ influx through R type channels. Nifedipine (10 M), an L
                                                                                                                                                                                                                                         type Ca2+ channel blocker, produced no measurable effect on either the AHPfast, the AHPmedium, or the AHPslow. By contrast, -conotoxin-
                                                                                                                                                                                                                                         GVIA (0.5 M), a selective N type Ca2+ channel blocker, always obliterated the AHPslow, and in 50% of the neurons abolished the
                                                                                                                                                                                                                                         AHPmedium (about half of the AHPmedium are Ca2+sensitive, see above), while leaving the AHPfast unaffected ( Fig. 9 and Table 4 .).
                                                                                                                                                                                                                                         These results indicate that the SK and BK type K+ channels are both regulated by Ca2+ influx through N type channels. BK channels are
                                                                                                                                                                                                                                         gated by influx Ca2+ directly (8), whereas SK channels are affected by influx Ca2+ indirectly (i.e., Ca2+ entering through N type VDCC
                                                                                                                                                                                                                                         triggers RY receptors to release Ca2+ from CICR pools). Such a sequence implies a functional coupling between N type Ca2+ channels
                                                                                                                                                                                                                                           FIG. 9. Effects of VDCC antagonists on AP-induced calcium currents, AHPslow and AP-induced Ca2+ transients. (A) Inward calcium
                                                                                                                                                                                                                                           currents recorded in isolated nodose neurons evoked by a prerecorded AP waveform from a holding potential of 60 mV. From Left
                                                                                                                                                                                                                                           to Right, control inward current in the presence of 2 mM [Ca2+]o and in the presence of 10 M nifedipine. After reestablishing control
                                                                                                                                                                                                                                           conditions, the neuron was exposed to 1 M -conotoxin-GVIA. The effects of 500 M cadmium were recorded in another neuron;
                                                                                                                                                                                                                                           the control current for this cell was similar to the first trace. (B) AHPslow evoked by a train of four APs (10 Hz) recorded in another
                                                                                                                                                                                                                                           nodose neuron. From Left to Right, AHPslow evoked in control conditions, in the presence of 100 M CdCl2, after washout, in the
                                                                                                                                                                                                                                           presence of 500 nM -conotoxin-GVIA, and after washout. (C) AP-induced Ca2+ transients recorded in two nodose neurons. From
                                                                                                                                                                                                                                           Left to Right, Ca2+ transients evoked by a train of eight APs in normal Locke solution, and in Locke solution containing 10 M
                                                                                                                                                                                                                                           nifedipine. In another neuron, 1 M -conotoxin-GVIA reduced the Ca2+ transient 50% (see Table 4 ). APs were evoked by 2.5-ms,
                                                                                                                                                                                                                                           10-Hz depolarizing current pulses.
                                                                                                                                                                                                                                         Table 3. Effects of Ca2+ channel blockers on action potential-induced inward Ca2+ currents
                                                                                                                                                                                                                                         Channel type                 Channel blocker                     Concentration M                      Reduction                n
                                                                                                                                                                                                                                         T                            Amiloride                           500                                   00                      18
                                                                                                                                                                                                                                         L                            Nifedipine                          10                                    44  5.6                 9
                                                                                                                                                                                                                                         P/Q                          -AGA IVA                           0.2                                   00                      2
                                                                                                                                                                                                                                         Q                            -CTX MVIIC                         0.25                                  00                      6
                                                                                                                                                                                                                                         N                            -CTX GVIA                          1                                     40  4.0                 15
                                                                                                                                                                                                                                         The blocking effect of amiloride, nifedipine, -agatoxin (AGA) IVA, -conotoxin (CTX) MVIIC, and -conotoxin (CTX) GVIA is
                                                                                                                                                                                                                                         expressed as percent reduction in the peak amplitude of the total calcium current  SEM. n corresponds to the number of cells for each
                                                                                                                                                                                                                                         condition.
                                                                                                                                                                                                                                         Table 4. Actions of specific Ca2+ channel blockers on the action potential-induced Ca2+ transient and the AHP slow
                                                                                                                                                                                                                                                                                                   Reduction, %
                                                                                                                                                                                                                                         Channel type               Channel blocker                Ca2+ transient             n         AHPslow amplitude                     n
                                                                                                                                                                                                                                         L                          Nifedipine                     57  7.7                   21        00                                   5
                                                                                                                                                                                                                                         N                          -CTX GVIA                     39  6.2                   4         100  0                               6
                                                                                                                                                                                                                                         T, R                       Nickel                         nd                                   00                                   5
                                                                                                                                                                                                                                         All                        Cadmium                        100  0                    2         100  0                               6
                                                                                                                                                                                                                                         The following concentrations of antagonists were used: nifedipine (10 M), -conotoxin GVIA (0.5 M or 1 M), nickel (50500 M),
the authoritative version for attribution.
                                                                                                                                                                                                                                              and R Y channels in the endoplasmic reticulum. We tested this proposition by examining the effects of VDCC antagonists on the
                                                                                                                                                                                                                                         magnitude of AP-induced Cat.
                                                                                                                                                                                                                                              Ca2+ influx through both L and N type Ca2+ channels triggers CICR. The magnitude of the Cat is a sensitive indicator of Ca2+
                                                                                                                                                                                                                                         release from the CICR pool. To determine the relative influence of Ca2+ influx through L and N type channels on release from the CICR
                                                                                                                                                                                                                                         pool, we applied selective VDCC antagonists and monitored the amplitude of Cat. Nifedipine (10 M) and -conotoxin-GVIA (0.51.0
                                                                                                                                                                                                                                         M) diminished the amplitude of the Cat by 57% and 39%, respectively ( Fig. 9 and Table 4 ). These results reveal that Ca2+ entering
                                                                                                                                                                                                                                         through either L or N type Ca2+ channels provides trigger Ca2+ to stimulate CICR. Given that the amount of Ca2+ influx through L and N
                                                                                                                                                                                                                                         type Ca2+ channels is comparable (44% and 40%, respectively, of total AP-induced Ca2+ influx; see Table 3 ), there must be a remarkable
                                                                                                                                                                                                                                         spatial arrangement between plasma membrane N type Ca2+ channels, endoplasmic reticulum R Y receptors, and plasma membrane SK
                                                                                                                                                                                                                                         channels. Our working hypothesis concerning the regulation of the AHPslow by Ca2+ is illustrated schematically in Fig. 10 .
                                                                                                                                                                                                                                                                                                         DISCUSSION
                                                                                                                                                                                                                                              Whether recorded in intact vagal sensory ganglia or in acutely isolated vagal afferent somata (nodose neurons), single APs can elicit
                                                                                                                                                                                                                                         an AHPslow that exhibits a delayed onset (50300 ms), a slow time to peak amplitude (0.30.5 s), and a particularly long duration (215 s)
                                                                                                                                                                                                                                         (14, 21). Inhibition of the AHPslow by numerous inflammatory mediators (e.g., bradykinin, prostanoids, histamine, serotonin, leukotriene
                                                                                                                                                                                                                                         C4; see Table 1 ) results in an increased neuronal excitability and a loss of spike-frequency adaptation. Thus, modulation of the AHPslow by
                                                                                                                                                                                                                                         these mediators provides a mechanism for peripheral nociceptor sensitization that may underlie allergic inflammation-induced hyperalgesia.
                                                                                                                                                                                                                                              One unresolved but important mechanistic question revolves around the delayed onset and protracted duration of the AHPslow. Many
                                                                                                                                                                                                                                         of our studies of nodose AHPslow were performed with acutely dissociated adult neurons, which are essentially spherical structures lacking
                                                                                                                                                                                                                                         dendritic and axonal processes. Thus, the delayed onset of the AHPslow cannot be due to slow diffusion of Ca2+ from distal sites of influx
                                                                                                                                                                                                                                         to somal SK channels. The high temperature coefficient (Q10 > 3.0) for the rising phase and the decay time constant of the nodose AHPslow
                                                                                                                                                                                                                                         ( 14 ) also argues against simple Ca2+ diffusion as an explanation for the slow kinetics of the AHPslow. The time course of the AHPslow
                                                                                                                                                                                                                                         could arise from unusual channel kinetics of the SK channels. This also appears unlikely if SK channels in nodose neurons have activation
                                                                                                                                                                                                                                         kinetics similar to those cloned from rat brain ( 22 ). Recombinant SK channels from rat brain have activation time constants that are
                                                                                                                                                                                                                                         orders of magnitude shorter than the rise time of the AHPslow. It is more likely that the time course of the AHPslow is a consequence of the
                                                                                                                                                                                                                                         Cat because of CICR.
                                                                                                                                                                                                                                           FIG. 10. Schematic diagram of the relation between plasma membrane Ca2+ channels, BK, and SK potassium channels and
                                                                                                                                                                                                                                           endoplasmic reticulum R Y receptors in primary vagal afferent neurons. Single APs evoke Ca2+ influx through L and N type VDCCs.
                                                                                                                                                                                                                                           Ca2+ influx through either of these channels can trigger release of Ca2+ from the endoplasmic reticulum via RY receptors. Whereas
                                                                                                                                                                                                                                           BK channels are activated directly by Ca2+ entering the neuron via N type VDCC, SK channels are activated indirectly. SK channels
                                                                                                                                                                                                                                           require Ca2+ from CICR pools released after Ca2+ influx through N type channels.
                                                                                                                                                                                                                                              If the AHPslow is directly dependent on Ca2+ released from the CICR pool, the AHPslow and the AP-induced rise in [Ca2+]i should
                                                                                                                                                                                                                                         display similar kinetics. Quantitative kinetic comparisons between these two variables are subject to some uncertainty, because the time
                                                                                                                                                                                                                                         course of the Cat reflects global changes in [Ca2+ ]I, whereas the kinetics of the AHPslow are determined by events at the plasma
                                                                                                                                                                                                                                         membrane. Nonetheless, we determined the time-to-peak and 10-to-90% decay time for both the AHPslow and the Cat elicited by one to
                                                                                                                                                                                                                                         eight APs ( 19 ). The time-to-peak for AHPslow was significantly slower than the Cat by nearly a factor of a two (1.0 s vs. 1.9 s); the Cat
                                                                                                                                                                                                                                         also decayed more rapidly than the AHPslow (3 s vs. 7 s). Analogous temporal discrepancies have been reported between the Cat and
                                                                                                                                                                                                                                         AHPslow in vagal motoneurons ( 23 ). Such temporal differences suggest that Ca2+ released from CICR pools does not act alone to gate
                                                                                                                                                                                                                                         AHPslow K+ channels. Cloned SK channels contain many potential phosphorylation sites ( 15 ); Ca2+-dependent phosphorylation and/or
                                                                                                                                                                                                                                         dephosphorylation may thus be additional processes in the signaltransduction pathway of AP-evoked AHPslow.
                                                                                                                                                                                                                                              Unambiguous data now exist showing that Ca2+ can directly activate SK channels in hippocampal neurons ( 24 ) and in Xenopus
                                                                                                                                                                                                                                         oocytes ( 22 ). In nodose neurons, it is less clear whether Ca2+ alone is sufficient to activate and sustain the AHPslow after an AP. In
                                                                                                                                                                                                                                         hippocampal neurons, flash photolysis of a caged Ca2+ chelator immediately truncates AP-induced AHPslow, suggesting that elevated
                                                                                                                                                                                                                                         intracellular Ca2+ is required to maintain the AHPslow ( 25 ). These results do not, however, distinguish between continuous Ca2+ gating of
                                                                                                                                                                                                                                         SK channel and the involvement of other Ca2+-dependent factors sustaining the longevity of the AHPslow. It is also possible that Ca2+-
                                                                                                                                                                                                                                         dependent factors act synergistically with Ca2+ to control SK channels ( 23 ). The nearly spherical morphology and large size of acutely
                                                                                                                                                                                                                                         isolated adult nodose neurons provide a favorable preparation to determine the nature of second messengers required to activate and
                                                                                                                                                                                                                                         sustain the AHPslow.
                                                                                                                                                                                                                                              In conclusion, a subset of vagal primary afferent neurons possess a slowly developing and long-lasting spike afterhyperpolarization,
                                                                                                                                                                                                                                         the AHPslow, that can profoundly affect the discharge frequency of these visceral afferent neurons. Although AP-evoked Ca2+ influx via
                                                                                                                                                                                                                                         both L and N type Ca2+ channels triggers CICR, only Ca2+ flux through N type channels activates the CICR-dependent AHPslow. This type
                                                                                                                                                                                                                                         of specificity suggests that spatial coupling between N type Ca2+ channels and SK channels may be critical for the generation of the
                                                                                                                                                                                                                                         AHPslow in nodose neurons. The exact mechanism coupling Cat to the AHPslow current remains to be determined.
the authoritative version for attribution.
                                                                                                                                                                                                                                              We thank our coworkers who participated in many of the experiments described in this manuscript: Drs. Akiva Cohen, Samir Jafri,
                                                                                                                                                                                                                                         and Bill Wonderlin, and Mr. Glen Taylor. The authors also thank Dr. Liz Katz and Mr. Eric Lancaster for their constructive suggestions on
                                                                                                                                                                                                                                         an earlier draft of this manuscript. This work was supported by National Institutes of Health Grants GM-46956 to J.P.Y.K., NS-22069 to
                                                                                                                                                                                                                                         D.W. and Training Grant NS-07375 to K.A.M.
                                                                                                                                                                                                                                         1. Undem, B. J. & Riccio, M. M. ( 1997 ) in Asthma , eds. Barnes, P. J. , Grunstein, M. M. , Leff, A. & Woolcock, A. J. ( Lippincott , Philadelphia ), pp.
                                                                                                                                                                                                                                             10091026 .
                                                                                                                                                                                                                                         2. Weinreich, D. ( 1995 ) Pulm. Pharmacol 8 , 173179 .
                                                                                                                                                                                                                                         3. Undem, B. J. , Hubbard, W. & Weinreich, D. ( 1993 ) J. Auton. Nerv. Syst. 44 , 3544 .
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                         in vivo ( 8 ), the heat stimulus causes a rapid depolarization and initiates a train of action potentials. The fact that this response is present in
                                                                                                                                                                                                                                         isolated nociceptors shows that no other cells are necessary to produce the response to heat; there is no signal molecule released from
                                                                                                                                                                                                                                         adjacent damaged cells to which the nociceptor responds. Nor is the response due to damage to the nociceptive neuron itselfas might
                                                                                                                                                                                                                                         occur if heat were causing a breakdown in the plasma membrane and a consequent depolarizationbecause the depolarization and action-
                                                                                                                                                                                                                                         potential firing ceases immediately when the stimulus is withdrawn and because similar responses can be elicited again and again on
                                                                                                                                                                                                                                         repeated application of the heat stimulus.
                                                                                                                                                                                                                                           FIG. 1. Depolarization and a train of action potentials initiated in a nociceptive neuron in culture by application of a brief heat
                                                                                                                                                                                                                                           stimulus. Membrane potential recorded by using whole-cell patch clamp (see ref. 6 for details).
                                                                                                                                                                                                                                              The reproducibility of the response in isolated nociceptors resembles that in other sensory receptors. Interestingly, though, the
                                                                                                                                                                                                                                         behavior of nociceptors in vivo is different. Repeated application of a strong stimulus leads to a progressive increase in the response in
                                                                                                                                                                                                                                         nociceptors in vivo but not in isolated nociceptors nor in other sensory receptors. This process, known as sensitization or hyperalgesia, is
                                                                                                                                                                                                                                         characteristic of nociceptors in vivo and has obvious protective value for the organism as a whole, in that the pain caused by a damaging
                                                                                                                                                                                                                                         stimulus becomes more urgent if the stimulus is repeated or maintained. The fact that sensitization is not observed in isolated nociceptors
                                                                                                                                                                                                                                         suggests that the phenomenon is not intrinsic to the neuron but instead has its origin in extracellular signals released from nearby damaged
                                                                                                                                                                                                                                         or inflamed tissue ( 9 ). Recent advances in our understanding of this process of sensitization are discussed below.
                                                                                                                                                                                                                                              The membrane current induced by heat in a voltage-clamped nociceptor is shown in Fig. 2 A. The current is activated rapidly (but not
                                                                                                                                                                                                                                         instantaneously) by heat, with a mean time to half activation of 35 ms at 50C ( 6 , 7 ). By contrast, neurons insensitive to heat show only
                                                                                                                                                                                                                                         a small current change, which occurs as rapidly as the solution change and therefore probably has a simple physical origin such as a
                                                                                                                                                                                                                                         temperature dependence of membrane leakage resistance. When the temperature dependence of the heat-sensitive current is examined (
                                                                                                                                                                                                                                         Fig. 2 B), the current can be seen to be activated above 42C and to increase exponentially as the temperature is raised further, much as is
                                                                                                                                                                                                                                         observed in nociceptors in vivo ( 1 , 2 ).
                                                                                                                                                                                                                                              Experiments examining the ionic selectivity of the heat-activated current ( 6 , 7 ) have shown that the heat-activated channel
                                                                                                                                                                                                                                         discriminates poorly amongst monovalent alkali cations, in common with many other ion channels such as those gated by glutamate,
                                                                                                                                                                                                                                         acetylcholine, or cyclic nucleotides. Calcium ions can by themselves carry current through the channel but, in addition, have the effect of
                                                                                                                                                                                                                                         partially blocking a current carried by monovalent ions. The channel must therefore possess a binding site in the pore region with a higher
                                                                                                                                                                                                                                         affinity for Ca2+ than for monovalent cations. Contrary to early reports ( 10 ), the channel does not seem to be blocked by Cs+ ions. The
                                                                                                                                                                                                                                         current-voltage relation shows outward rectification and a reversal potential of around 0 mV under physiological conditions ( 6 , 10 , 11 ).
                                                                                                                                                                                                                                           FIG. 2. Responses of membrane current in isolated nociceptors to heat. (A) Application of a rapid step change in temperature (from
                                                                                                                                                                                                                                           room temperature to 49C; time course shown by the top trace) elicits an inward current with a short delay (35 ms) in a nociceptive
                                                                                                                                                                                                                                           neuron (lower of the two membrane current traces). In heat-insensitive neurons, a much smaller current change is elicited with no
                                                                                                                                                                                                                                           delay (top current trace). Neurons were voltage-clamped by the whole-cell patch-clamp method at 70 mV. (B). Current as a function
the authoritative version for attribution.
                                                                                                                                                                                                                                           of temperature in a heat-sensitive neuron (lower trace) and in a heat-insensitive neuron (upper trace). Modified from ref. 6.
                                                                                                                                                                                                                                              Single heat-activated channels have a conductance of around 3040 pS ( 10 , 11 ). The single channel conductance itself is only
                                                                                                                                                                                                                                         weakly temperature-dependent, in common with other ion channels, and the pronounced dependence of current on temperature is caused
                                                                                                                                                                                                                                         by a strong temperature dependence of the probability of channel opening. The time constants of channel opening can be deduced from the
                                                                                                                                                                                                                                         characteristics of the current noise produced when several channels are present simultaneously in a cell-attached membrane patch (see
                                                                                                                                                                                                                                         Fig. 3 ). Application of heat activates inward current and current noise in heat-sensitive but not in heat-insensitive neurons ( Fig. 3 A). The
                                                                                                                                                                                                                                         power spectrum of heat-induced noise ( Fig. 3 B) indicates the existence of two Lorentzian components with time constants 1 = 17 ms and
                                                                                                                                                                                                                                         2 = 0.49 ms. A simple two-state model reproducing these features is
                                                                                                                                                                                                                                              C1  C2  O,
                                                                                                                                                                                                                                                   1    2
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         where 1 is the time constant of transition from the closed state C1 to C2 and 2 is the time constant of transition from the closed state C2 to
                                                                                                                                                                                                                                         the open state (O). The similarity of 1 to the half-time of channel opening after application of a heat jump ( Fig. 2 A) suggests that it is the
                                                                                                                                                                                                                                         transition between C1 and C2 that is the main temperature-dependent event.
                                                                                                                                                                                                                                           FIG. 3. Noise associated with opening of heat-activated ion channels. (A) Examples of cell-attached patch-clamp recordings of the
                                                                                                                                                                                                                                           responses of a heat-insensitive neuron (top recording) and a heat-sensitive neuron (bottom recording) to temperature steps. Patch
                                                                                                                                                                                                                                           pipette contained only 154 mM NaCl/10 mM Hepes to maximize current through heat-sensitive ion channels and was held at 0 mV.
                                                                                                                                                                                                                                           Heat-sensitive currents through channels outside the patch were prevented by bathing the rest of the cell in solution free of permeant
                                                                                                                                                                                                                                           ions (154 mM N-methyl glucamine/10 mM Hepes). (B) Power spectrum of cell-attached current activated by a temperature step to 49
                                                                                                                                                                                                                                           C. For each experiment, 10 consecutive traces (16,384 samples per trace at 10 kHz; filtered at 4 kHz) were acquired, first at room
                                                                                                                                                                                                                                           temperature then at 49C, and the power spectra were calculated. Heat-induced power spectrum (points) was calculated as the
                                                                                                                                                                                                                                           difference between the two spectra. Power spectrum fitted with the sum of two Lorentzian functions (sum shown as a solid line;
                                                                                                                                                                                                                                           component spectra as a dashed line) with half-power frequencies as shown, corresponding to time constants of 17 ms and 0.49 ms
                                                                                                                                                                                                                                           (V.V., P.C., and P.A.M., unpublished data).
                                                                                                                                                                                                                                              Electrophysiologists who work on cultured sensory neurons tend to think of their cells as a bimodal population, consisting of
                                                                                                                                                                                                                                         nociceptive and nonnociceptive neurons. Whole-animal physiologists who work on nociceptors in situ know differently; nociceptors come
                                                                                                                                                                                                                                         in many different varieties, with properties such as heat, mechanical, and chemical sensitivity present to variable extents in different single-
                                                                                                                                                                                                                                         unit recordings. The main division, of course, is between slowly conducting, unmyelinated nociceptive nerve fibers, which commonly
                                                                                                                                                                                                                                         respond to a wide range of stimuli (polymodal fibers) and more rapidly conducting myelinated nerve fibers, which frequently respond to a
                                                                                                                                                                                                                                         smaller subset of noxious stimuli, but amongst which heat-sensitive units are also commonly encountered ( 12 , 13 ). A corresponding
                                                                                                                                                                                                                                         division of nerve-cell bodies is seen, both in dorsal root ganglia and in cultured preparations, into small-diameter dark neurons and large-
                                                                                                                                                                                                                                         diameter pale neurons ( 14 ). Heat sensitivity is, perhaps reassuringly, seen in both cell types in culture ( 10 , 15 ), but there is a
                                                                                                                                                                                                                                         quantitative difference: the smaller cells have a threshold of around 45C, whereas larger cells form a different population with a threshold
                                                                                                                                                                                                                                         of 51C ( 15 ). Interestingly, only the former population responds to capsaicin, suggesting that there is more than one heat-sensitive
                                                                                                                                                                                                                                         channel at work in this diverse population of nociceptors (see further discussion below).
                                                                                                                                                                                                                                              Thermosensation in C. elegans. The nematode worm C. elegans is capable of seeking out a preferred temperature at which to feed,
                                                                                                                                                                                                                                         and mutants unable to detect temperature can therefore be selected by isolating individuals that stray from preferred-temperature areas.
                                                                                                                                                                                                                                         These worms have a mutation either in a gene, tax-4, that codes for the -subunit of an ion channel gated by cyclic nucleotides ( 16 ) or in
                                                                                                                                                                                                                                         a second gene, tax-2, that codes for a -subunit ( 17 ). The fact that these channels can be gated by cyclic nucleotides suggests (but does
                                                                                                                                                                                                                                         not prove) that the mechanism of thermosensation is the modulation of the pathway that controls the level of cyclic nucleotides, rather than
                                                                                                                                                                                                                                         the direct action of heat on the ion channel itself. In this respect, detection of nonnoxious temperatures in C. elegans is different from
                                                                                                                                                                                                                                         mammalian noxious heat sensation. The latter depends only on expression of a heat-sensitive ion channel, which can be seen to function in
                                                                                                                                                                                                                                         isolated membrane patches and therefore is not gated by diffusible messengers controlled by intracellular signaling pathways ( 11 , 18 ,
                                                                                                                                                                                                                                         19 ), whereas thermosensation in C. elegans seems to depend on cyclic nucleotides as intracellular messengers, and the thermosensitive
                                                                                                                                                                                                                                         element is therefore presumably some stage in the pathway modulating the level of cyclic nucleotides. There are many forms of
                                                                                                                                                                                                                                         mammalian thermosensation, as outlined in the introduction, and it is quite possible that signaling pathways are involved in some of these,
                                                                                                                                                                                                                                         even though they do not seem to be directly responsible for heat sensation in the nociceptors of higher vertebrates.
                                                                                                                                                                                                                                              Sensitization of Nociceptors. The process of sensitization (or hyperalgesia) is familiar to us all: a stimulus strong enough to cause
                                                                                                                                                                                                                                         tissue damage hurts more with time, and even after the stimulus has been removed, the damaged area is hypersensitive to touch and to
                                                                                                                                                                                                                                         temperature. This phenomenon can be attributed partly to changes in pain transmission in the spinal cord or at higher levels, but an
                                                                                                                                                                                                                                         important component results from processes occurring at the site of injury. A large number of molecules released by tissue damage are
                                                                                                                                                                                                                                         known to act as mediators of hyperalgesia. Examples include neuropeptides, prostaglandins, histamine, platelet-activating factor, and
                                                                                                                                                                                                                                         bradykinin ( 9 , 20 ).
the authoritative version for attribution.
                                                                                                                                                                                                                                              With so many different factors able to cause hyperalgesia, it is perhaps no surprise that more than one cellular mechanism is involved.
                                                                                                                                                                                                                                         One recently elucidated mechanism involves activation of protein kinase A. External inflammatory messengers such as prostaglandins,
                                                                                                                                                                                                                                         serotonin, and adenosine activate adenylate cyclase and consequently increase the level of cAMP, leading to activation of protein kinase A
                                                                                                                                                                                                                                         ( 21 , 22 ). The principal physiologically important target of protein kinase A seems to be a recently identified voltage-sensitive Na
                                                                                                                                                                                                                                         channel ( 23 ), which, unlike the more usual neuronal Na channel, is not blocked by tetrodotoxin. The effect of phosphorylation of the
                                                                                                                                                                                                                                         tetrodotoxin-resistant Na channel is to lower its threshold, thereby making it more likely that an action potential will be elicited ( 21 , 22 ).
                                                                                                                                                                                                                                         This membrane ionic current is probably not the only one modulated by cAMP, as actions on a K+ current and on a voltage and cyclic
                                                                                                                                                                                                                                         nucleotide-gated conductance have also been identified ( 24 , 25 ). All of these cAMP-dependent mechanisms, however, operate in the
                                                                                                                                                                                                                                         same direction, in that they sensitize the nociceptive nerve terminal to any stimulus that is capable of exciting it, because the effect is to
                                                                                                                                                                                                                                         reduce the
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         threshold for action potential firing, rather than on the specific receptor current induced by the stimulus.
                                                                                                                                                                                                                                               A second and more specific mechanism uses activation of protein kinase C (PKC) to sensitize the response to heat. Fig. 4 A shows
                                                                                                                                                                                                                                         that the inflammatory mediator bradykinin potently increases the membrane current activated by a heat pulse ( 6 ). Bradykinin is known to
                                                                                                                                                                                                                                         activate both phospholipase C and phospholipase A2, thereby releasing a number of potential intracellular signaling molecules. The
                                                                                                                                                                                                                                         enhancement of the heat response can be shown to be due to activation of PKC, however, and not to other possible intracellular mediators,
                                                                                                                                                                                                                                         because it can be mimicked by phorbol esters, which are specific activators of PKC ( Fig. 4 B). Fig. 4 C shows that activation of PKC
                                                                                                                                                                                                                                         increases the current activated by a heat stimulus and shifts the relation between temperature and membrane current to lower temperatures.
                                                                                                                                                                                                                                         These observations predict that normally innocuous temperatures, such as body warmth, will therefore become painful after sensitization,
                                                                                                                                                                                                                                         an observation that corresponds well both with experiments on intact preparations and with our personal experience that even the warmth
                                                                                                                                                                                                                                         of a hand can cause a sensation of pain when applied to an injured area of the body. Other evidence supporting the identity of PKC as an
                                                                                                                                                                                                                                         intracellular mediator of sensitization includes the findings that sensitization can be reversed by PKC inhibitors and can be prolonged by
                                                                                                                                                                                                                                         phosphatase inhibitors, which prevent dephosphorylation after a protein target has been phosphorylated by PKC ( 6 ). The possibility that
                                                                                                                                                                                                                                         mediators other than bradykinin may also employ the PKC pathway to induce sensitization has not yet been investigated and certainly
                                                                                                                                                                                                                                         deserves to be.
                                                                                                                                                                                                                                           FIG. 4. Phosphorylation by PKC sensitizes the heat response of nociceptors. (A) Response of membrane current to a 49C heat pulse
                                                                                                                                                                                                                                           before and after exposure to bradykinin (Bk). (B) Similar effect to that seen in A is observed after treatment by the specific PKC
                                                                                                                                                                                                                                           activator phorbol myristate acetate (PMA). (C) Current vs. temperature relations before and after PMA treatment, showing
                                                                                                                                                                                                                                           sensitization of the heat response. [Reproduced with permission from ref. 6 (Copyright 1996, Proceedings of the National Academy
                                                                                                                                                                                                                                           of Sciences of the United States of America)].
                                                                                                                                                                                                                                              Desensitization of Nociceptors. When a long pulse of moderate heat, insufficiently strong to cause cell damage and to release the
                                                                                                                                                                                                                                         extracellular mediators responsible for sensitization, is applied to a heat-sensitive nociceptor in vivo, gradual adaptation or desensitization
                                                                                                                                                                                                                                         in the firing frequency is observed ( 26 ). A similar phenomenon is seen in isolated nociceptors ( Fig. 5 ), showing that desensitization,
                                                                                                                                                                                                                                         unlike sensitization, is intrinsic to the nociceptor. Recent experiments in our lab have shown that desensitization is triggered by an influx
                                                                                                                                                                                                                                         of calcium ions from the external medium through the heat-sensitive ion channel. In this respect, desensitization of the heat response
                                                                                                                                                                                                                                         resembles the desensitization in response to prolonged application of capsaicin ( 27 ), which is triggered by activation of the calcium-
                                                                                                                                                                                                                                         dependent phosphatase calcineurin by an influx of calcium through the capsaicin-gated channel itself. These observations suggest that both
                                                                                                                                                                                                                                         the heat-activated channel and the capsaicin-gated channel are desensitized when dephosphorylated by calcineurin, one of many
                                                                                                                                                                                                                                         similarities between the two (see below). The molecular mechanisms of sensitization of the heat-activated channel (phosphorylation by
                                                                                                                                                                                                                                         PKC; see above) and desensitization (dephosphorylation by calcineurin) may therefore be simply complementary aspects of the same
                                                                                                                                                                                                                                         process, in which the heat sensitivity of the channel is regulated by phosphorylation (see Fig. 5 and discussion below).
                                                                                                                                                                                                                                              The Capsaicin Receptor, Vanilloid Receptor Subtype 1 (VR1), and Its Relation to Heat Sensation. Capsaicin, the active
                                                                                                                                                                                                                                         ingredient of chili peppers, has been known for some time to depolarize nociceptive nerve terminals by a direct action on an ion channel (
                                                                                                                                                                                                                                         28 ). Capsaicin is not part of the normal environment of most animals. Therefore, it had always been supposed that the capsaicin receptor
                                                                                                                                                                                                                                         was gated physiologically by an endogenous agonist, just as the morphine-receptor family is activated physiologically not by morphine but
                                                                                                                                                                                                                                         by endogenous opiates. Capsaicin-responding neurons can be activated by low pH, and, as pH can drop considerably during inflammation,
                                                                                                                                                                                                                                         hydrogen ions were a plausible candi
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 5. The heat-sensitive current in a nociceptor undergoes desensitization in response to a maintained pulse of heat (P.C., A.M.,
                                                                                                                                                                                                                                           V.V., and P.A.M., unpublished data).
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                           FIG. 6. Possible model for sensitization of the heat response. Acidification of the external solution causes protonation of an external
                                                                                                                                                                                                                                           site on the heat-sensitive ion channel and consequent sensitization ( 19 ). Phosphorylation at the internal surface has a similar effect (
                                                                                                                                                                                                                                           6 ). Both effects are reversible. PP, phosphatase.
                                                                                                                                                                                                                                              The process of sensitization, which shifts the relation between temperature and channel opening to lower temperatures (see Fig. 3 C),
                                                                                                                                                                                                                                         must act by stabilizing the more disordered, higher-temperature state of the channel in such a way that lower temperatures are needed to
                                                                                                                                                                                                                                         induce channel opening. How might this interesting and physiologically important process be operating? One possibility is that opposite
                                                                                                                                                                                                                                         changes in the charge on either side of the membrane may be important. The work of Tominaga et al. ( 19 ) has shown that protonation of
                                                                                                                                                                                                                                         an external site of VR1 induces sensitization, and work in our own lab has shown that phosphorylation of an internal site of the heat-
                                                                                                                                                                                                                                         sensitive receptor induces an apparently identical sensitized state ( 6 ). If VR1 and the heat-sensitive receptors are one and the same, as is
                                                                                                                                                                                                                                         suggested by most lines of evidence (see above), then we can put these two observations together in a simple (and speculative) model of
                                                                                                                                                                                                                                         the sensitization process ( Fig. 6 ). In this model, addition of positive charge to the external face of the membrane or addition of negative
                                                                                                                                                                                                                                         charge to the internal face have equivalent effects, with both manipulations leading to stabilization of a disordered state of the protein and
                                                                                                                                                                                                                                         consequently to sensitization of the response to heat.
                                                                                                                                                                                                                                         1. Treede, R. D. , Meyer, R. A. , Raja, S. N. & Campbell, J. N. ( 1992 ) Prog. Neurobiol. 38 , 397421 .
                                                                                                                                                                                                                                         2. Belmonte, C. & Gallar, J. ( 1996 ) in Neurobiology of Nociceptors , eds. Belmonte, C. & Cervero, F. ( Oxford Univ. Press , Oxford ), Vol. 6 , pp. 146
                                                                                                                                                                                                                                              183 .
the authoritative version for attribution.
                                                                                                                                                                                                                                         3. Rang, H. P. , Bevan, S. & Dray, A. ( 1994 ) Textbook of Pain , eds. Melzack, R. & Wall, P. ( Churchill Livingstone , Edinburgh ), Vol. 3 , pp. 5778 .
                                                                                                                                                                                                                                         4. Baccaglini, P. I. & Hogan, P. G. ( 1983 ) Proc. Natl. Acad. Sci. USA 80 , 594598 .
                                                                                                                                                                                                                                         5. Gilabert, R. & McNaughton, P. A. ( 1997 ) J. Neurosci. Methods 71 , 191198 .
                                                                                                                                                                                                                                         6. Cesare, P. & McNaughton, P. A ( 1996 ) Proc. Natl. Acad. Sci. USA 93 , 1543515439 .
                                                                                                                                                                                                                                         7. Cesare, P. & McNaughton, P. A. ( 1997 ) Curr. Opin. Neurobiol. 7 , 493499 .
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                               EDWARD R. PERL*
                                                                                                                                                                                                                                               Department of Cell and Molecular Physiology, CB 7545, University of North Carolina, Chapel Hill, NC 27599
                                                                                                                                                                                                                                               ABSTRACT Control of expression of molecular receptors for chemical messengers and modulation of these receptors
                                                                                                                                                                                                                                         activity are now established as ways to alter cellular reaction. This paper extends these mechanisms to the arena of pathological
                                                                                                                                                                                                                                         pain by presenting the hypothesis that increased expression of -adrenergic receptors in primary afferent neurons is part of the
                                                                                                                                                                                                                                         etiology of pain in classical causalgia. It is argued that partial denervation by lesion of peripheral nerve or by tissue destruction
                                                                                                                                                                                                                                         induces a change in peripheral nociceptors, making them excitable by sympathetic activity and adrenergic substances. This
                                                                                                                                                                                                                                         excitation is mediated by -adrenergic receptors and has a time course reminiscent of experimental denervation supersensitivity.
                                                                                                                                                                                                                                         The change in neuronal phenotype is demonstrable after lesions of mixed nerves or of the sympathetic postganglionic supply.
                                                                                                                                                                                                                                         Similar partial denervations also produce a substantial increase in the number of dorsal root ganglion neurons evidencing the
                                                                                                                                                                                                                                         presence of -adrenergic receptors. The hypothesis proposes the increased presence of -adrenergic receptors in primary afferent
                                                                                                                                                                                                                                         neurons to result from an altered gene expression triggered by cytokines/growth factors produced by disconnection of peripheral
                                                                                                                                                                                                                                         nerve fibers from their cell bodies. These additional adrenergic receptors are suggested to make nociceptors and other primary
                                                                                                                                                                                                                                         afferent neurons excitable by local or circulating norepinephrine and epinephrine. For central pathways, the adrenergic excitation
                                                                                                                                                                                                                                         would be equivalent to that produced by noxious events and would consequently evoke pain. In support, evidence is cited for a
                                                                                                                                                                                                                                         form of denervation supersensitivity in causalgia and for increased expression of human -adrenergic receptors after loss of
                                                                                                                                                                                                                                         sympathetic activity.
                                                                                                                                                                                                                                               This essay is an outgrowth of a Colloquium session in which new evidence was presented on how molecular receptors for chemical
                                                                                                                                                                                                                                         synaptic mediators can specify and regulate neuronal responses in systems associated with pain mechanisms. These data build on the
                                                                                                                                                                                                                                         concept that not only the nature, but also the magnitude, of the transfer of information between cells is at least as much a function of
                                                                                                                                                                                                                                         receptive molecules as of the chemical messengers. Therefore, selective regulation of receptor expression and quantitative control of
                                                                                                                                                                                                                                         receptor activity are factors defining or modulating synaptic function. Importantly, such concepts, when applied to disease, open novel
                                                                                                                                                                                                                                         possibilities of pharmaceutical manipulation and treatment. My purpose is to extend such considerations of receptor regulation to a
                                                                                                                                                                                                                                         pathological process involving pain.
                                                                                                                                                                                                                                               There now is considerable agreement that in mammals, the detection and signaling of tissue damage or pathology, that is,
                                                                                                                                                                                                                                         nociception, is a normal somatosensory function. In this view, pain, one of the reactions to tissue injury, represents the sensory
                                                                                                                                                                                                                                         concomitant of nociception. By logical extension, pain in the absence of peripheral tissue damage is abnormal, in other words, pathological.
                                                                                                                                                                                                                                               Causalgia and Postsympathectomy Pain. S. Weir Mitchell ( 1 ) named a syndrome causalgia after its most prominent symptom, a
                                                                                                                                                                                                                                         burning pain referred to a particular body region appearing spontaneously or after innocuous stimulation. As classically described,
                                                                                                                                                                                                                                         causalgia appears after partial disruption of the innervation to a limb, typically after injury to a large mixed nerve. It probably is relevant
                                                                                                                                                                                                                                         that the full-blown syndrome is not usually reported after lesions of smaller, purely cutaneous nerves. Some years after the original
                                                                                                                                                                                                                                         descriptions, Rene Leriche ( 2 ) pointed out that the syndrome of causalgia had features suggesting abnormal sympathetic nervous system
                                                                                                                                                                                                                                         functioning and proposed sympathectomy as a treatment. Subsequently, the list of disorders in which pain was presumably related to
                                                                                                                                                                                                                                         sympathetic nervous activity expanded beyond the original descriptions of classical causalgia and acquired other terminologies. It is not
                                                                                                                                                                                                                                         clear that all of these later additions to the category of sympathetically related pain disorders share a common etiology and pathology to
                                                                                                                                                                                                                                         the classical causalgic syndrome. For the purpose of focusing our consideration on a disorder with a common causative process, the
                                                                                                                                                                                                                                         following starts from classical causalgia without implying extension to either more general or to more specific terminologies and
                                                                                                                                                                                                                                         classifications: e.g., reflex sympathetic dystrophy ( 3 ), complex regional pain syndrome ( 4 ), and various others ( 5 ).
                                                                                                                                                                                                                                               As outlined in Fig. 1 , the classical syndrome of causalgia includes the following features. (i) It follows partial denervation of a
                                                                                                                                                                                                                                         region, usually by traumatic injury of a large mixed nerve. (ii) The partially denervated area is hypalgesic. (iii) Days to weeks after the
                                                                                                                                                                                                                                         disturbance of innervation, spontaneous pain appears, typically burning in nature, referred to the partially denervated and nearby regions.
                                                                                                                                                                                                                                         (iv) Pain is produced or increased by normally nonpainful stimuli, e.g., skin cooling or light touch (allodynia). (v) Abnormal sympathetic
                                                                                                                                                                                                                                         function is evident in the region (e.g., vasomotion, perspiration). (vi) The pain is aggravated by emotional upset. (vii) Trophic changes
                                                                                                                                                                                                                                         appear in the partially denervated tissues and nearby regions including abnormal coloration and turgor of the skin, unusual growth of hair,
                                                                                                                                                                                                                                         and changes in bone and other subcutaneous tissues. Certain of the physical signs are suggestive of chronic inflammation.
                                                                                                                                                                                                                                               After Leriches suggestion, regional sympathectomy or regional sympathetic block has been used as a therapy for this syndrome with,
                                                                                                                                                                                                                                         in many cases, at least temporary success ( 6 , 7 ). In cases with successful outcome, the abnormal pain is reduced or abolished, and there
                                                                                                                                                                                                                                         is amelioration of trophic changes ( 2 , 6 , 7 ). During remission of the signs and symptoms after sympathectomy or sympathetic block,
                                                                                                                                                                                                                                         local injection of norepinephrine into the skin of the previously painful region has been reported to recreate the former causalgic pain (ref.
                                                                                                                                                                                                                                         8 ; see also ref. 9 ). These observations suggest that sympathetic
the authoritative version for attribution.
                                                                                                                                                                                                                                         activity and adrenergic mediators have a part in the aberrant pain and other features of causalgia.
                                                                                                                                                                                                                                             Classical Causalgia
                                                                                                                                                                                                                                             Partial injury of a mixed nerve or of peripheral innervation
                                                                                                                                                                                                                                         immunoreactivity
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         to the 2A-adrenergic receptor antibody does not appear after localized artificial inflammation produced by injection of formalin or
                                                                                                                                                                                                                                         Freunds complete adjuvant, and immunoreactivity to a 2C antibody is not increased after sciatic nerve injury ( 29 ). An earlier
                                                                                                                                                                                                                                         autoradiographic study with the partial 2 agonist, p-iodoclonidine (125I-labeled) had indicated increased binding in ipsilateral DRG after
                                                                                                                                                                                                                                         partial or complete nerve transection; however, the diameter spectrum of the p-iodoclonidine-labeled DRG neurons (mainly small-
                                                                                                                                                                                                                                         diameter) partially differs from that with the 2A-adrenergic receptor antibody (refs. 10 and 29 ; K. Nishiyama and E.R.P., unpublished
                                                                                                                                                                                                                                         data).
                                                                                                                                                                                                                                             Linking -Adrenergic Receptors to Pain in Causalgia
                                                                                                                                                                                                                                            Partial interruption of primary afferent and sympathetic innervation of a region
                                                                                                                                                                                                                                            Reaction to injury by neurons and associated cells: production of cytokines for growth/regeneration
                                                                                                                                                                                                                                            Activation of gene(s) producing -ARs in injured DRG neurons and in remaining intact nociceptors
                                                                                                                                                                                                                                            Activation of -ARs in nociceptors by local and circulating adrenergic agents
                                                                                                                                                                                                                                            Impulse traffic in peripheral and central pathways for nociception and pain
                                                                                                                                                                                                                                            Peripheral and central changes leading to allodynia
                                                                                                                                                                                                                                            Secondary changes in sensory and affective mechanisms from persisting pain-related input
                                                                                                                                                                                                                                             FIG. 3. Factors in the hypothesis connecting increased expression of -adrenergic receptors to the pain of causalgia.
                                                                                                                                                                                                                                               An Hypothesis. The effects of experimental nerve injury on the responsiveness of cutaneous nociceptors to adrenergic substances
                                                                                                                                                                                                                                         and on -adrenergic receptor expression in dorsal root ganglia suggest a possible relationship to the etiology and symptoms of causalgia.
                                                                                                                                                                                                                                         The most salient factors are outlined in Fig. 2 . This leads to the hypothesis summarized by Fig. 3 . It is proposed that injury to part of the
                                                                                                                                                                                                                                         innervation to a bodily region, e.g., partial transection of a mixed nerve supplying part of a limb, induces production of cytokines and/or
                                                                                                                                                                                                                                         growth factors by the injured neurons and associated cells (e.g., Schwann cells). These chemical factors, among other effects, mediate
                                                                                                                                                                                                                                         responses to injury or are associated with regrowth and lead to altered gene expression in uninjured neurons of the nerve and region.
                                                                                                                                                                                                                                         Disconnection of sympathetic postganglionic fibers from their targets, by itself and in conjunction with injury of primary afferent fibers,
                                                                                                                                                                                                                                         contributes to these signals, leading to altered expression of -adrenergic receptors. The fact that classical causalgia usually follows injury
                                                                                                                                                                                                                                         to large mixed nerves raises the possibility that the required lesion for the full syndrome is the combined interruption of primary afferent
                                                                                                                                                                                                                                         fibers and sympathetic postganglionic fibers. Possibly the loss of the presence of sympathetic mediators in the vicinity of primary afferent
                                                                                                                                                                                                                                         terminals because of interruption of sympathetic postganglionic fibers is part of the pathophysiological process. The net result after such
                                                                                                                                                                                                                                         partial loss of innervation is that afferent neurons, normally expressing few -adrenergic receptors capable of producing an excitatory
                                                                                                                                                                                                                                         response, develop them. As a result, sensory neurons, particularly nociceptors, become excitable through these newly formed receptors.
                                                                                                                                                                                                                                         The afferent neuron excitation would occur by norepinephrine locally released by the remaining sympathetic supply to the vasculature or
                                                                                                                                                                                                                                         norepinephrine and epinephrine circulating from other parts of the body. For central mechanisms, signals produced in nociceptors by
                                                                                                                                                                                                                                         adrenergic agents are equivalent to those evoked by noxious events and lead to the sensation of pain. Signals interpreted as the result of
                                                                                                                                                                                                                                         noxious stimuli can result from activation by -adrenergic receptors in DRG neurons by trace quantities of norepinephrine and
                                                                                                                                                                                                                                         epinephrine. The outcome is spontaneous pain or sensitization of nociceptors.
                                                                                                                                                                                                                                               Observations on human subjects offer circumstantial support for facets of this concept. Loss or decrease of sympathetic activity and
                                                                                                                                                                                                                                         the consequent decrease in circulating sympathetic postganglionic mediators has been shown to increase expression of -adrenergic
                                                                                                                                                                                                                                         receptors ( 27 , 28 , 33 ). Those observations were made on blood platelets. By inference, one can argue that the same process could affect
                                                                                                                                                                                                                                         DRG neurons. Regional sympathectomy in experimental animals does increase binding of -adrenergic agonists in DRG neurons of the
                                                                                                                                                                                                                                         innervated region (ref. 10 and K. Nishiyama and E.R.P., unpublished data). Thus, one manipulation that leads to induction of adrenergic
                                                                                                                                                                                                                                         excitation of nociceptors, loss of sympathetic innervation, increases -adrenergic receptor expression in some tissues.
                                                                                                                                                                                                                                               It is pertinent that the affected limb in persons suffering from pain disorders fitting the criteria of classical causalgia exhibits lower
                                                                                                                                                                                                                                         concentrations of norepinephrine and a degradation product (3,4-dihydroxphenylethyleneglycol) in its venous return than contralaterally.
                                                                                                                                                                                                                                         This could imply that the affected limb has less functioning sympathetic innervation. At the same time, the affected limb exhibits
                                                                                                                                                                                                                                         increased activity by sympathetically innervated tissues ( 34 , 35 ). These human data suggest that a process akin to denervation
                                                                                                                                                                                                                                         supersensitivity may operate in classical causalgia and possibly other varieties of sympathetically related pain disorders.
                                                                                                                                                                                                                                               To repeat, my suggestion here is that primary afferent neuron excitation by adrenergic agents in classical causalgia results from novel
                                                                                                                                                                                                                                         -adrenergic receptor production in dorsal root ganglia neurons evoked by direct and indirect effects of injury to peripheral innervation.
                                                                                                                                                                                                                                         The increased adrenergic receptor expression, in part, involves primary afferent neurons, particularly nociceptors, with intact connections
                                                                                                                                                                                                                                         to the periphery and the central nervous system. As a consequence of the novel -adrenergic receptor production, some of these afferent
                                                                                                                                                                                                                                         neurons develop an excitatory response to trace amounts of adrenergic substances in peripheral tissues. Such excitation would be the start
                                                                                                                                                                                                                                         of abnormal signals activating pain pathways and central pain-related mechanisms.
                                                                                                                                                                                                                                               The concept just outlined, like most hypotheses, has difficulties. First, it cannot explain all parts of a complex syndrome. In
                                                                                                                                                                                                                                         particular, it does not account for the trophic changes, allodynia, and psychological alterations. The hypothesis suggests only that nerve
                                                                                                                                                                                                                                         injury and partial denervation unleash a set of circumstances leading to an abnormal production of -adrenergic receptors in sensory
                                                                                                                                                                                                                                         neurons. Experimental studies suggest that the adrenergic receptor type may be of the 2 (possibly also 1) type ( 28 , 29 , 23 ). These
                                                                                                                                                                                                                                         adrenergic receptors become part of a messenger system whereby adrenergic substances excite or sensitize peripheral sensory neurons
                                                                                                                                                                                                                                         related to nociception and pain, which represents a step in the process leading to spontaneous pain and to activation of central pathways.
                                                                                                                                                                                                                                         Subsequently, the abnormally initiated central activity can lead to sensitization and other plastic changes in central neuronal mechanisms.
                                                                                                                                                                                                                                         Although not an explanation of all signs and symptoms of causalgia, this proposal provides a possible etiology of the pathological process
                                                                                                                                                                                                                                         and some insight into factors that could operate to maintain the process. Second, there is the issue of the effects mediated by 2-adrenergic
                                                                                                                                                                                                                                         receptors that usually are presumed in neurons to mediate
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         inhibitory actions. In this context, it should be remembered that 2-adrenergic receptors are intermediate arteriolar smooth muscle
                                                                                                                                                                                                                                         constriction. Therefore, this class of receptors is capable of being part of an excitatory signal-transduction process ( 36 ); furthermore, the
                                                                                                                                                                                                                                         signaling system induced by nerve injury may not be identical to that occurring in neurons normally.
                                                                                                                                                                                                                                              Does the idea of a change in cellular phenotype by the enhanced production of membrane receptors possibly apply to other
                                                                                                                                                                                                                                         situations? A similar process could operate in other versions of sympathetically related pain. It could also relate to Raynauds disease,
                                                                                                                                                                                                                                         another pathological process which, in part, appears to represent overreaction to sympathetic mediators and could possibly result from an
                                                                                                                                                                                                                                         increased expression of adrenergic receptors ( 37 ). Furthermore, enhanced reactions to adrenergic mediators by the vasculature have also
                                                                                                                                                                                                                                         been postulated for certain forms of hypertension ( 38 , 39 ). To conclude, the concept of increased expression of molecular receptors as a
                                                                                                                                                                                                                                         mechanism of disease, and in particular of pathological pain, deserves serious consideration and further exploration.
                                                                                                                                                                                                                                              I thank Ms. S. Derr for her assistance. Preparation of this paper was aided by grants NS 10321 and NS 14899 of the National Institute
                                                                                                                                                                                                                                         of Neurological Disorders and Stroke.
                                                                                                                                                                                                                                         1. Mitchell, W. ( 1872 ) Injuries of Nerves and Their Consequences ( Lippincott , Philadelphia ).
                                                                                                                                                                                                                                         2. Leriche, R. ( 1916 ) Presse Md. 24 , 178180 .
                                                                                                                                                                                                                                         3. Evans, J. A. ( 1946 ) Surg. Clin. North Am. 26 , 435448 .
                                                                                                                                                                                                                                         4. Merskey, H. & Bogduk, N. ( 1995 ) in Classification of Chronic Pain: Descriptions of Chronic Pain Syndromes and Definition of Terms ( IASP
                                                                                                                                                                                                                                              Press , Seattle ).
                                                                                                                                                                                                                                         5. Kozin, F. , McCarty, D. J. , Sims, J. & Genant, H. ( 1976 ) Am. J. Med. 60 , 321331 .
                                                                                                                                                                                                                                         6. Schumacker, H. B. , Speigel, I. J. & Upjohn, R. H. ( 1948 ) Surg. Gynecol. Obstet. 86 , 7686 .
                                                                                                                                                                                                                                         7. Richards, R. L. ( 1967 ) Arch. Neurol. 16 , 339350 .
                                                                                                                                                                                                                                         8. Wallin, E. , Torebjrk, E. & Hallin, R. ( 1976 ) in Sensory Functions of the Skin in Primates, with Special Reference to Man , ed. Zotterman, Y. (
                                                                                                                                                                                                                                              Pergamon , Oxford ), pp. 489502 .
                                                                                                                                                                                                                                         9. Torebjrk, E. , Wahren, L. , Wallin, G. , Hallin, R. & Koltzenburg, M. ( 1995 ) Pain 63 , 1120 .
                                                                                                                                                                                                                                         10. Perl, E. R. ( 1994 ) in Progress in Pain Research and Management , eds. Fields, H. L. & Liebeskind, J. C. ( IASP Press , Seattle ), pp. 129150 .
                                                                                                                                                                                                                                         11. Perl, E. R. ( 1994 ) in Touch, Temperature, and Pain in Health and Disease: Mechanisms and Assessments, Progress in Pain Research and
                                                                                                                                                                                                                                              Management eds. Boivie, J. , Hansson, P. & Lindblom, U. ( IASP Press , Seattle ), Vol. 3 , pp. 231248 .
                                                                                                                                                                                                                                         12. Tracy, G. D. & Cockett, F. B. ( 1957 ) Lancet i (272) , 1214 .
                                                                                                                                                                                                                                         13. Litwin, M. S. ( 1962 ) Arch. Surg. 84 , 591595 .
                                                                                                                                                                                                                                         14. Raskin, N. H. , Levinson, S. A. , Hoffman, P. M. , Pickett, J. B. E. & Fields, H. L. ( 1974 ) Am. J. Surg. 128 , 7578 .
                                                                                                                                                                                                                                         15. Churcher, M. D. ( 1984 ) Lancet ii (8395) , 131133 .
                                                                                                                                                                                                                                         16. Devor, M. & Jnig, W. ( 1981 ) Neurosci. Lett. 24 , 4347 .
                                                                                                                                                                                                                                         17. Devor, M. ( 1983 ) J. Auton. Nerv. Syst. 7 , 371384 .
                                                                                                                                                                                                                                         18. Shea, V. & Perl, E. R. ( 1985 ) J. Neurophysiol. 54 , 491501 .
                                                                                                                                                                                                                                         19. Barasi, S. & Lynn, B. ( 1986 ) Brain Res. 378 , 2127 .
                                                                                                                                                                                                                                         20. OHalloran, K. D. & Perl, E. R. ( 1997 ) Brain Res. 759 , 233240 .
                                                                                                                                                                                                                                         21. Sato, J. & Perl, E. R. ( 1991 ) Science 251 , 16081610 .
                                                                                                                                                                                                                                         22. Bossut, D. F. & Perl, E. R. ( 1995 ) J. Neurophysiol. 73 , 17211723 .
                                                                                                                                                                                                                                         23. Ali, Z. , Ringkamp, M. , Hartke, T. V. , Chien, H. F. , Flavahan, N. A. , Campbell, J. N. & Meyer, R. A. ( 1999 ) J. Neurophysiol. 81 , 455466 .
                                                                                                                                                                                                                                         24. Bossut, D. F. , Shea, V. & Perl, E. R. ( 1995 ) J. Neurophysiol. 75 , 514517 .
                                                                                                                                                                                                                                         25. Cannon, W. B. & Rosenblueth , ( 1949 ) The Supersensitivity of Denervated Structures: A Law of Denervation ( McMillan , New York ).
                                                                                                                                                                                                                                         26. Arnett, C. O. & J. A. Davis ( 1979 ) J. Pharmacol. Exp. Ther. 211 , 394400 .
                                                                                                                                                                                                                                         27. Davies, I. B. , Sudera, D. , Sagnella, G. , Marchesi-Saviotti, E. , Mathias, C. , Bannister, R. & Sever, P. S. ( 1982 ) J. Clin. Invest. 69 , 779784 .
                                                                                                                                                                                                                                         28. Egan, B. , Neubig, R. & Julius, S. ( 1985 ) Clin. Pharmacol. Ther. 38 , 519524 .
                                                                                                                                                                                                                                         29. Birder, L. A. & Perl, E. R. ( 1999 ) J. Physiol. (London) 515 , 533542 .
                                                                                                                                                                                                                                         30. Jenkins, R. & Hunt, S. P. ( 1991 ) Neurosci. Lett. 129 , 107110 .
                                                                                                                                                                                                                                         31. Jenkins, R. , McMahon, S. B. , Bond, A. B. & Hunt, S. P. ( 1993 ) Eur. J. Neurosci. 5 , 751759 .
                                                                                                                                                                                                                                         32. Baranowski, A. P. , Anand, U. & McMahon, S. B. ( 1992 ) Neurosci. Lett. 141 , 5356 .
                                                                                                                                                                                                                                         33. Davies, I. B. & Sever, P. S. ( 1988 ) in Autonomic Failure: A Textbook of Clinical Disorders of the Autonomic Nervous System , ed. Bannister, R. (
                                                                                                                                                                                                                                              Oxford Univ. Press , Oxford ), pp. 348366 .
                                                                                                                                                                                                                                         34. Drummond, P. D. , Finch, P. M. & Smythe, G. A. ( 1991 ) Brain 114 , 20252036 .
                                                                                                                                                                                                                                         35. Drummond, P. D. , Finch, P. M. , Edvinsson, L. & Goadsby, P. J. ( 1994 ) Clin. Auto. Res. 4 , 113116 .
                                                                                                                                                                                                                                         36. Nichols, A. J. & Ruffolo, R. R. , Jr. ( 1991 ) in Progress in Basic and Clinical Pharmacology , ed. Ruffolo, R. R., Jr. (Karger, Basel), Vol. 8 , pp.
                                                                                                                                                                                                                                              115179 .
                                                                                                                                                                                                                                         37. Edwards, J. M. , Phinney, E. S. , Taylor, L. M., Jr. , Keenan, E. J. & Porter, J. M. ( 1987 ) J. Vasc. Surg. 5 , 3845 .
                                                                                                                                                                                                                                         38. Michel, M. C. , Insel, P. A. & Brodde, O. ( 1989 ) FASEB J. 3 , 139144 .
                                                                                                                                                                                                                                         39. De Champlain, J. ( 1989 ) J. Hypertension 8, Suppl. 7 , S77S85 .
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                              This paper was presented at the National Academy of Sciences colloquium The Neurology of Pain, held December 1113, 1998,
                                                                                                                                                                                                                                         at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                               KENNETH L. CASEY
                                                                                                                                                                                                                                               Neurology Service, Veterans Affairs Medical Center, University of Michigan, 2215 Fuller Road, Ann Arbor, MI 48105
                                                                                                                                                                                                                                               ABSTRACT Pain is a unified experience composed of interacting discriminative, affective-motivational, and cognitive
                                                                                                                                                                                                                                         components, each of which is mediated and modulated through forebrain mechanisms acting at spinal, brainstem, and cerebral
                                                                                                                                                                                                                                         levels. The size of the human forebrain in relation to the spinal cord gives anatomical emphasis to forebrain control over
                                                                                                                                                                                                                                         nociceptive processing. Human forebrain pathology can cause pain without the activation of nociceptors. Functional imaging of
                                                                                                                                                                                                                                         the normal human brain with positron emission tomography (PET) shows synaptically induced increases in regional cerebral
                                                                                                                                                                                                                                         blood flow (rCBF) in several regions specifically during pain. We have examined the variables of gender, type of noxious stimulus,
                                                                                                                                                                                                                                         and the origin of nociceptive input as potential determinants of the pattern and intensity of rCBF responses. The structures most
                                                                                                                                                                                                                                         consistently activated across genders and during contact heat pain, cold pain, cutaneous laser pain or intramuscular pain were the
                                                                                                                                                                                                                                         contralateral insula and anterior cingulate cortex, the bilateral thalamus and premotor cortex, and the cerebellar vermis. These
                                                                                                                                                                                                                                         regions are commonly activated in PET studies of pain conducted by other investigators, and the intensity of the brain rCBF
                                                                                                                                                                                                                                         response correlates parametrically with perceived pain intensity. To complement the human studies, we developed an animal
                                                                                                                                                                                                                                         model for investigating stimulus-induced rCBF responses in the rat. In accord with behavioral measures and the results of human
                                                                                                                                                                                                                                         PET, there is a progressive and selective activation of somatosensory and limbic system structures in the brain and brainstem
                                                                                                                                                                                                                                         following the subcutaneous injection of formalin. The animal model and human PET studies should be mutually reinforcing and
                                                                                                                                                                                                                                         thus facilitate progress in understanding forebrain mechanisms of normal and pathological pain.
                                                                                                                                                                                                                                               Forebrain Mediation of Pain. Pain is a conscious experience that includes discriminative, affective-motivational, and cognitive
                                                                                                                                                                                                                                         components that produce the unified sensation of pain. These components are each mediated through separate, interactive forebrain
                                                                                                                                                                                                                                         mechanisms ( 1 ). For example, the ability to localize somatic stimuli in time, space, and along a continuum of intensities is greatly
                                                                                                                                                                                                                                         impaired following lesions limited to the primary somatosensory (S1) cortex or the ventral posterolateral thalamus. These lesions do not
                                                                                                                                                                                                                                         produce analgesia, however, because the aversive nature of noxious stimuli, although poorly localized, is still evident in the behavior of
                                                                                                                                                                                                                                         animals and the verbal reports of humans ( 2 ). Neurons in the S1 cortex and ventral posterolateral thalamus, including those responding
                                                                                                                                                                                                                                         primarily to noxious stimuli, have small, contralateral receptive fields consistent with the mediation of spatial stimulus localization ( 3 ). In
                                                                                                                                                                                                                                         contrast, lesions within the anterior cingulate cortex have no effect on innocuous or nociceptive somesthetic discriminative functions, but
                                                                                                                                                                                                                                         impair the recognition of the noxious or aversive quality of the stimulus in animals and the perceived affective quality of pain in humans (
                                                                                                                                                                                                                                         4 , 5 ). Anterior cingulate neurons that respond to noxious stimuli have large, often bilateral receptive fields, consistent with a limited role
                                                                                                                                                                                                                                         in spatial discriminative capacity ( 6 ). There is no comparable information about the neuronal substrate for the cognitive dimension of
                                                                                                                                                                                                                                         pain, but there are numerous studies and observations showing the profound influences of attention, suggestion, and emotional state on the
                                                                                                                                                                                                                                         perception of pain ( 7 ). The broad range of environmental influences, such as attention, fear, and the placebo effect on the perception of
                                                                                                                                                                                                                                         pain suggests that cortical association areas and their subcortical connections are critical participants in mediating the cognitive aspects of
                                                                                                                                                                                                                                         pain.
                                                                                                                                                                                                                                               The Forebrain Modulation of Pain. The processing of nociceptive stimuli is modulated by the forebrain at spinal, brainstem, and
                                                                                                                                                                                                                                         diencephalic levels. Stimulation of the cerebral cortex or thalamus can facilitate or suppress the responses of spinothalamic or
                                                                                                                                                                                                                                         trigeminothalamic tract neurons ( 8 , 9 ). In the awake monkey, the response of trigeminothalamic cells to noxious heat depends on
                                                                                                                                                                                                                                         behavioral state ( 10 , 11 ). Corticobulbar and corticothalamic neurons have marked effects on the excitability of brainstem and thalamic
                                                                                                                                                                                                                                         cells that receive nociceptive input ( 12  16 ).
                                                                                                                                                                                                                                               Because of the large volume of the human forebrain in relation to that of the spinal cord (77% vs. 2% of central nervous system
                                                                                                                                                                                                                                         volume), these descending modulatory influences may assume greater importance in humans than in other species, such as the laboratory
                                                                                                                                                                                                                                         rat, where the forebrain is less anatomically dominant (31% vs. 35% of central nervous system volume) ( 17 ). The human spinothalamic
                                                                                                                                                                                                                                         tract, for example, contains an estimated 2,000 to 5,000 fibers whereas the corticospinal tract, which includes fibers terminating in the
                                                                                                                                                                                                                                         superficial layers of the dorsal horn ( 18 , 19 ), is estimated to contain from 5  105 to 1  106 fibers ( 20 , 21 ). Corticothalamic influences
                                                                                                                                                                                                                                         are also likely to be dominant in the human; in the cat, approximately 50% of the estimated 5,000 to 9,000 synapses on thalamocortical
                                                                                                                                                                                                                                         projection neurons are presumed to be of cortical origin, whereas only 15% are formed by ascending afferent fibers ( 22 ).
                                                                                                                                                                                                                                               The Physiological Rationale of Positron Emission Tomography (PET). Synaptic activity generates increases in cerebral blood
                                                                                                                                                                                                                                         flow (CBF). This physiological fact is the basis for both PET and functional magnetic resonance imaging (fMRI). The most commonly
                                                                                                                                                                                                                                         used fMRI method relies on local shifts in the magnetic field that accompany the shift from deoxyhemoglobin to oxyhemoglobin within
                                                                                                                                                                                                                                         activated perfused tissue ( 23 ). PET and fMRI are complementary methods of assessing brain activity. This article will be limited to a
                                                                                                                                                                                                                                         discussion of PET.
the authoritative version for attribution.
                                                                                                                                                                                                                                               The first indication that brain activity increases global CBF was reported by Roy and Sherrington over 100 years ago ( 24 ).
                                                                                                                                                                                                                                         Subsequent radioactive tracer techniques revealed increases in regional CBF (rCBF) during sensory stimulation or the performance of
                                                                                                                                                                                                                                         motor tasks ( 25 ). Recent studies that use the technique of optical imaging have demonstrated that cortical blood flow responses occur
                                                                                                                                                                                                                                         within 3 sec of sensory stimulation and are initially restricted to the 300- to 500-m dimensions of cortical columns before spreading to
                                                                                                                                                                                                                                         involve the surrounding 3 mm to 5 mm of cortical tissue ( 26  28 ). The biochemical coupling of rCBF and synaptic activity is unknown
                                                                                                                                                                                                                                         and is still an area of active investigation. Studies of regional cerebral glucose utilization show that it and rCBF are normally tightly
                                                                                                                                                                                                                                         coupled and that the coupling occurs within the synaptic neuropil. The degree of coupling may vary among regions and under special
                                                                                                                                                                                                                                         experimental conditions ( 29 ), but it is reliably present in the normal mammal. Blocking the production of nitric oxide has no effect on
                                                                                                                                                                                                                                         synaptically induced rCBF responses in the rat somatosensory system ( 30 ). There is evidence that adenosine may be a critical link in this
                                                                                                                                                                                                                                         process, but it is likely that the action of several mediators may be important ( 31 ).
                                                                                                                                                                                                                                               A reduction of rCBF should occur when synaptic activity is suppressed below some resting or background level. Indeed, reductions in
                                                                                                                                                                                                                                         rCBF are observed in many PET studies of rCBF ( 32 ). However, the physiological significance of reduced rCBF is uncertain; it does not
                                                                                                                                                                                                                                         necessarily indicate the presence of inhibitory synaptic activity, because both inhibitory and excitatory synaptic activity can contribute to
                                                                                                                                                                                                                                         increases in synaptic metabolism ( 33 ). It is possible that some of the observed reductions in rCBF reflect autoregulatory mechanisms for
                                                                                                                                                                                                                                         global CBF, these reductions may not affect neuronal function; others may signal the removal of synaptic excitation (disfacilitation) by an
                                                                                                                                                                                                                                         inhibitory process located outside the area of rCBF decrease. In any event, it is not now possible to establish the valence of synaptic
                                                                                                                                                                                                                                         activity by rCBF estimation methods.
                                                                                                                                                                                                                                               PET Methodology. In most current studies, water (as H215O) or [15O]butanol is injected intravenously, or carbon dioxide (as C15O2)
                                                                                                                                                                                                                                         is inhaled and converted in the lungs to H215O. The 15O has a half-life of 122 sec. This length of time is sufficient for CBF measurements,
                                                                                                                                                                                                                                         because a bolus injection (e.g., 50 mCi) of this compound is nearly completely diffused into brain tissue on the first arterial pass ( 34 ).
                                                                                                                                                                                                                                         The count of emissions from a given volume of brain tissue is therefore a good estimate of the perfusion of that brain region during the
                                                                                                                                                                                                                                         counting period (approximately 60 sec for a typical scan).
                                                                                                                                                                                                                                               With the analytical methods that we use, we find that there are approximately 95,000 voxels in the gray matter of the average human
                                                                                                                                                                                                                                         brain. At our facility, a three-dimensional voxel is a cube 2.25 mm on each side. However, the spatial resolution of PET is limited by the
                                                                                                                                                                                                                                         smoothing introduced by image reconstruction filters and by the ability of the radiation detectors to differentiate the radiation emitted from
                                                                                                                                                                                                                                         two separate point sources. For PET, this distance, the full width at half maximum, is between 6 and 9 mm. However, the spatial accuracy
                                                                                                                                                                                                                                         in the localization of an activation focus is improved (to less than half the full width at half maximum) when subtraction images are made.
                                                                                                                                                                                                                                               Each image set is normalized to whole brain counts, and mean radioactivity concentration images are created by estimating rCBF
                                                                                                                                                                                                                                         across all subjects with stereotactic anatomical standardization techniques. Image voxel intensities are normalized to global cerebral
                                                                                                                                                                                                                                         activity with the use of a linear proportional model to remove baseline differences in global CBF between scans and subjects ( 35 ). In our
                                                                                                                                                                                                                                         facility, CBF images are aligned onto the coordinates of a standard stereotactic atlas ( 36 ), by using anatomical landmarks identified
                                                                                                                                                                                                                                         within the PET images of each individual so that the CBF differences are compared within the same brain regions ( 37  39 ). To
                                                                                                                                                                                                                                         determine whether a task or a stimulus has produced an increase in rCBF, the rCBF computed during a control condition is subtracted
                                                                                                                                                                                                                                         from that computed during the test condition. The resulting subtraction image, then, shows those brain regions with differences in CBF
                                                                                                                                                                                                                                         between the two conditions.
                                                                                                                                                                                                                                               A voxel-by-voxel statistical subtraction analysis (Z-score) with adjustment for multiple comparisons is performed by estimating the
                                                                                                                                                                                                                                         smoothness of subtraction images ( 40 ) following three-dimensional Gaussian filtering to enhance signal-to-noise ratio and compensate
                                                                                                                                                                                                                                         for residual anatomical variance. Typically, only those voxels with normalized CBF values larger than 60% of the global value are
                                                                                                                                                                                                                                         analyzed, because these voxels represent the gray matter of the brain. Voxels showing a significantly increased CBF compared with the
                                                                                                                                                                                                                                         average noise variance computed across all voxels (pooled variance) are identified ( 41 ). The critical level of significance is determined
                                                                                                                                                                                                                                         by using this information to adjust P = 0.05 ( 42 ). With this method, the results of interest are revealed primarily through the data analysis.
                                                                                                                                                                                                                                         However, it is also possible to perform correlations between the intensity of the rCBF responses throughout the brain and some behavioral
                                                                                                                                                                                                                                         parameter of interest, such as the perceived intensity of stimulation ( 43 ).
                                                                                                                                                                                                                                               In addition, volumes of interest (VOI) may be established within brain structures selected because of a priori hypotheses and the
                                                                                                                                                                                                                                         results of previously published PET studies. The size and shape of each VOI may be standardized across studies or determined separately
                                                                                                                                                                                                                                         according to functional criteria. We presently use a method similar to that described by Burton ( 44 ), in which voxels showing significant
                                                                                                                                                                                                                                         peak increases in CBF between comparison conditions are identified within the brain structure of interest; the voxels are progressively
                                                                                                                                                                                                                                         expanded in three dimensions to include contiguous voxels that meet the statistical criterion established by the voxel-by-voxel Z-score
                                                                                                                                                                                                                                         analysis. To determine the statistical significance of rCBF increases, a paired t statistic is computed for each VOI from the average
                                                                                                                                                                                                                                         percentage increase in CBF across all subjects. Levels of significance are established, based on the Bonferroni correction for multiple
                                                                                                                                                                                                                                         comparisons among VOI.
                                                                                                                                                                                                                                               Important Variables in the Conduct and Interpretation of PET Studies of Pain. There are now numerous studies from various
                                                                                                                                                                                                                                         facilities that have used PET during the application of experimental pain. The results are difficult to compare because they are affected by
                                                                                                                                                                                                                                         intersubject variability, type of stimulus, method of scanning, and data analysis methods. To assess the effect of some of these variables,
                                                                                                                                                                                                                                         we have conducted several investigations in normal subjects with a variety of stimulation methods. The variables we have considered thus
                                                                                                                                                                                                                                         far include gender, the physical characteristics of the stimulus, and the sources of nociceptive afferent input.
                                                                                                                                                                                                                                               Gender. The prevailing evidence suggests that although there is no reliable gender difference in pain thresholds, pain tolerance is
                                                                                                                                                                                                                                         generally higher in male than in female subjects ( 45 ). PET studies find gender differences in resting rCBF ( 46 ) or in the cerebral
                                                                                                                                                                                                                                         metabolic rate of glucose utilization ( 47  49 ). These findings suggest that there may be underlying gender differences in the neural
                                                                                                                                                                                                                                         mechanisms that mediate pain perception. Accordingly, we performed PET studies in normal right-handed male (n = 10) and female (n =
                                                                                                                                                                                                                                         10) subjects (18 to 39 years old) as they discriminated differences in the intensity of innocuous and noxious heat stimuli applied to the left
                                                                                                                                                                                                                                         forearm ( 50 ). Thermal stimuli were 40C or 50C heat, applied with a thermode as repetitive 5-sec contacts to the left volar forearm.
                                                                                                                                                                                                                                         Both male and female subjects rated the 40C stimuli as warm but not painful and the 50C stimuli as painful, but females rated the 50C
                                                                                                                                                                                                                                         stimuli as significantly more intense than did the males (P = 0.0052). Both genders showed a bilateral activation of premotor cortex during
                                                                                                                                                                                                                                         heat pain in addition to the activation of a number of contralateral structures, including the posterior insula, anterior cingulate cortex, and
the authoritative version for attribution.
                                                                                                                                                                                                                                         overlap of the activation patterns occurred between genders. However, direct image subtraction showed that females had significantly
                                                                                                                                                                                                                                         greater activation of the contralateral prefrontal cortex compared with males. A VOI comparison (t-statistic) also showed greater activation
                                                                                                                                                                                                                                         of the contralateral insula and thalamus in females compared with males (P < 0.05). These pain-related differences in brain activation may
                                                                                                                                                                                                                                         be attributed to gender, perceived pain intensity, or both factors. These results show that gender differences are important considerations in
                                                                                                                                                                                                                                         the investigation of forebrain responses to noxious stimuli.
                                                                                                                                                                                                                                           FIG. 1. Statistical map of rCBF responses of 10 males (M) and 10 females (F) to repetitive noxious heat stimulation (50C) of the left
                                                                                                                                                                                                                                           volar forearm. Color coding of Z scores as indicated by flame bar at right. The right hemisphere of the MRI stereotactic template is
                                                                                                                                                                                                                                           on the readers left. The numbers below columns of images indicate millimeters above a plane connecting the anterior and posterior
                                                                                                                                                                                                                                           commissures. In both genders, there is significant activation of the contralateral anterior cingulate cortex (+41, +37), premotor, and
                                                                                                                                                                                                                                           insular cortex (+15, +7), ipsilateral insula (+7, +15), and bilateral cerebellar vermis (12). Voxel-by-voxel analysis indicated that
                                                                                                                                                                                                                                           some structures were significantly activated (Z > 4.0) only in males (contralateral sensorimotor cortex, +52; contralateral lenticular
                                                                                                                                                                                                                                           nucleus, +2; ipsilateral prefrontal cortex, +15) and others only in females (contralateral prefrental cortex, +32; anterior insula, +2;
                                                                                                                                                                                                                                           thalamus, +15; ipsilateral lenticular nucleus, +2; contralateral cerebellum, 25). Direct comparisons of percent increase in rCBF,
                                                                                                                                                                                                                                           however, revealed that the only difference is that the contralateral thalamus, anterior insula, and prefrontal cortex show a greater
                                                                                                                                                                                                                                           response in females compared with males. Reproduced from ref. 50 with permission from the International Association for the Study
                                                                                                                                                                                                                                           of Pain.
                                                                                                                                                                                                                                               Physical Characteristics of the Noxious Stimulus. Subtraction images have been interpreted as revealing those cerebral structures
                                                                                                                                                                                                                                         that have increased synaptic activity related specifically to the central processing of the neuronal signals produced by noxious heat. In our
                                                                                                                                                                                                                                         earlier report ( 51 ), we controlled for the cerebral processes mediating the discrimination of heat intensity within the innocuous range. By
                                                                                                                                                                                                                                         cooling the skin, we were able to produce innocuous, easily perceptible differences in the degree of skin temperature increase and to
                                                                                                                                                                                                                                         duplicate the intensity differences that were applied within the noxious range. However, it is possible that by cooling the skin, we reduced
                                                                                                                                                                                                                                         the perceived difference between the innocuous stimuli below that which would exist normally. The resulting rCBF responses may have
                                                                                                                                                                                                                                         been reduced below the sensitivity of our PET analysis. To test this possibility, we performed a series of PET studies on subjects who were
                                                                                                                                                                                                                                         asked to discriminate the differences between innocuous warm stimuli delivered to the volar forearm at normal baseline skin temperature.
                                                                                                                                                                                                                                         We wished to determine whether this procedure would lead to a demonstration of rCBF increases that could be compared with those
                                                                                                                                                                                                                                         elicited by noxious heat stimulation. An equally important and related issue is whether other methods of producing pain result in the same
                                                                                                                                                                                                                                         intensity and pattern of rCBF increases as the
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         increases elicited by repetitive noxious heat applied to the skin. To examine this question, we performed PET studies on normal subjects as
                                                                                                                                                                                                                                         they experienced the deep, aching pain produced by immersion of one hand in 6C water for 105 sec. We were then able to compare the
                                                                                                                                                                                                                                         PET rCBF results obtained during warm discrimination and during tonic, noxious cold immersion with the results previously obtained
                                                                                                                                                                                                                                         during repetitive cutaneous noxious heat ( 51 ).
                                                                                                                                                                                                                                              We studied three groups of nine normal, healthy subjects, 18 to 39 years old, all of whom were given instruction and practice in the
                                                                                                                                                                                                                                         use of the visual analog scale for the estimation of stimulus intensity and unpleasantness. One group was assigned a warm discrimination
                                                                                                                                                                                                                                         task, another group rated innocuous and noxious heat intensity and unpleasantness, and the third group participated in the ice-water
                                                                                                                                                                                                                                         immersion study. In the warm discrimination study, two intensities of innocuous heat (36C and 43C) were applied with a thermode as
                                                                                                                                                                                                                                         repetitive 5-sec contacts to the volar forearm throughout the scan. Neither stimulus was rated as painful. All subjects discriminated the 43
                                                                                                                                                                                                                                         C stimulus from the 36C stimulus (P < 0.0001). Significant increases in rCBF to the 43C stimuli were found in the contralateral ventral
                                                                                                                                                                                                                                         posterior thalamus, lenticular nucleus, medial prefrontal cortex (Brodmanns areas 10 and 32), and the cerebellar vermis. In the study of
                                                                                                                                                                                                                                         noxious and innocuous heat, all subjects rated the 50C stimuli as painful and the 40C stimuli as warm, but not painful. Significant rCBF
                                                                                                                                                                                                                                         increases to 50C stimuli were found contralaterally in the lenticular nucleus, thalamus, anterior cingulate cortex, premotor cortex, and the
                                                                                                                                                                                                                                         secondary somatosensory (S2) and insular cortices. The ipsilateral premotor cortex and thalamus as well as the medial dorsal midbrain and
                                                                                                                                                                                                                                         cerebellar vermis showed significant rCBF increases. CBF increases just below the threshold for statistical significance were seen in the
                                                                                                                                                                                                                                         contralateral sensorimotor cortex (S1/M1).
                                                                                                                                                                                                                                              In the ice-water immersion study, the left hand was immersed to the wrist throughout each of six scans in water kept at an average
                                                                                                                                                                                                                                         temperature of either 20.5C  1.15C or 6.02C  1.18C on alternate scans. All subjects rated the intensity of the stimuli on a scale in
                                                                                                                                                                                                                                         which 0 = no pain and 10 = barely tolerable pain. Subjects rated the 20C water immersion as painless (average rating  SD of 0.18 
                                                                                                                                                                                                                                         0.48), but gave ratings indicating intense pain during immersion in 6C water (7.89  1.45). All subjects expressed the perception of the
                                                                                                                                                                                                                                         pain as very cold, steady, and deep. Highly significant increases in rCBF were found contralaterally in the sensorimotor cortex (M1/ S1),
                                                                                                                                                                                                                                         premotor cortex, anterior cingulate cortex, and in the region of the anterior insula and lenticular nucleus. Ipsilateral increases in rCBF were
                                                                                                                                                                                                                                         seen in the lateral prefrontal cortex (Brodmanns areas 10 and 46), anterior cingulate cortex, the region of the insular and opercular
                                                                                                                                                                                                                                         precentral cortices, and the thalamus. The cerebellar vermis also showed a significant increase in rCBF. CBF increases just below the
                                                                                                                                                                                                                                         threshold for statistical significance were seen in the contralateral thalamus.
                                                                                                                                                                                                                                              Comparisons of rCBF response magnitude were made among the five stereotactically concordant brain regions that showed
                                                                                                                                                                                                                                         significant responses in both the heat pain and cold pain conditions: the cerebellar vermis, ipsilateral thalamus, contralateral premotor
                                                                                                                                                                                                                                         cortex, contralateral anterior cingulate cortex, and the region of the contralateral anterior insula and lenticular nucleus. Each region showed
                                                                                                                                                                                                                                         a higher increase in rCBF during the cold pain study (3.26%  0.061%) than during the heat pain study (2.85%  0.124%; paired t4 = 3.60;
                                                                                                                                                                                                                                         P < 0.022).
                                                                                                                                                                                                                                              The results show that in conscious humans, two forms of noxious stimulation that are different in temporal pattern, afferent fiber
                                                                                                                                                                                                                                         activation, and perceived spatiotemporal and qualitative characteristics produce similar, but not identical, patterns of brain rCBF increases.
                                                                                                                                                                                                                                         These pain-related response patterns are each quite different from the brain responses observed during the discrimination between two
                                                                                                                                                                                                                                         intensities of innocuous heat stimuli. The results suggest that the increased rCBF responses observed during noxious stimulation reflect
                                                                                                                                                                                                                                         physiological differences in neuronal activity that are related to both nociceptive processing and the perception of pain. The overlap in the
                                                                                                                                                                                                                                         spatial distribution of rCBF increases during noxious cutaneous heat and noxious deep-cold stimulation suggests that a reproducible
                                                                                                                                                                                                                                         pattern of rCBF responses may occur that is common to the perceptions of pain produced by different stimuli. Differences in the intensity
                                                                                                                                                                                                                                         and spatial patterns of these pain-related rCBF increases may reflect physiological differences in neuronal nociceptive processing that are
                                                                                                                                                                                                                                         linked with these two forms of pain perception.
                                                                                                                                                                                                                                              The Source of Nociceptive Input. In comparing the rCBF changes induced by cutaneous-contact heat pain with that induced by
                                                                                                                                                                                                                                         deep-cold pain, we found that the brain activation patterns showed a considerable overlap; the contralateral anterior cingulate, anterior
                                                                                                                                                                                                                                         insula/lenticular nucleus, premotor cortex, and the ipsilateral thalamus and cerebellar vermis were activated by both forms of noxious
                                                                                                                                                                                                                                         stimulation ( 52 ). Because cold noxious stimulation activates cutaneous, subcutaneous, muscle, periosteal, and venous nociceptors ( 53 ),
                                                                                                                                                                                                                                         we wished to compare two forms of noxious stimulation that more selectively activate nociceptive afferents from different sources.
                                                                                                                                                                                                                                              For each of eight PET scans, 11 normal subjects rated the intensity of cutaneous and intramuscular stimuli delivered to the
                                                                                                                                                                                                                                         nondominant (left) forearm on a visual analog scale; stimulus intensity was adjusted to approximate pain threshold levels. Cutaneous pain
                                                                                                                                                                                                                                         was produced with a high-energy CO2 laser stimulator. Muscle pain was elicited with high-intensity intramuscular electrical stimulation.
                                                                                                                                                                                                                                         The pain intensity ratings and the differences between noxious and innocuous ratings were similar for cutaneous and intramuscular stimuli
                                                                                                                                                                                                                                         (P > 0.05). After stereotactic registration, statistical pixel-by-pixel summation (Z-score) and VOI analyses of subtraction images were
                                                                                                                                                                                                                                         performed. Direct statistical comparisons between cutaneous and intramuscular stimulations showed no reliable differences between these
                                                                                                                                                                                                                                         two forms of noxious stimulation, indicating that a substantial overlap occurred in brain activation patterns. These activated cerebral
                                                                                                                                                                                                                                         structures may represent those recruited early in nociceptive processing, because both forms of stimuli were near pain threshold.
                                                                                                                                                                                                                                              Increases in rCBF of 3.5% or more were seen in the contralateral S2, anterior insular, anterior cingulate, prefrontal, and inferior
                                                                                                                                                                                                                                         parietal cortices; and in the contralateral thalamus, lenticular nucleus, ipsilateral premotor cortex, and cerebellum. Cutaneous laser
                                                                                                                                                                                                                                         stimulation was relatively ineffective in evoking rCBF responses in the contralateral anterior cingulate or in the lenticular nucleus.
                                                                                                                                                                                                                                         Intramuscular stimulation was similarly ineffective in activating the contralateral prefrontal and ipsilateral premotor cortex. However, each
                                                                                                                                                                                                                                         form of stimulation evoked responses of sufficient magnitude in each structure, but a direct statistical comparison failed to differentiate
                                                                                                                                                                                                                                         significantly between them. The similar cerebral activation patterns suggest that the perceived differences between acute skin and muscle
                                                                                                                                                                                                                                         pain are mediated by differences in intensity and temporospatial patterns of neuronal activity within similar sets of forebrain structures.
                                                                                                                                                                                                                                              An Emerging Pattern. In summarizing the data obtained from right-handed subjects in our facility, we found that certain structures
                                                                                                                                                                                                                                         were activated by noxious stimuli across a wide range of conditions. The pattern of activation is best represented by inspection of the
                                                                                                                                                                                                                                         results of our study of gender ( Fig. 1 ). Among the most prominent activation sites are the contralateral insular cortex, primarily the
                                                                                                                                                                                                                                         anterior portion, and the cerebellar vermis. These structures responded to each form of noxious stimulation in all groups of subjects.
                                                                                                                                                                                                                                         Bilateral insular activation is seen, but it often does not reach statistical significance in voxel-by-voxel analysis. The contralateral
                                                                                                                                                                                                                                         thalamic responses to noxious stimuli are equally robust, but are
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         more frequently bilateral. The contralateral anterior cingulate cortex responded to all noxious stimuli except cutaneous laser pulses; clear
                                                                                                                                                                                                                                         bilateral responses were observed only during deep cold pain. Finally, the premotor cortex (Brodmanns area 6) responded bilaterally in all
                                                                                                                                                                                                                                         studies, except the study performed with a cutaneous laser, where the response was ipsilateral only; and the study performed with
                                                                                                                                                                                                                                         intramuscular stimulation, where the response was not significant. Other structures, including the S1 and S2, lateral prefrental
                                                                                                                                                                                                                                         (Brodmanns areas 10 and 46), and inferior parietal (Brodmanns area 40) cortices were significantly activated in a minority of our studies.
                                                                                                                                                                                                                                               The insular cortex has been considered a component of cerebral pain mechanisms based primarily on clinical information ( 54  57 ).
                                                                                                                                                                                                                                         Because of its anatomical connections, this region is likely to mediate affective, mnemonic, and autonomic features of pain ( 58 ). The
                                                                                                                                                                                                                                         insula receives input from the ventral medial posterior thalamus, a region that receives direct nociceptive input from the superficial dorsal
                                                                                                                                                                                                                                         horn ( 59 , 60 ). The spinothalamic tract projects also to the medial and intralaminar thalamus, the origin of thalamocortical fibers to the
                                                                                                                                                                                                                                         anterior cingulate gyrus; nociceptive neurons are found in both structures ( 6 , 61  63 ). Furthermore, clinical and experimental evidence
                                                                                                                                                                                                                                         shows that the anterior cingulate cortex is critical for normal pain-related behaviors ( 64  66 ). The activation of these thalamocortical
                                                                                                                                                                                                                                         pathways to the insular and cingulate cortices is therefore consistent with other information about nociceptive processing. How the
                                                                                                                                                                                                                                         cerebellum and premotor cortices fit into this picture is currently unclear. Based on evidence from other PET studies, it is possible that
                                                                                                                                                                                                                                         cortical and subcortical motor mechanisms become activated in anticipation of movements intended to escape the noxious stimulation (
                                                                                                                                                                                                                                         67 ).
                                                                                                                                                                                                                                               The contralateral S1 cortex responds significantly in a minority of our studies. Often there is a peak of activation within S1, but it
                                                                                                                                                                                                                                         fails to reach statistical significance in most of our studies. The activation of S1 across other PET studies of pain also seems quite variable.
                                                                                                                                                                                                                                         This observation raises again the question of the role of S1 in the cerebral processing of pain. Neurophysiological studies leave little doubt
                                                                                                                                                                                                                                         that nociceptive information reaches the S1 cortex ( 3 , 68 , 69 ). Clinical observations and PET studies show that S1 is critical for
                                                                                                                                                                                                                                         somesthetic discriminative performance, but that surgical extirpation of S1 does little to relieve pain ( 70 , 71 ). The conditions that require
                                                                                                                                                                                                                                         S1 activity for nociceptive processing have yet to be determined and present an interesting challenge for the future.
                                                                                                                                                                                                                                               Each of the five structures identified above (six, counting each hemithalmus separately) is known to participate in cerebral functions
                                                                                                                                                                                                                                         other than nociceptive processing and pain. It is premature to consider that this particular pattern of activation is unique for pain.
                                                                                                                                                                                                                                         Nonetheless, in nearly all other PET studies of experimentally applied pain, the structures named here have been activated by using a wide
                                                                                                                                                                                                                                         variety of stimuli and different data processing methods. There are other important variables to investigate. The issue of cerebral
                                                                                                                                                                                                                                         asymmetry in pain processing has yet to be addressed systematically; it is probably an important factor in pathological pain states in
                                                                                                                                                                                                                                         humans ( 72 ). It is likely that further experience with wholebrain imaging methods will allow us to identify a pain-specific pattern of
                                                                                                                                                                                                                                         cerebral activation.
                                                                                                                                                                                                                                               Meanwhile we have the opportunity to test specific hypotheses about the participation of each of these regions in nociceptive
                                                                                                                                                                                                                                         processing and pain. Introducing conditions that perturb the cerebral activation pattern can test hypotheses about the mechanisms of pain
                                                                                                                                                                                                                                         and analgesia. For example, Rainville et al. ( 73 ), by using hypnotic suggestion, were able to uncouple perceived intensity from perceived
                                                                                                                                                                                                                                         unpleasantness to demonstrate a strong correlation between the ratings of unpleasantness and the degree of rCBF increase within a portion
                                                                                                                                                                                                                                         of the anterior cingulate gyrus. And Derbyshire et al. ( 43 ) have shown that several cerebral regions, including those named above, show a
                                                                                                                                                                                                                                         significant correlation of rCBF response magnitude with perceived stimulus intensity. Information relevant to perceived stimulus intensity
                                                                                                                                                                                                                                         thus appears to be distributed widely, not simply to structures such as the S1 cortex that are known to mediate discriminative function. The
                                                                                                                                                                                                                                         ability of PET to provide information about nociceptive processing in the awake human brain offers an opportunity to study the effects of
                                                                                                                                                                                                                                         neuropathic pain, central nervous system damage, and the unique effects of analgesics. For example, although opioid analgesia specifically
                                                                                                                                                                                                                                         attenuates pain-activated, but not vibration-activated, cerebral responses, it strongly activates the anterior cingulate cortex (K.L.C., P.
                                                                                                                                                                                                                                         Svenssen, T. J. Morrow, J. Raz, C. S. Jone, and S. Minoshima, unpublished data). Such a result suggests the involvement of both spinal
                                                                                                                                                                                                                                         and supraspinal sites of analgesic action, including the participation of descending inhibitory modulation.
                                                                                                                                                                                                                                               Future progress in the analysis of the physiology of this pain-related network will require the development of an animal model for
                                                                                                                                                                                                                                         invasive studies that cannot be performed in humans. We have recently developed a model for studies of nocifensive behaviors in the rat.
                                                                                                                                                                                                                                               An Animal Model for Future rCBF Studies. We used rCBF in an animal model to identify the patterns of forebrain nociceptive
                                                                                                                                                                                                                                         processing that occur during early and late phases of the well-established formalin test of inflammatory pain in the rat ( 74 ). During the
                                                                                                                                                                                                                                         early phase, immediately after the injection of formalin into the dorsal hindpaw, pain behaviors are frequently elicited that are most
                                                                                                                                                                                                                                         intense. This phase continues for approximately 5 min, after which nociception is considerably reduced. The late phase is marked by the
                                                                                                                                                                                                                                         return of moderate to high levels of pain-related behaviors, beginning 10 to 15 minutes after formalin injection and continuing for 1 h.
                                                                                                                                                                                                                                         The early phase is thought to be caused by the direct activation of peripheral nociceptors by formalin, whereas the late phase is believed to
                                                                                                                                                                                                                                         be related to the development of inflammation and sensitization of central nociceptive neurons ( 75  78 ).
                                                                                                                                                                                                                                               We measured normalized rCBF increases by an autoradiographic method that uses the radiotracer [99m]Tc-exametazime. Rats were
                                                                                                                                                                                                                                         restraint-adapted to a soft towel for 2 to 3 weeks. To examine changes in rCBF during the early acute pain phase of the formalin test, we
                                                                                                                                                                                                                                         injected the left hindfoot of the restrained rat with 0.05 ml of a 2.5% solution of formalin. After 2 min, we injected each animal with an
                                                                                                                                                                                                                                         intravenous bolus of radiotracer. The same procedure was followed in the late phase of the test, but we injected the radiotracer 20 minutes
                                                                                                                                                                                                                                         later. After the first injection, these adapted rats showed little or no movement while in the restraint. Two to five minutes after the
                                                                                                                                                                                                                                         radiotracer injection, the rat was overdosed with anesthetic (chloral hydrate, 300 mg/kg i.v.) and decapitated; slides of the frozen brain
                                                                                                                                                                                                                                         were prepared for routine histological staining and quantitative autoradiography. Eighteen regions of interest (ROIs) were selected,
                                                                                                                                                                                                                                         representing various structures within the limbic and somatosensory systems. Densitometric analysis of autoradiograms was performed
                                                                                                                                                                                                                                         with microcomputer-assisted video imaging. Anatomic location of selected ROIs was accomplished by overlaying matching transparencies
                                                                                                                                                                                                                                         from a standard stereotactic atlas. We converted the film densities to apparent tissue radioactivity concentrations (nCi/mg) by comparing
                                                                                                                                                                                                                                         them with the film optical densities of 14C-labeled standards, allowing ROI comparisons across different films and animals. An index of
                                                                                                                                                                                                                                         activation was then calculated from individual ROI activities as a percentage of the average total activity of the entire brain. Significant
                                                                                                                                                                                                                                         differences in activation for each ROI were detected between experimental groups by using ANOVA with post-hoc t tests (P  0.05).
                                                                                                                                                                                                                                               During the early phase of the formalin test, a highly significant (31%) increase in rCBF occurred in the contralateral hindlimb cortex.
                                                                                                                                                                                                                                         At the same time the retrosplenial portion of the cingulate cortex and the midbrain periaqueductal gray
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         were activated bilaterally (31% and 7.8%, respectively). During the late phase, these structures remained active but the hindlimb activation
                                                                                                                                                                                                                                         became bilateral. In addition, the intensity of periaqueductal gray activation increased to 20% and was joined by significant rCBF
                                                                                                                                                                                                                                         increases in the interpeduncular and paraventricular nuclei (66% and 30%, respectively), in the habenular complex (58%), anterior dorsal
                                                                                                                                                                                                                                         nucleus of the thalamus (30%), and the parietal cortex (30%) adjacent to the hindlimb cortex. The somatotopic organization of the
                                                                                                                                                                                                                                         somatosensory thalamus and the small number of neurons excited by hindlimb stimulation probably resulted in an underestimation of
                                                                                                                                                                                                                                         specific thalamic nuclei activity. Nonetheless, we detected blood flow increases in the ventral posterolateral thalamus (8.7%) and in the
                                                                                                                                                                                                                                         medial thalamus (9.0%) that did not reach statistical significance but did tend to be greater in the late phase compared with the early phase
                                                                                                                                                                                                                                         of the formalin test.
                                                                                                                                                                                                                                               These results show that specific structures known to be important in nociceptive processing and modulation are selectively activated
                                                                                                                                                                                                                                         in the awake rat during the formalin test. Activation of a structure may be related to nociception, antinociception, or both. The contralateral
                                                                                                                                                                                                                                         hindlimb cortex and midbrain periaqueductal gray received nociceptive input and were active during the early phase. In the late phase,
                                                                                                                                                                                                                                         bilateral activity was seen throughout the forebrain, with the recruitment of limbic system components, each of which has been shown to
                                                                                                                                                                                                                                         participate in mediating or modulating nocifensive behaviors. In addition to the well-known analgesic effect of periaqueductal gray
                                                                                                                                                                                                                                         stimulation, interpeduncular nucleus stimulation modulates antinociceptive circuitry in the medullary raphe nuclei ( 79 ), and stimulation
                                                                                                                                                                                                                                         of the paraventricular nucleus produces analgesia ( 80 ). Analgesia also follows the microinjection of morphine and electrical stimulation
                                                                                                                                                                                                                                         within the habenular complex ( 81 , 82 ). Activation of the cingulate cortex is consistent with the activation of one of its major inputs, the
                                                                                                                                                                                                                                         anterior dorsal thalamic nuclei, and is in accord with the limbic cortical activation seen in human PET studies. Overall, the bilateral
                                                                                                                                                                                                                                         activation of somatosensory and limbic structures agrees with 2-deoxyglucose studies of glucose uptake in rats with chronic constriction
                                                                                                                                                                                                                                         injures of the sciatic nerve ( 83 ). Here we show that rCBF analysis is useful in studying central responses to acute and chronic stimuli.
                                                                                                                                                                                                                                               The Future of Pain Imaging. This developing technology may undergo significant improvements in both spatial and temporal
                                                                                                                                                                                                                                         resolution. Currently, PET provides a quantitative, statistically reliable method for assessing the activity of large brain and brainstem
                                                                                                                                                                                                                                         regions. Hypotheses can then focus on the conditions necessary and sufficient to activate one or more regions in a group of subjects.
                                                                                                                                                                                                                                         Although it is now possible to obtain reliable and quantitative information from single subjects with PET, fMRI has the ability to focus
                                                                                                                                                                                                                                         with great precision on rCBF responses in specific regions. Working together in a complementary manner, the two procedures should help
                                                                                                                                                                                                                                         develop a more precise understanding of the functional organization of pain and nociceptive processing. This progress will be facilitated
                                                                                                                                                                                                                                         by the parallel use of animal models, allowing questions about dynamics and functional connectivity to be addressed by selective
                                                                                                                                                                                                                                         stimulation, lesion, and drug microinjection studies.
                                                                                                                                                                                                                                               The clinical impact of this effort will be apparent as we develop an understanding of how the central nervous system adapts to
                                                                                                                                                                                                                                         chronic nociceptive input and injury. The changes in nociceptive processing demonstrated at the spinal cord level in experimental animals
                                                                                                                                                                                                                                         are likely to affect nociceptive processing and hence pain at higher levels. Such studies may have an important impact on descending
                                                                                                                                                                                                                                         modulatory influences, especially in forebrain-dominated animals such as humans. Evidence has accumulated showing that peripheral
                                                                                                                                                                                                                                         injury can profoundly affect thalamic and cortical sensory processes over long periods of time ( 84  86 ). In some cases, these plastic
                                                                                                                                                                                                                                         changes can be correlated with pain ( 87 ). A significant minority of patients with injury or disease of the central nervous system also
                                                                                                                                                                                                                                         suffer chronic, often unremitting pain as a consequence of the central lesion(s) ( 88 ). The pathophysiology of this condition is unknown,
                                                                                                                                                                                                                                         but the methods discussed here hold the promise for better solutions to the treatment and prevention of these chronic pain conditions.
                                                                                                                                                                                                                                         1. Melzack, R. & Casey, K. L. ( 1968 ) in The Skin Senses , eds. Kenshalo, D. R. & Thomas, C. C. ( Thomas , Springfield, IL ), pp. 423439 .
                                                                                                                                                                                                                                         2. Head, H. & Holmes, G. ( 1911 ) Brain 34 , 102254 .
                                                                                                                                                                                                                                         3. Kenshalo, D. R., Jr. , & Isensee, O. ( 1983 ) J. Neurophysiol. 50 , 14791496 .
                                                                                                                                                                                                                                         4. Foltz, E. L. & White, L. E. ( 1962 ) J. Neurosurg. 19 , 89100 .
                                                                                                                                                                                                                                         5. Hurt, R. W. & Ballantine, H. T. ( 1973 ) Clin. Neurosurg. 21 , 334351 .
                                                                                                                                                                                                                                         6. Sikes, R. W. & Vogt, B. A. ( 1992 ) J. Neurophysiol. 68 , 17201732 .
                                                                                                                                                                                                                                         7. Price, D. D. ( 1988 ) Psychological and Neural Mechanisms of Pain ( Raven , New York ), pp. 1241 .
                                                                                                                                                                                                                                         8. Gerhart, K. D. , Yezierski, R. P. , Fang, Z. R. & Willis, W. D. ( 1983 ) J. Neurophysiol. 49 , 406423 .
                                                                                                                                                                                                                                         9. Yezierski, R. P. , Gerhart, K. D. , Schrock, B. J. & Willis, W. D. ( 1983 ) J. Neurophysiol. 49 , 424441 .
                                                                                                                                                                                                                                         10. Dubner, R. , Hoffman, D. S. & Hayes, R. L. ( 1981 ) J. Neurophysiol. 46 , 444464 .
                                                                                                                                                                                                                                         11. Hayes, R. L. , Dubner, R. & Hoffman, D. S. ( 1981 ) J. Neurophysiol. 46 , 428443 .
                                                                                                                                                                                                                                         12. Basbaum, A. I. & Fields, H. L. ( 1984 ) Annu. Rev. Neurosci. 7 , 309338 .
                                                                                                                                                                                                                                         13. Yuan, B. , Morrow, T. J. & Casey, K. L. ( 1986 ) J. Neurosci. 6 , 36113617 .
                                                                                                                                                                                                                                         14. Shin, H. C. & Chapin, J. K. ( 1990 ) Somatosens. Mot. Res. 7 , 421434 .
                                                                                                                                                                                                                                         15. Alloway, K. D. , Johnson, M. J. & Wallace, M. B. ( 1993 ) J. Neurophysiol. 70 , 892908 .
                                                                                                                                                                                                                                         16. Liu, X. B. , Honda, C. N. & Jones, E. G. ( 1995 ) J. Comp. Neurol. 352 , 6991 .
                                                                                                                                                                                                                                         17. Swanson, L. W. ( 1995 ) Trends Neurosci. 18 , 471474 .
                                                                                                                                                                                                                                         18. Cheema, S. S. , Rustioni, A. & Whitsel, B. L. ( 1984 ) J. Comp. Neurol. 225 , 276290 .
                                                                                                                                                                                                                                         19. Ralston, D. D. & Ralston, H. J. , III ( 1985 ) J. Comp. Neurol. 242 , 325337 .
                                                                                                                                                                                                                                         20. Blinkov, S. M. & Glezer, I. I. ( 1968 ) The Human Brain in Figures and Tables ( Plenum , New York ).
                                                                                                                                                                                                                                         21. Towe, A. L. ( 1995 ) J. Brain Res. 36 , 393398 .
                                                                                                                                                                                                                                         22. Liu, X.-B. , Honda, C. N. & Jones, E. G. ( 1995 ) J. Comp. Neurol. 352 , 6991 .
                                                                                                                                                                                                                                         23. Ogawa, S. , Lee, T.-M., Kay, A. R. & Tank, D. ( 1990 ) Proc. Natl. Acad. Sci. USA 87 , 98689872 .
                                                                                                                                                                                                                                         24. Roy, C. S. & Sherrington, C. S. ( 1890 ) J. Physiol. (London) 11 , 85108 .
                                                                                                                                                                                                                                         25. Roland, P. E. ( 1993 ) Brain Activation ( Wiley-Liss , New York ).
                                                                                                                                                                                                                                         26. Malonek, D. & Grinvald, A. ( 1996 ) Science 272 , 551554 .
                                                                                                                                                                                                                                         27. Wang, G. , Tanaka, K. & Tanifuji, M. ( 1996 ) Science 272 , 1665 1668 .
                                                                                                                                                                                                                                         28. MacVicar, B. A. ( 1997 ) Neuroscientist 3 , 381388 .
                                                                                                                                                                                                                                         29. Mraovitch, S. , Calando, Y. , Pinard, E. , Pearce, W. J. & Seylaz, J. ( 1992 ) Neuroscience 49 , 451466 .
                                                                                                                                                                                                                                         30. Adachi, K. , Takahashi, S. , Melzer, P. , Campos, K. L. , Nelson, T. , Kennedy, C. & Sokoloff, L. ( 1994 ) Am. J. Physiol. 267 , H2155 H2162 .
                                                                                                                                                                                                                                         31. Dirnagl, U. , Niwa, K. , Lindauer, U. & Villringer, A. ( 1994 ) Am. J. Physiol. 267 , H296H301 .
                                                                                                                                                                                                                                         32. Seitz, R. J. & Roland, P. E. ( 1992 ) Acta Neurol. Scand. 86 , 6067 .
                                                                                                                                                                                                                                         33. Auker, C. R. , Meszler, R. M. & Carpenter, D. O. ( 1983 ) J. Neurophysiol. 49 , 15041516 .
the authoritative version for attribution.
                                                                                                                                                                                                                                         39. Minoshima, S. , Koeppe, R. A. , Frey, K. A. & Kuhl, D. E. ( 1994 ) J. Nucl. Med. 35 , 15281536 .
                                                                                                                                                                                                                                         40. Friston, K. J. , Frith, C. D. , Liddle, P. F. & Frackowiak, R. S. ( 1991 ) J. Cereb. Blood Flow Metab. 11 , 690699 .
                                                                                                                                                                                                                                         41. Worsley, K. J. , Evans, A. C. , Marrett, S. & Neelin, P. ( 1992 ) J. Cereb. Blood Flow Metab. 12 , 900918 .
                                                                                                                                                                                                                                         42. Adler, R. J. & Hasofer, A. M. ( 1976 ) Ann. Probabil. 4 , 112 .
                                                                                                                                                                                                                                         43. Derbyshire, S. W. , Jones, A. K. , Gyulai, F. , Clark, S. , Townsend, D. & Firestone, L. L. ( 1997 ) Pain 73 , 431445 .
                                                                                                                                                                                                                                         44. Burton, H. , Videen, T. O. & Raichle, M. E. ( 1993 ) Somatosens. Mot. Res. 10 , 297308 .
                                                                                                                                                                                                                                         45. Goolkasian, P. ( 1985 ) Psychol. Women Q. 9 , 1528 .
                                                                                                                                                                                                                                         46. Gur, R. C. , Gur, R. E. , Obrist, W. D. , Hungerbuhler, J. P. , Younkin, D. , Rosen, A. D. , Skolnick, B. E. & Reivich, M. ( 1982 ) Science 217 , 659
                                                                                                                                                                                                                                              661 .
                                                                                                                                                                                                                                         47. Gur, R. C. , Mozley, L. H. , Mozley, P. D. , Resnick, S. M. , Karp, J. S. , Alavi, A. , Arnold, S. E. & Gur, R. E. ( 1995 ) Science 267 , 528531 .
                                                                                                                                                                                                                                         48. Andreason, P. J. , Zametkin, A. J. , Guo, A. C. , Baldwin, P. & Cohen, R. M. ( 1994 ) Psychiatry Res. 51 , 175183 .
                                                                                                                                                                                                                                         49. Azari, N. P. , Rapoport, S. I. , Grady, C. L. , DeCarli, C. , Haxby, J. V. , Schapiro, M. B. & Horwitz, B. ( 1992 ) Brain Res. 574 , 198208 .
                                                                                                                                                                                                                                         50. Paulson, P. E. , Minoshima, S. , Morrow, T. J. & Casey, K. L. ( 1998 ) Pain 76 , 223229 .
                                                                                                                                                                                                                                         51. Casey, K. L. , Minoshima, S. , Berger, K. L. , Koeppe, R. A. , Morrow, T. J. & Frey, K. A. ( 1994 ) J. Neurophysiol. 71 , 802807 .
                                                                                                                                                                                                                                         52. Casey, K. L. , Minoshima, S. , Morrow, T. J. & Koeppe, R. A. ( 1996 ) J. Neurophysiol. 76 , 571581 .
                                                                                                                                                                                                                                         53. Klement, W. & Arndt, J. O. ( 1992 ) J. Physiol. (London) 449 , 7383 .
                                                                                                                                                                                                                                         54. Biemond, A. ( 1956 ) A. M. A. Arch. Neurol. Psychiatry 75 , 231244 .
                                                                                                                                                                                                                                         55. Schmahmann, J. D. & Leifer, D. ( 1992 ) Arch. Neurol. 49 , 10321037 .
                                                                                                                                                                                                                                         56. Bassetti, C. , Bogousslavsky, J. & Regli, F. ( 1993 ) Neurology 43 , 19421949 .
                                                                                                                                                                                                                                         57. Greenspan, J. D. & Winfield, J. A. ( 1992 ) Pain 50 , 2939 .
                                                                                                                                                                                                                                         58. Saper, C. B. ( 1982 ) J. Comp. Neurol. 210 , 163173 .
                                                                                                                                                                                                                                         59. Craig, A. D. , Bushnell, M. C. , Zhang, E.-T. & Blomqvist, A. ( 1994 ) Nature (London) 372 , 770773 .
                                                                                                                                                                                                                                         60. Craig, A. D. & Bushnell, M. C. ( 1994 ) Science 265 , 252255 .
                                                                                                                                                                                                                                         61. Casey, K. L. ( 1966 ) J. Neurophysiol. 29 , 727750 .
                                                                                                                                                                                                                                         62. Dong, W. K. , Ryu, H. & Wagman, I. H. ( 1978 ) J. Neurophysiol. 41 , 15921613 .
                                                                                                                                                                                                                                         63. Bushnell, M. C. & Duncan, G. H. ( 1989 ) Exp. Brain Res. 78 , 415418 .
                                                                                                                                                                                                                                         64. Vogt, B. A. , Sikes, R. W. & Vogt, L. J. ( 1993 ) in Neurobiology of Cingulate Cortex and Limbic Thalamus: A Comprehensive Handbook , eds.
                                                                                                                                                                                                                                              Vogt, B. A. & Gabriel, M. ( Birkhauser , Boston ).
                                                                                                                                                                                                                                         65. Devinsky, O. , Morrell, M. J. & Vogt, B. A. ( 1995 ) Brain 118 , 279306 .
                                                                                                                                                                                                                                         66. Gabriel, M. & Poremba, A. ( 1995 ) in The Role of Pain in Cingulate Cortical and Limbic Thalamic Mediation of Avoidance Learning , eds. Besson,
                                                                                                                                                                                                                                              J.-M. , Guilbaud, G. & Ollat, H. ( John Libbey, Eurotext , Paris ) pp. 197212 .
                                                                                                                                                                                                                                         67. Hsieh, J.-C. , Hagermark, O. , Stahle-Backdahl, M. , Ericson, K. , Eriksson, L. , Stone-Elander, S. & Ingvar, M. ( 1994 ) J. Neurophysiol. 72 , 3004
                                                                                                                                                                                                                                              3008 .
                                                                                                                                                                                                                                         68. Kenshalo, D. R. , Chudler, E. H. , Anton, F. & Dubner, R. ( 1988 ) Brain Res. 454 , 378382 .
                                                                                                                                                                                                                                         69. Chudler, E. H. , Anton, F. , Dubner, R. & Kenshalo, D. R. , Jr. ( 1990 ) J. Neurophysiol. 63 , 559569 .
                                                                                                                                                                                                                                         70. White, J. C. & Sweet, W. H. ( 1969 ) Pain and the Neurosurgeon. A Forty-Year Experience ( Thomas , Springfield, IL ).
                                                                                                                                                                                                                                         71. Sweet, W. H. ( 1982 ) in New Perspectives in Cerebral Localization , eds. Thompson, R. A. & Green, J. R. ( Raven , New York ), pp. 205242 .
                                                                                                                                                                                                                                         72. Nasreddine, Z. S. & Saver, J. L. ( 1997 ) Neurology 48 , 11961199 .
                                                                                                                                                                                                                                         73. Rainville, P. , Duncan, G. H. , Price, D. D. , Carrier, M. & Bushnell, M. C. ( 1997 ) Science 277 , 968971 .
                                                                                                                                                                                                                                         74. Morrow, T. J. , Paulson, P. E. , Danneman, P. J. & Casey, K. L. ( 1998 ) Pain 75 , 355365 .
                                                                                                                                                                                                                                         75. Coderre, T. J. , Vaccarino, A. L. & Melzack, R. ( 1990 ) Brain Res. 535 , 155158 .
                                                                                                                                                                                                                                         76. Coderre, T. J , Katz, J. , Vaccarino, A. L. & Melzack, R. ( 1993 ) Pain 52 , 259285 .
                                                                                                                                                                                                                                         77. Wiebe, J. P. & Kavaliers, M. ( 1988 ) Brain Res. 461 , 150157 .
                                                                                                                                                                                                                                         78. Brown, A. G. ( 1981 ) Organization in the Spinal Cord ( Springer , New York ).
                                                                                                                                                                                                                                         79. Hentall, I. D. & Budhrani, V. M. ( 1990 ) Brain Res. 522 , 322324 .
                                                                                                                                                                                                                                         80. Yirmiya, R. , Ben-Eliyahu, S. , Shavit, Y. , Marek, P. & Liebeskind, J. C. ( 1990 ) Brain Res. 537 , 169174 .
                                                                                                                                                                                                                                         81. Cohen, S. R. & Melzack, R. ( 1985 ) Brain Res. 359 , 131139 .
                                                                                                                                                                                                                                         82. Cohen, S. R. & Melzack, R. ( 1986 ) Neurosci. Lett. 70 , 165169 .
                                                                                                                                                                                                                                         83. Mao, J. , Mayer, D. J. & Price, D. D. ( 1993 ) J. Neurosci. 13 , 26892702 .
                                                                                                                                                                                                                                         84. Kaas, J. H. ( 1991 ) Annu. Rev. Neurosci. 14 , 137167 .
                                                                                                                                                                                                                                         85. Dubner, R. & Ruda, M. A. ( 1992 ) Trends Neurosci. 15 , 96103 .
                                                                                                                                                                                                                                         86. Kaas, J. H. , Florence, S. L. & Neeraj, J. ( 1997 ) Neuroscientist 3 , 123130 .
                                                                                                                                                                                                                                         87. Flor, H. , Elbert, T. , Knecht, S. , Wienbruch, C. , Pantev, C. , Birbaumer, N. , Larbig, W. & Taub, E. ( 1995 ) Nature (London) 375 , 482484 .
                                                                                                                                                                                                                                         88. Casey, K. L. ( 1991 ) Pain and Central Nervous System Disease: The Central Pain Syndromes ( Raven , New York ), pp. 1280 .
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                           FIG. 1. Transverse section of the spinal cord of a patient whose colon cancer pain was relieved by a limited midline myelotomy at
                                                                                                                                                                                                                                           T10. The section was taken at a level just rostral to the site of the lesion and was stained for myelin. A bilateral demyelinated area in
                                                                                                                                                                                                                                           the fasciculus gracilis is seen below the arrow. The drawing shows the demyelinated area in black. [Figure reproduced with
                                                                                                                                                                                                                                           permission from ref. 5 (Copyright 1996, International Association for the Study of Pain).]
                                                                                                                                                                                                                                         determined that the surgical lesion interrupted the medial fasciculus gracilis bilaterally, as shown in Fig. 1 . The lesion did not appear to
                                                                                                                                                                                                                                         intrude into the central gray matter.
                                                                                                                                                                                                                                           FIG. 2. Effects of DC and VLC lesions at T10 in rats (n = 20) on responses to mechanical stimulation of the skin (Brush, Press, and
                                                                                                                                                                                                                                           Pinch) and to graded intensities of CRD (20, 40, 60 and 80 mm Hg). The responses were normalized. * indicates a significant change.
                                                                                                                                                                                                                                           [Figure reproduced with permission from ref. 7 (Copyright 1996, The American Physiological Society).]
                                                                                                                                                                                                                                           FIG. 3. Effects of microdialysis administration of drugs that block nociceptive synaptic transmission into the sacral spinal cord on
                                                                                                                                                                                                                                           responses of gracile neurons (n = 10) to graded intensities of CRD. The drugs included morphine and CNQX (6-cyano-7-
                                                                                                                                                                                                                                           nitroquinoxaline-2,3-dione). Naloxone was given systemically to reverse the action of morphine. In some animals (n = 3), a lesion
                                                                                                                                                                                                                                           was placed in the DC. * indicates a significant change. [Figure reproduced with permission from ref. 11 (Copyright 1996, The
                                                                                                                                                                                                                                           American Physiological Society).]
                                                                                                                                                                                                                                              Experimental studies therefore were begun to determine whether there is a visceral nociceptive pathway in the dorsal column (DC).
                                                                                                                                                                                                                                         Recordings were made in the ventral posterolateral (VPL) nucleus of the rat thalamus from neurons that responded to colorectal distention
                                                                                                                                                                                                                                         (CRD) or to inflammation of the colon ( 5 , 7 ). The neurons also responded to mechanical stimuli applied to the skin. Sequential lesions of
                                                                                                                                                                                                                                         the DC and the spinothalamic tract in the ventrolateral column (VLC) were made to see whether these affected the responses to noxious
                                                                                                                                                                                                                                         stimulation of the colon or to stimulation of the cutaneous receptive field. A DC lesion was found to reduce the responses to CRD by 60
                                                                                                                                                                                                                                         80%, whereas a VLC lesion only reduced such responses by 20% ( Fig. 2 B). The DC lesion also
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 4. Morphological studies of the origins of axons in the medial DC that might mediate the transmission of visceral nociceptive
                                                                                                                                                                                                                                           information. The retrograde tracer, WGA-HRP, was injected into the DCs at the cervical level, taken up by axons, and transported
                                                                                                                                                                                                                                           caudally to label numerous cell bodies in the central region of the spinal cord adjacent to the central canal (CC). [Figure reproduced
                                                                                                                                                                                                                                           with permission from ref. 5 (Copyright 1996, International Association for the Study of pain).]
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         profoundly reduced the responses to weak stimulation of the skin but had little effect on those to noxious pinch ( Fig. 2 A). A VLC lesion
                                                                                                                                                                                                                                         nearly eliminated responses to pinch but had only limited effects on responses to weak stimulation of the skin. A DC lesion also reduced
                                                                                                                                                                                                                                         the heightened activity of VPL neurons after colon inflammation; a VLC lesion had much less effect. A small electrolytic or kainic acid
                                                                                                                                                                                                                                         lesion in the gracile nucleus dramatically reduced the responses of VPL neurons to CRD ( 8 ). It was concluded that the DC is more
                                                                                                                                                                                                                                         important than the VLC for transmitting nociceptive signals from the colon to the VPL nucleus ( 7 ). Similar results with respect to CRD
                                                                                                                                                                                                                                         were obtained in monkeys ( 9 ).
                                                                                                                                                                                                                                           FIG. 5. Summary diagram depicting the course of axons arising from postsynaptic DC neurons in the visceral processing region
                                                                                                                                                                                                                                           adjacent to the central canal. Axons arising from neurons near the central canal at sacral levels of the spinal cord ascend in the
                                                                                                                                                                                                                                           midline of the DC adjacent to the medial septum to innervate the medial gracile nucleus. Axons arising from neurons near the central
                                                                                                                                                                                                                                           canal at thoracic levels of the spinal cord ascend in the DC adjacent to the dorsal intermediate septum and innervate both the gracile
                                                                                                                                                                                                                                           and cuneate nuclei.
                                                                                                                                                                                                                                               The visceral nociceptive signals that reach the gracile nucleus could be transmitted by the collaterals of primary afferent neurons that
                                                                                                                                                                                                                                         ascend directly to the gracile nucleus or by the axons of postsynaptic DC neurons (see review in ref. 10 ). Which of the routes was the
                                                                                                                                                                                                                                         more effective was tested by blocking nociceptive transmission from the colon in the sacral spinal cord by using microdialysis
                                                                                                                                                                                                                                         administration of morphine or of the non-N-methyl-D-aspartic acid receptor antagonist CNQX (6-cyano-7-nitroquinoxaline-2,3-dione) (
                                                                                                                                                                                                                                         11 ). Access to the spinal cord by colon afferents was restricted to the pelvic nerve distribution by sectioning the hypogastric nerves
                                                                                                                                                                                                                                         bilaterally. Administration of either drug blocked the responses of gracile neurons to CRD ( Fig. 3 ). Naloxone reversed the action of
                                                                                                                                                                                                                                         morphine. Similar effects were observed when recordings were made from identified postsynaptic DC neurons in the central gray region
                                                                                                                                                                                                                                         of the sacral cord. Neurons in this region are known to have a strong visceral input, and it proved to be the case for postsynaptic DC
                                                                                                                                                                                                                                         neurons located in this region. It was concluded that most of the responses of gracile neurons to CRD distention (or colon inflammation)
                                                                                                                                                                                                                                         depended on activation of postsynaptic DC neurons, although it could not be ruled out that some of the responses were the result of
                                                                                                                                                                                                                                         conduction in directly projecting primary afferents.
                                                                                                                                                                                                                                           FIG. 6. Reduction in the writhing reflex (WR) evoked by balloon distention of the duodenum after a DC lesion, (a) The intensities of
                                                                                                                                                                                                                                           the WR evoked by graded distention (by 0.1 to 0.7 ml) of the duodenum are shown. No differences in the behavioral response were
                                                                                                                                                                                                                                           seen between rats tested before the sham surgery (pre-SH) and rats tested after the sham surgery (SH). The scale used was from 0
                                                                                                                                                                                                                                           (normal) to 4 (stretching of the body, extension of the hind limbs). (b) The effect of the DC lesion on the behavioral response to
                                                                                                                                                                                                                                           duodenal distention is shown (prelesion vs. after lesion). (c) A comparison of the behavior in the sham surgery and lesion groups. The
                                                                                                                                                                                                                                           numbers of animals used are shown. * indicate significant changes. [Figure reproduced with permission from ref. 13 (Copyright
                                                                                                                                                                                                                                           1998, Lippincot Williams & Wilkins, http://lww.com ).]
the authoritative version for attribution.
                                                                                                                                                                                                                                         anterograde tracer made into the central canal region of the sacral cord (C.-C. Wang, W.D.W., and K.N.W., unpublished work). The
                                                                                                                                                                                                                                         visceral postsynaptic DC pathway arising from the sacral spinal cord ascends in the DC midline adjacent to the medial septum. Similar
                                                                                                                                                                                                                                         injections into the central gray region at a midthoracic level label axons that ascend adjacent to the dorsal intermediate septum. These
                                                                                                                                                                                                                                         axonal projections also have terminations in the gracile and cuneate nuclei. A summary diagram representing these pathways is shown in
                                                                                                                                                                                                                                         Fig. 5 .
                                                                                                                                                                                                                                           FIG. 7. Functional MRI (fMRI) study of the effects of a DC lesion on blood volume changes evoked in the brain after noxious
                                                                                                                                                                                                                                           distention of the colon in monkeys. (Left) The brain slices shown were taken from a monkey that was subjected to sham surgery at
                                                                                                                                                                                                                                           T10. The fMRI images were from a comparable location before (Upper) and then 4 months after the sham surgery (Lower). (Right)
                                                                                                                                                                                                                                           The images were made before and 4 months after a DC lesion at T10 in a different monkey. Colored regions indicate locations where
                                                                                                                                                                                                                                           regional cerebral blood volume increased relative to the nonstimulated state.
                                                                                                                                                                                                                                                                        BEHAVIORAL EVIDENCE FOR A DC VISCERAL PAIN PATHWAY
                                                                                                                                                                                                                                              To determine whether the DC helps mediate pain originating in abdominal viscera, experiments were done in awake, behaving rats on
                                                                                                                                                                                                                                         the effects of a DC lesion on the behavioral responses to pancreatitis and noxious chemical stimulation of the pancreas and to duodenal
                                                                                                                                                                                                                                         distention.
                                                                                                                                                                                                                                              A pancreatitis model induced by infusion of glycodeoxycholic acid into the pancreas and i.p. caerulian was shown to reduce home-
                                                                                                                                                                                                                                         cage exploratory behaviors (rearing) in rats ( 12 ). Decreases in normal activity did not occur in animals that had received lesions of their
                                                                                                                                                                                                                                         DC at the C1 spinal level 1 week before induction of the pancreatitis, suggesting that the DC lesion provided a protective effect. More
                                                                                                                                                                                                                                         recent unpublished data confirmed that responses of VPL neurons to stimulation of the pancreas with bradykinin also are reduced by a DC
                                                                                                                                                                                                                                         lesion and also by spinal administration of morphine at a midthoracic level. The effect of morphine is naloxone reversible.
                                                                                                                                                                                                                                              In another series of experiments, a balloon catheter was chronically implanted in the duodenum in rats through the stomach wall (
                                                                                                                                                                                                                                         13 ). Graded distention of the duodenum in awake, behaving animals resulted in graded intensities of the writhing reflex. A lesion of the
                                                                                                                                                                                                                                         DC at C2 produced a dramatic reduction in the intensity of the writhing reflex ( Fig. 6 ). However, to be effective, the lesion had to include
                                                                                                                                                                                                                                         the region of the dorsal intermediate septum bilaterally. Parallel experiments were done in anesthetized rats to determine the effects of a
                                                                                                                                                                                                                                         DC lesion at C2 on the responses of VPL neurons to duodenal distention. Again, a lesion of the DC that included the region of the dorsal
                                                                                                                                                                                                                                         intermediate septum bilaterally resulted in a profound reduction in the responses of the VPL neurons. The requirement for the placement
                                                                                                                                                                                                                                         of the DC lesions laterally to the midline is explained by the morphological study described in the previous section and illustrated in
                                                                                                                                                                                                                                         Fig. 5 . Axons originating from postsynaptic dorsal horn neurons in the central gray region of the midthoracic spinal cord travel toward the
                                                                                                                                                                                                                                         DC nuclei near the dorsal intermediate septum.
                                                                                                                                                                                                                                                                                    FUNCTIONAL MRI STUDIES IN MONKEYS
                                                                                                                                                                                                                                              In a recent unpublished study, the brain structures involved in mediating visceral nociceptive responses have been investigated by
                                                                                                                                                                                                                                         using functional MRI. Monkeys were anesthetized with isoflurane and placed in a 4.7-Tesla magnet for imaging regional cerebral blood
                                                                                                                                                                                                                                         volume. The blood was labeled with a superparamagentic iron oxide compound to enhance the contrast in the image in proportion to
                                                                                                                                                                                                                                         increases in regional blood volume. Relative cerebral blood flow was estimated by gradient echo bolus tracking, and changes in cerebral
the authoritative version for attribution.
                                                                                                                                                                                                                                         CRD consistently caused regional blood volume increases in a number of brain structures, including the VPL nucleus. Blood volume
                                                                                                                                                                                                                                         changes were not reduced after sham surgery, but they were essentially completely eliminated after the DC lesion. The effects of the DC
                                                                                                                                                                                                                                         lesion persisted for at least 4 months ( Fig. 7 ).
                                                                                                                                                                                                                                              We thank Kelli Gondesen and Jingna Wei for their expert technical assistance. The work was supported by National Institutes of
                                                                                                                                                                                                                                         Health Grant NS 11255.
                                                                                                                                                                                                                                         1. Gybels, J. M. & Sweet, W. H. ( 1989 ) Neurosurgical Treatment of Persistent Pain (Karger, Basel).
                                                                                                                                                                                                                                         2. Hitchcock, E. R. ( 1970 ) J. Neurol. Neurosurg. Psychiatry 33 , 224230 .
                                                                                                                                                                                                                                         3. Schvarcz, J. R. ( 1984 ) Acta Neurochir. 33, Suppl. , 431435 .
                                                                                                                                                                                                                                         4. Gildenberg, P. L. & Hirshberg, R. M. ( 1984 ) J. Neurol. Neurosurg. Psychiatry 47 , 9496 .
                                                                                                                                                                                                                                         5. Hirshberg, R. M. , Al-Chaer, E. D. , Lawand, N. B. , Westlund, K. N. & Willis, W. D. ( 1996 ) Pain 67 , 291305 .
                                                                                                                                                                                                                                         6. Nauta, H. J. W. , Hewitt, E. , Westlund, K. N. & Willis, W. D. ( 1997 ) J. Neurosurg. 86 , 538542 .
                                                                                                                                                                                                                                         7. Al-Chaer, E. D. , Lawand, N. B. , Westlund, K. N. & Willis, W. D. ( 1996 ) J. Neurophysiol 76 , 26612674 .
                                                                                                                                                                                                                                         8. Al-Chaer, E. D. , Westlund, K. N. & Willis, W. D. ( 1997 ) J. Neurophysiol. 78 , 521527 .
                                                                                                                                                                                                                                         9. Al-Chaer, E. D. , Feng, Y. & Willis, W. D. ( 1998 ) J. Neurophysiol. 79 , 31433150 .
                                                                                                                                                                                                                                         10. Willis, W. D. & Coggeshall, R. E. ( 1991 ) Sensory Mechanisms of the Spinal Cord ( Plenum , New York ).
                                                                                                                                                                                                                                         11. Al-Chaer, E. D , Lawand, N. B. , Westlund, K. N. & Willis, W. D. ( 1996 ) J. Neurophysiol. 76 , 26752690 .
                                                                                                                                                                                                                                         12. Houghton, A. K. , Kadura, S. & Westlund, K. N. ( 1997 ) NeuroReport 8 , 37953800 .
                                                                                                                                                                                                                                         13. Feng, Y. , Cui, M. , Al-Chaer, E. D. & Willis, W. D. ( 1998 ) Anesthesiology 89 , 411420 .
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                               TONY L. YAKSH * , XIAO-YING HUA* , IVETA KALCHEVA*, NATSUKO NOZAKI-TAGUCHI *  , AND MARTIN MARSALA*
                                                                                                                                                                                                                                              *  Department of Anesthesiology, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA 92093-0818; and
                                                                                                                                                                                                                                         Department of Anesthesiology, Chiba University School of Medicine, 18-1 Inohana Chuo-ku Chiba-shi, 260 Japan
                                                                                                                                                                                                                                               ABSTRACT Behavioral models indicate that persistent small afferent input, as generated by tissue injury, results in a
                                                                                                                                                                                                                                         hyperalgesia at the site of injury and a tactile allodynia in areas adjacent to the injury site. Hyperalgesia reflects a sensitization of
                                                                                                                                                                                                                                         the peripheral terminal and a central facilitation evoked by the persistent small afferent input. The allodynia reflects a central
                                                                                                                                                                                                                                         sensitization. The spinal pharmacology of these pain states has been defined in the unanesthetized rat prepared with spinal
                                                                                                                                                                                                                                         catheters for injection and dialysis. After tissue injury, excitatory transmitters (e.g., glutamate and substance P) acting though N-
                                                                                                                                                                                                                                         methyl-D-aspartate (NMDA) and neurokinin 1 receptors initiate a cascade that evokes release of (i) NO, (ii) cyclooxygenase
                                                                                                                                                                                                                                         products, and (iii) activation of several kinases. Spinal dialysis show amino acid and prostanoid release after cutaneous injury.
                                                                                                                                                                                                                                         Spinal neurokinin 1, NMDA, and non-NMDA receptors enhance spinal prostaglandin E2 release. Spinal prostaglandins facilitate
                                                                                                                                                                                                                                         release of spinal amino acids and peptides. Activation by intrathecal injection of receptors on spinal C fiber terminals (,/ opiate,
                                                                                                                                                                                                                                         2 adrenergic, neuropeptide Y) prevents release of primary afferent peptides and spinal amino acids and blocks acute and
                                                                                                                                                                                                                                         facilitated pain states. Conversely, consistent with their role in facilitated processing, NMDA, cyclooxygenase 2, and NO synthase
                                                                                                                                                                                                                                         inhibitors act to diminish only hyperalgesia. Importantly, spinal delivery of several of these agents diminishes human injury pain
                                                                                                                                                                                                                                         states. This efficacy emphasizes (i) the role of facilitated states in humans, (ii) shows the importance of spinal systems in human
                                                                                                                                                                                                                                         pain processing, and (iii) indicates that these preclinical mechanisms reflect processes that regulate the human pain experience.
                                                                                                                                                                                                                                               Local tissue injury and inflammation yields well-defined escape behaviors in animals and pain reports in humans. Examination of the
                                                                                                                                                                                                                                         histochemistry and electrophysiology of spinal systems has revealed considerable detail regarding the elements of systems that are
                                                                                                                                                                                                                                         activated by these stimuli. Nevertheless, the functional contribution of different spinal systems in pain processing ultimately must be
                                                                                                                                                                                                                                         defined in terms of the systems in which such end points can be measured, e.g., the behavior of the intact organism. We will consider
                                                                                                                                                                                                                                         below how certain spinal systems contribute to the observed behavioral states.
                                                                                                                                                                                                                                                                          BEHAVIORAL EFFECTS OF CUTANEOUS STIMULI AFTER INJURY
                                                                                                                                                                                                                                               An acute, unconditioned, thermal, or mechanical stimulus sufficient to activate polymodel nociceptive afferents (C fibers) depolarizes
                                                                                                                                                                                                                                         populations of dorsal horn wide dynamic range (WDR) neurons that project supraspinally. This output in turn evokes a supraspinally
                                                                                                                                                                                                                                         organized escape behavior. The hot plate test (thermal stimulus to the paw) or the local injection of an irritant such as formalin or
                                                                                                                                                                                                                                         capsaicin where the unconditioned stimulus evokes a somatotopically directed behavior (e.g., withdrawal or licking) are behavioral
                                                                                                                                                                                                                                         paradigms believed to reflect this underlying mechanism ( 1 ). The more intense the stimulus, the more robust will be the afferent volley
                                                                                                                                                                                                                                         and the more vigorous or shorter latencied is the escape behavior ( 2 ).
                                                                                                                                                                                                                                               An acute stimulus of intensity and duration that leads to tissue injury also produces an acute discharge. In addition, the injury leads to
                                                                                                                                                                                                                                         the local release of active factors that evoke and sustain persistent activity in the sensory afferents innervating the injured or inflamed
                                                                                                                                                                                                                                         tissue ( 3 ). Thus, in contrast to the acute response, injury leads to persistent activity in populations of small afferents and also may activate
                                                                                                                                                                                                                                         afferent populations that are excited only in the presence of local factors generated by the injury (e.g., silent nociceptors) ( 4 ).
                                                                                                                                                                                                                                         Electrophysiological studies have shown that the persistent activation of spinal WDR neurons by small, but not large, afferents, will lead:
                                                                                                                                                                                                                                         (i) a progressive enhancement of the WDR response to each subsequent input, and (ii) an increase in the dimensions of the peripheral
                                                                                                                                                                                                                                         receptive field to which the spinal neuron will respond ( 5 ). This electrophysiological observation parallels behavioral changes in which
                                                                                                                                                                                                                                         the animal displays an enhanced response to a given stimulus or a reduction in the intensity of the stimulus required to evoke an escape
                                                                                                                                                                                                                                         response. Thus, the injection of an irritant (formalin) into one hind paw evokes a high frequency barrage during the first 1020 min
                                                                                                                                                                                                                                         followed by a modest ongoing discharge over the next hour ( 6 ). Coincident with the initial afferent barrage, WDR neurons display an
                                                                                                                                                                                                                                         initial burst of activity followed by a period of quiescence and then a progressive enhanced barrage ( 7 ). In rats, injection of formalin
                                                                                                                                                                                                                                         results in a prominent licking and flinching of the injected paw with the incidence of flinching showing a biphasic time course that
                                                                                                                                                                                                                                         parallels that reported for the discharge of spinal WDR neurons (see Fig. 1 ). The first-phase behavior is the result of an initial intense
                                                                                                                                                                                                                                         afferent barrage. The secondphase behavior is believed to represent the induction of a state of spinal facilitation in which the diminished
                                                                                                                                                                                                                                         formalin-initiated afferent input yields a prominent response.
                                                                                                                                                                                                                                               Alternately, after a mild local burn, there is a decreased nociceptive threshold to heat within the burn area (a 1 thermal hyperalgesia)
                                                                                                                                                                                                                                         and a pain response generated by light touch applied to uninjured skin regions adjacent to the area of injury (a 2 tactile allodynia) (see
                                                                                                                                                                                                                                         Fig. 2 ) ( 8 ). Importantly, at least the initiating component of this hyperalgesia reflects on
the authoritative version for attribution.
                                                                                                                                                                                                                                         small afferent input. The intradermal injection of capsaicin, an agent known to selectively activate C fibers, can induce a 2 allodynia in
                                                                                                                                                                                                                                         humans and animals ( 9 ). This altered sensory condition persists after the termination of the pain produced by the capsaicin injection and
                                                                                                                                                                                                                                         extends anatomically beyond the local site in which the capsaicin was shown to exert an effect.
                                                                                                                                                                                                                                           FIG. 1. (Upper) C fiber activity recorded in situ in rats from single sural nerve fibers, identified by their conduction velocity and
                                                                                                                                                                                                                                           modality as C fibers. Immediately after formalin injection (as indicated by the dashed line) into their receptive fields, high activity
                                                                                                                                                                                                                                           was observed in high-threshold C nociceptive afferent fibers (as well as in A beta and A delta fibers, data not shown). At later
                                                                                                                                                                                                                                           intervals, activity was observed in all mechanically sensitive C fibers, at rates that were less (1/22/3) than those achieved initially
                                                                                                                                                                                                                                           (adapted from ref. 6 ). (Lower) Frequency of flinching as measured by an automated motion detector is plotted at 5-min intervals after
                                                                                                                                                                                                                                           the injection of formalin into the paw at the time indicated by the vertical dashed line. As indicated, the flinching behavior displays a
                                                                                                                                                                                                                                           biphasic occurrence (phase 1 and phase 2). The data represent the mean  SEM of eight rats.
                                                                                                                                                                                                                                                          ROLE OF SPINAL AND PERIPHERAL SYSTEMS IN THE POST-TISSUE INJURY PAIN STATE
                                                                                                                                                                                                                                               The behavioral sequelae outlined above, showing a hyperalgesic/allodynic state after tissue injury, may result from a peripheral
                                                                                                                                                                                                                                         sensitization secondary to the injury and/or to a change in central processing initiated by the persistent small afferent input generated by
                                                                                                                                                                                                                                         the injury. Blockade of spinal activation by the spinal delivery of a local anesthetic ( 11 ) or a selective blockade of small afferent input by
                                                                                                                                                                                                                                         the intrathecal infusion of a short-lasting opiate during the initial period of injury ( 12 ) will attenuate the second phase of the formalin
                                                                                                                                                                                                                                         response and abolish the 2 tactile allodynia, but not the 1 hyperalgesia observed secondary to a mild thermal injury (N.N.-T. and T.L.Y.,
                                                                                                                                                                                                                                         unpublished observations). Importantly, as the pain behavior observed during the second phase after formalin injection is blocked by the
                                                                                                                                                                                                                                         injection of local anesthetic into the paw ( 13 ), it is clear that the exaggerated responding indeed depends on the concurrent low-level
                                                                                                                                                                                                                                         afferent input observed during the second phase of the formalin test (see Fig. 1 ). These findings thus support the hypothesis that (i) the
                                                                                                                                                                                                                                         initial injury-induced afferent barrage generated in an opiatesensitive spinal system initiates a cascade that supports the 2 allodynia
                                                                                                                                                                                                                                         observed after injury, and (ii) the 1 hyperalgesia is mediated in part by a peripheral sensitization of small opiatesensitive C fibers.
                                                                                                                                                                                                                                           FIG. 2. Time course of change in mechanical threshold necessary to evoke acute withdrawal (Upper) or the thermal escape latency to
                                                                                                                                                                                                                                           evoke withdrawal (Lower) in the normal (noninjured) and injured paw. The injury was induced with the exposure of the shaded area
                                                                                                                                                                                                                                           indicated in the paw diagram (Left) to a 52C thermal stimulus applied for 45 sec at the time indicated in the graphs by the vertical
                                                                                                                                                                                                                                           dashed line. As indicated, the test stimuli were applied to the sites as indicated. Only a modest change in tactile thresholds were
                                                                                                                                                                                                                                           observed at the injury site, and no change in thermal escape thresholds were noted in the off injury site (data not shown). Hence the
                                                                                                                                                                                                                                           lower response latency corresponds to a 1 thermal hyperalgesia and 2 tactile allodynia. Contralateral paws showed no systematic
                                                                                                                                                                                                                                           change. Mechanical thresholds were determined with Von Frey hairs, and the thermal escape thresholds were assessed with
                                                                                                                                                                                                                                           Hargreaves apparatus ( 10 ). All points represent mean  SEM of five animals. B, baseline threshold.
the authoritative version for attribution.
                                                                                                                                                                                                                                         through an effect mediated by the tachykinin neurokinin 1 (NK-1) and the glutamatergic -amino-3-hydroxy-5-
                                                                                                                                                                                                                                         methylisoxazole-4propionic acid (AMPA)/N-methyl-D-aspartate (NMDA) receptors, respectively. In situ hybridization shows labeling for
                                                                                                                                                                                                                                         NK-1 and NMDA receptor units in the dorsal gray matter, particularly in the substantia gelatinosa where small afferents are known to
                                                                                                                                                                                                                                         terminate, (iv) Electrophysiologically, NK-1 and AMPA receptor antagonists will diminish small afferentevoked excitation. NMDA
                                                                                                                                                                                                                                         antagonists do not appear to reduce monosynaptically mediated afferent-evoked excitation and thus are not believed to be immediately
                                                                                                                                                                                                                                         postsynaptic to the primary afferent terminal, though some binding may be on the C-fiber terminal itself (see refs. 5 and 14 for references).
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                           FIG. 3. Schematic summarizes the organization of several dorsal horn systems that contribute to the processing of nociceptive
                                                                                                                                                                                                                                           information. Primary afferent C fibers release peptide (e.g., sP/CGRP, etc.) and excitatory amino acid (glutamate) products. Small
                                                                                                                                                                                                                                           DRG as well as some postsynaptic elements contain NOS) and are able on depolarization to release NO. Peptides and excitatory
                                                                                                                                                                                                                                           amino acids evoke excitation in second-order neurons. For glutamate, direct monosynaptic excitation is mediated by non-NMDA
                                                                                                                                                                                                                                           receptors (i.e., acute primary afferent excitation of WDR neurons is not mediated by the NMDA or NK-1 receptor). Interneurons
                                                                                                                                                                                                                                           excited by afferent barrage induce excitation in second-order neuron via a NMDA receptor, which leads to an increase in intracellular
                                                                                                                                                                                                                                           Ca2+, activation of phospholipase A2, NOS, and phosphorylating enzymes. COX products (PG) and NO are formed and released.
                                                                                                                                                                                                                                           These agents diffuse extracellularly and facilitate transmitter release (retrograde transmission) from primary and nonprimary afferent
                                                                                                                                                                                                                                           terminals by either a direct cellular action (e.g., NO) or by an interaction with a specific class of receptors [e.g., PG type E (EP)
                                                                                                                                                                                                                                           receptors for prostanoids]. Phosphorylation of intracellular protein (e.g., enzymes and receptors such as NMDA) leads to additional
                                                                                                                                                                                                                                           enhanced sensitivity. See text for other details.
                                                                                                                                                                                                                                              sP. The spinal delivery of NK-1 receptor agonists results in a mild acute pain behavior and a subsequent reduced response latency
                                                                                                                                                                                                                                         to thermal stimuli (thermal hyperalgesia). Blockade of the NK-1 receptor by intrathecal antagonists ( 19 ) or down-regulation of NK-1
                                                                                                                                                                                                                                         receptor expression by intrathecal treatment with NK-1 receptor mRNA antisense ( 20 ) has no effect on acute nociceptive thresholds, but
                                                                                                                                                                                                                                         reduces the second phase of the formalin response. Intrathecal injection of NK-1 antagonists after phase 1 reduces their effect on the
                                                                                                                                                                                                                                         second phase ( 19 ).
                                                                                                                                                                                                                                              Glutamate Receptors. Repetitive small afferent input (as that which occurs after tissue injury) will evoke spinal glutamate release
                                                                                                                                                                                                                                         (see Fig. 4 ) ( 21 , 22 ). The spinal delivery of agonists for the ionotrophic glutamate receptors (NMDA/ AMPA) will evoke a potent
                                                                                                                                                                                                                                         spontaneous pain behavior and a subsequent thermal hyperalgesia and tactile allodynia ( 23 ).
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         Blockade of spinal AMPA receptors by intrathecal antagonists ( 24 ) will elevate acute nociceptive thresholds, as well as the first and
                                                                                                                                                                                                                                         second phase of the formalin test. In contrast, intrathecal NMDA antagonists have little effect on acute nociception, but diminish the
                                                                                                                                                                                                                                         second phase of the formalin test ( 25 ). As with the NK-1 antagonists, NMDA antagonists in the formalin model show a diminished effect
                                                                                                                                                                                                                                         on phase two when delivered after phase 1 ( 26 ), which reflects the fact that after an acute injurious stimulus, such as with formalin
                                                                                                                                                                                                                                         injection, there is an initiating barrage of activity leading to transmitter release (see Fig. 4 ). This release, for example of glutamate, sP, and
                                                                                                                                                                                                                                         prostanoids, leads to biochemical changes within the spinal cord that must persist after the initial occupancy of the NMDA (or NK-1
                                                                                                                                                                                                                                         receptor) receptor during phase 1 has passed.
                                                                                                                                                                                                                                               Prostaglandins (PGs). PGs are released in vivo from the spinal cord by a peripherally injurious stimulus that is associated with small
                                                                                                                                                                                                                                         afferent activation (see Fig. 4 ) ( 28 ) and by the direct spinal delivery of NK-1 and glutamate receptor agonists ( 22 , 29 ). Prostanoid
                                                                                                                                                                                                                                         receptors are present in the dorsal horn and on DRG ( 30 ). Activation of prostanoid receptors has been shown to increase the opening of
                                                                                                                                                                                                                                         voltage-sensitive Ca channels and to enhance primary afferent peptide release ( 31 ). Consistent with these events, intrathecal PG receptor
                                                                                                                                                                                                                                         agonists will evoke a hyperalgesia and allodynia ( 32 ). The spinal delivery of cyclooxygenase (COX) inhibitors or antagonists has no
                                                                                                                                                                                                                                         effect on acute nociceptive processing, but will reduce the phase 2 of the formalin test at doses that are inactive when given systemically (
                                                                                                                                                                                                                                         33 , 34 ). Consistent with the observation that NMDA/ NK-1 antagonists can block certain hyperalgesic states, spinal NMDA agonists can
                                                                                                                                                                                                                                         evoke a hyperalgesic state and this hyperalgesia also can be blocked by spinal COX inhibitors ( 23 ). Several COX isozymes have been
                                                                                                                                                                                                                                         identified ( 35 ). Current evidence indicates that COX-2 is constitutive in the spinal cord (but only in a few peripheral organs). The
                                                                                                                                                                                                                                         intrathecal delivery of selective COX inhibitors suggests that the hyperalgesia induced by intrathecal NK1/NMDA is mediated in part by
                                                                                                                                                                                                                                         the local release of PGs. In recent work, we have shown that spinal COX2 inhibitors are also effective in models of inflammatory
                                                                                                                                                                                                                                         hyperalgesia (ref. 36 and D. M. Dirig and T.L.Y., unpublished observations).
                                                                                                                                                                                                                                           FIG. 4. Time course of flinching behavior (Top) and concurrent assessment of lumbar spinal glutamate (Middle), and PGs E2
                                                                                                                                                                                                                                           (Bottom) release measured in unanesthetized rats before and after the injection of formalin (5%/50 l) into the left hind paw of the rat
                                                                                                                                                                                                                                           at time = 0. At 10 min, the rats received an intrathecal (IT) injection of saline (vehicle: n = 4) or morphine (10 g; n = 7). Release
                                                                                                                                                                                                                                           was assessed by using a chronically implanted loop dialysis probe ( 27 ), and the intrathecal injection was through a chronic
                                                                                                                                                                                                                                           intrathecal catheter. Each line presents the mean of four and seven rats, respectively. Error bars deleted for clarity. *, P < 0.05 versus
                                                                                                                                                                                                                                           formalin; #, P < 0.05 vs. IT saline.
                                                                                                                                                                                                                                               NO. NO is synthesized by NO synthase (NOS). Evidence of in vivo NO release from cord secondary to repetitive afferent stimulation
                                                                                                                                                                                                                                         and by intrathecal NMDA has been presented ( 21 ). The hyperalgesia induced by intrathecal NMDA and the second phase of the formalin
                                                                                                                                                                                                                                         test has been shown to be reduced by intrathecal competitive NOS inhibitors ( 37 ).
                                                                                                                                                                                                                                               Kinases and Phosphorylation. Increasing intracellular Ca2+I, though the inosital triphosphate pathway by activation of NK receptor
                                                                                                                                                                                                                                         and/or by the influx of Ca2+ through voltagegated Ca2+ channels or ionophores (NMDA receptor) ( 38 ), activates kinases that
                                                                                                                                                                                                                                         phosphorylate and phosphatases, which dephosphorylate local proteins. Phosphorylating enzyme systems consist of several classes of
                                                                                                                                                                                                                                         kinases that are distinguished by structure and the pharmacology of their inhibitors. In the spinal dorsal horn, cAMP-dependent kinase (
                                                                                                                                                                                                                                         39 ) and camkinase II ( 40 ) have been observed in the spinal dorsal horn and DRG. Protein kinase (PK) C consists of a large family of
                                                                                                                                                                                                                                         isozymes. In the spinal cord PKC ( 41 ) and PKC ( 42 ) are limited to the spinal gray, with PKC being reported to be largely in cells in
                                                                                                                                                                                                                                         lamina I and II inner of the dorsal horn. Although it is possible that any or all of the phosphorylating enzymes noted above may play a
                                                                                                                                                                                                                                         role, the use of inhibitors for PKA and PKC have shown the particular importance of this family of kinases in regulating spinal facilitation.
the authoritative version for attribution.
                                                                                                                                                                                                                                         Many hyperalgesic states are mediated by a spinal NMDA receptor. The NMDA receptor is multiply phosphorylated by PKA and PKC (
                                                                                                                                                                                                                                         43 ). Intrathecal delivery of PKC inhibitors has been shown to stereospecifically diminish the hyperalgesic effects of intrathecal NMDA.
                                                                                                                                                                                                                                         In addition, the augmented activity in dorsal horn neurons after intradermal mustard oil ( 44 , 45 ) or spinal NMDA ( 46 ) is reduced by the
                                                                                                                                                                                                                                         local spinal delivery of PKC inhibitors. PKA and PKC inhibitors, but not inactive isomers, will diminish capsaicin-evoked hyperpathia (
                                                                                                                                                                                                                                         45 ) and the second phase of the formalin test ( 47 , 48 ).
                                                                                                                                                                                                                                                                                                  SYSTEM INTERACTIONS
                                                                                                                                                                                                                                              As reviewed above, after local injury, the behaviorally defined components of post-tissue injury pain states reflect an increased
                                                                                                                                                                                                                                         receptive field and a left shift in the stimulus response curve for spinal dorsal horn neurons, which is evoked initially and then sustained in
                                                                                                                                                                                                                                         part by persistent small afferent input. The contribution of these changes in spinal function to the behaviorally relevant nociceptive state is
                                                                                                                                                                                                                                         substantiated by comparing the pharmacology associated with the effects on the behavior of the unanesthetized animal with the effects of
                                                                                                                                                                                                                                         the drugs on the underlying electrophysiology. Table 1 provides a summary of the effects of several classes of agonists and
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         antagonists given intrathecally in rats on acute pain behavior (as measured by thermal escape) and facilitated processing (as defined by
                                                                                                                                                                                                                                         their effects on the second phase of the formalin test). Based on such observations, it is possible to formulate a heuristic picture of the
                                                                                                                                                                                                                                         organization of several pharmacologically defined spinal systems that mediate the response of the animal to a strong and injurious
                                                                                                                                                                                                                                         stimulus. Thus, repetitive afferent input increases excitatory amino acid and peptide release from primary afferents that serve to initially
                                                                                                                                                                                                                                         depolarize dorsal horn neurons. Persistent depolarization serves to increase intracellular calcium, activating a variety of intracellular
                                                                                                                                                                                                                                         enzymes (COX-2 and NOS) and various kinases (PKC). PGs and NO are released spinally and serve to acutely enhance the subsequent
                                                                                                                                                                                                                                         release of afferent peptides and glutamate. Activation of local kinases serves to phosphorylate membrane receptors and channels. As an
                                                                                                                                                                                                                                         example, the NMDA receptor when phosphorylated displays an enhanced calcium flux (see Fig. 3 ). The role of these system-level
                                                                                                                                                                                                                                         changes in spinal nociceptive processing in pain behavior is supported by the analgesic effects of spinally delivered agents known to
                                                                                                                                                                                                                                         reduce small afferent transmitter release (, opioid, and 2 adrenergic agonists) and the antihyperalgesic actions of spinally delivered
                                                                                                                                                                                                                                         NK-1 and NMDA receptor antagonists, as well as inhibitors of spinal COX-2, NOS, and PKC.
                                                                                                                                                                                                                                         Table 1. Spinal drug action in nociceptive processing in animals models and human pain states
                                                                                                                                                                                                                                         Agonists                        Rat acute thermal escape        Rat phase 2 formalin      Human pain states               Reference
                                                                                                                                                                                                                                          agonist                       +                               +                         Morphine                        (16)
                                                                                                                                                                                                                                           agonist                       +                               +                         DADL                            (16, 49)
                                                                                                                                                                                                                                         2 agonist                      +                               +                         Clonidine                       (17, 50)
                                                                                                                                                                                                                                         Aden A-1 agonist                +/                             +                         R-PIA/adenosine                 (5153)
                                                                                                                                                                                                                                         GABA-A/B agonist                +/                             +                         Baclofen*                       (54, 55)
                                                                                                                                                                                                                                         GABAPENTIN                      0                               +                                                         (56)
                                                                                                                                                                                                                                         NMDA antagonist                 0                               +                         Ketamine/CPP                    (25, 57, 58)
                                                                                                                                                                                                                                         AMPA antagonist                 +                               +                                                         (24, 59)
                                                                                                                                                                                                                                         Metab Glu-antagonist            +/                             +                                                         (60)
                                                                                                                                                                                                                                         NK-1 agonist                    0                               +                                                         (20)
                                                                                                                                                                                                                                         COX inhibitor                   0                               +                         Lysine acetylsalicylate         (23, 61)
                                                                                                                                                                                                                                         EP-antagonist                   0                               +                                                         (34)
                                                                                                                                                                                                                                         NOS inhibitor                   0                               +                                                         (37)
                                                                                                                                                                                                                                         AChase inhibitor (mus)          +                               +                         Neostigmine                     (6266)
                                                                                                                                                                                                                                         N-Ca Ch blocker                 0                               +                         Ziconotide (SNX-111)            (67, 68)
                                                                                                                                                                                                                                         GABA, -aminobutyric acid; DADL, d-Ala2-d-Leu5-encephalin; R-PIA, R(-)N6-(2-phenylisopropyl) adenosine; CPP, ()-3-(2-
                                                                                                                                                                                                                                         carboxypiperazin-4-yl)-propyl-1-phosphonic acid; EP, PG type E.
                                                                                                                                                                                                                                         *Used intrathecally for spasticity.
                                                                                                                                                                                                                                              Several additional points should be considered in interpreting these data. The above comments are limited to several specific
                                                                                                                                                                                                                                         components of dorsal horn biology. Other systems that no doubt play an important role in spinal nociceptive processing, such as the
                                                                                                                                                                                                                                         purinergic receptors ( 69 ) and the metabotrophic glutamate receptors ( 70 ) are not considered. In each case, the effects of manipulations
                                                                                                                                                                                                                                         associated with a single system, e.g., NK-1, glutamate, or AMPA, are considered. In the case
                                                                                                                                                                                                                                           FIG. 5. Graph plots the relative intrathecal potency of several opioids as defined in rats on the hot plate test versus the relative
                                                                                                                                                                                                                                           potency when given epidurally (EP) or intrathecally (IT) in human postoperative or cancer pain. The axis plot the potency of the
                                                                                                                                                                                                                                           agent (in g for rat or mg in human) relative to the potency of morphine given in that model. These calculations are based on a
                                                                                                                                                                                                                                           standard analgesic dose for intrathecal morphine in rats on the hot plate (10 g) and after intrathecal (0.5 mg) or epidural (3 mg)
                                                                                                                                                                                                                                           delivery for pain in humans. Human data are based on reported doses necessary to produce an adequate clinical analgesia (data
                                                                                                                                                                                                                                           derived from ref. 16 ). DADL, d-Ala2-d-Leu5-encephalin.
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         of the primary afferent, terminals are known to routinely contain and likely release combinations of amino acids (glutamate), peptides
                                                                                                                                                                                                                                         (CGRP, sP, vasoactive intestinal polypeptide; ref. 71 ) and peptides (such as growth factors, ref. 72 ). The net combination of these drug
                                                                                                                                                                                                                                         effects are poorly studied. In the case of sP and NMDA, both contribute to the postsynaptic action ( 73 ). This observed excitation is
                                                                                                                                                                                                                                         consistent with (i) the ability of either agent to activate the second-order neurons, (ii) a depolarization by sP serving to remove the Mg2+
                                                                                                                                                                                                                                         block, and (iii) sP activating by local kinases to phosphorylate the NMDA channel. Agents that block the opening of Ca2+ channels in
                                                                                                                                                                                                                                         primary afferents likely block all transmitter release from that terminal, which accounts for the potent antinociception that is produced by
                                                                                                                                                                                                                                         these agents in contrast to that produced by antagonists for specific receptors (e.g., NK-1 or glutamate). The present comments focus
                                                                                                                                                                                                                                         primarily on the events that occur in the interval around the injury period. Over extended intervals of hours to days there is an up-
                                                                                                                                                                                                                                         regulation of receptors (NK-1) ( 74 ) and enzymes [COX ( 75 ) and NOS ( 76 )], leading to additional changes in system function. The
                                                                                                                                                                                                                                         evidence presented here clearly reflects the functional complexity of the events that occur secondary to a focal injury, leading to a
                                                                                                                                                                                                                                         persistent small afferent barrage. The fact that such stimuli will lead to a local 1 hyperalgesia and 2 tactile allodynia raises the likelihood
                                                                                                                                                                                                                                         that specific components of the post-tissue injury pain state may have distinct components. Thus, as noted, after a mild, local tissue injury
                                                                                                                                                                                                                                         a 1 hyperalgesia and a 2 tactile allodynia are noted. Treatment with spinal opiates during the injury phase will prevent the appearance of
                                                                                                                                                                                                                                         the allodynia, but not the hyperalgesia. This finding suggests that the allodynia after an acute injury depends on a cascade that is initiated,
                                                                                                                                                                                                                                         but not sustained, by the injury stimulus. In contrast, the hyperalgesia does not appear to depend on that cascade to be made manifest. Such
                                                                                                                                                                                                                                         differences may reflect on the clinical phenomena of preemptive analgesia ( 77 ). In pre-emptive analgesia the patient receiving opiates
                                                                                                                                                                                                                                         during the surgery is hypothesized to require less analgesic postoperatively. To the extent that the postoperative pain state reflects the
                                                                                                                                                                                                                                         allodynic component noted here, that would indeed be true. To the extent that the postoperative state involves a hyperalgesic mechanisms,
                                                                                                                                                                                                                                         the differences produced by intraoperative opiates might be slight. Finally, the early discussions on the events that occur during the periods
                                                                                                                                                                                                                                         immediately after injury focused on the phenomena as if it were a unitary phenomena. The initial observations, for example, that
                                                                                                                                                                                                                                         demonstrated that dorsal horn wind-up and several inflammatory models all were diminished by spinal NMDA receptor antagonists
                                                                                                                                                                                                                                         supported such homogeneity. It is now clear that variations in mechanism can be defined even with acute injury stimulus conditions. Thus,
                                                                                                                                                                                                                                         on examining the allodynia observed after the healing of a skin incision ( 59 ) or the hyperalgesia induced by a local burn (N.N.-T. and
                                                                                                                                                                                                                                         T.L.Y., unpublished observations), the hyperpathia was noted to be poorly diminished by NMDA receptor antagonists.
                                                                                                                                                                                                                                                                                              HUMAN SPINAL PROCESSING
                                                                                                                                                                                                                                               Although the above work is of importance in defining the biology of spinal processing in the mechanistic sense, such preclinical
                                                                                                                                                                                                                                         insights also appear to be relevant to our understanding of spinal system function in humans. Two points can be made: (i) comparability of
                                                                                                                                                                                                                                         the behavioral components and (ii) parallels in pharmacological activity.
                                                                                                                                                                                                                                               Comparability of Behavioral Pain Components in Humans and Animal Models. As in the preclinical models, after focal tissue
                                                                                                                                                                                                                                         injury (whether experimental or pathological) in humans, there is a clearly defined 1 hyperalgesia and 2 off-site tactile allodynia ( 78 )
                                                                                                                                                                                                                                         Though as yet poorly studied, it is clear that the postoperative or postinjury pain state in humans possess the same complexity (see refs. 79
                                                                                                                                                                                                                                         and 80 ). Still, the typical postoperative pain evaluation typically is limited to a univariate assessment (e.g., visual analogue score or
                                                                                                                                                                                                                                         postoperative narcotic consumption). Although clinically practical, such limited surveys may well obscure the benefits or actions of a drug
                                                                                                                                                                                                                                         that influences one of the components of the pain state.
                                                                                                                                                                                                                                               Comparability of Spinal Pharmacology in Humans and Animal Models. The pharmacology and activity of drug effects at the
                                                                                                                                                                                                                                         spinal level as defined in rodent systems have been shown to be extraordinarily predictive of the activity in human pain states. The best
                                                                                                                                                                                                                                         evaluated pharmacology is that of the opiates that have been widely examined in both humans and animals. As presented in Fig. 5 ,
                                                                                                                                                                                                                                         plotting the spinal potency of such agents relative to morphine in rats (intrathecal) and humans (epidural or intrathecal) reveals a high
                                                                                                                                                                                                                                         correlation. More importantly, a variety of nonopioid agents have been delivered intrathecally or epidurally in animal models and then in
                                                                                                                                                                                                                                         humans. Table 1 summarizes such work in which humans have received the respective novel class of agent. Importantly, it should be
                                                                                                                                                                                                                                         noted that agents, which, unlike opiates, have little effect on acute pain behavior (e.g., thermal escape) are indeed active in human clinical
                                                                                                                                                                                                                                         pain states.
                                                                                                                                                                                                                                               These results jointly (i) support the importance of spinal processing in human pain states, (ii) emphasize the functional and
                                                                                                                                                                                                                                         pharmacological comparability of these systems across species, and (iii) provide an important source of targets for the development of
                                                                                                                                                                                                                                         novel clinically relevant analgesics processing can be expected to yield even greater rewards.
                                                                                                                                                                                                                                         1. Yaksh, T. L. , Lynch III, C , Zapol, W. M. , Maze, M. , Biebuyck, J.F. & Saidman, L. J. , eds. ( 1997 ) Anesthesia: Biologic Foundations ( Lippincott ,
                                                                                                                                                                                                                                              Philadelphia ), pp. 685718 .
                                                                                                                                                                                                                                         2. Dirig, D. M. & Yaksh, T. L. ( 1995 ) Pain 62 , 321328 .
                                                                                                                                                                                                                                         3. Handwerker, H. O. ( 1991 ) Agents Actions 32 , 9199 .
                                                                                                                                                                                                                                         4. Messlinger, K. ( 1997 ) Anaesthesist 46 , 142153 .
                                                                                                                                                                                                                                         5. Dickenson, A. H. , Stanfa, L. C. , Chapman, V. & Yaksh, T. L. ( 1997 ) in Anesthesia: Biologic Foundations , eds. Yaksh, T. L. , Lynch III, C. ,
                                                                                                                                                                                                                                              Zapol, W. M. , Maze, M. , Biebuyck, J. F. & Saidman, L. J. ( Lippincott , Philadelphia ), pp. 611624 .
                                                                                                                                                                                                                                         6. Puig, S. & Sorkin L. S. ( 1996 ) Pain 64 , 345355 .
                                                                                                                                                                                                                                         7. Dickenson, A. H. & Sullivan, A. F. ( 1987 ) Pain 30 , 349360 .
                                                                                                                                                                                                                                         8. Nozaki-Taguchi, N. & Yaksh, T. L. ( 1998 ) Neurosci. Lett. 254 , 2528 .
                                                                                                                                                                                                                                         9. Simone, D. A. , Sorkin, L. S. , Oh, U. , Chung, J. M. , Owens, C. , LaMotte, R. H. , Willis, W. D. ( 1991 ) J. Neurophysiol. 66 , 228246 .
                                                                                                                                                                                                                                         10. Dirig, D. M. , Salami, A. , Rathbun, M. L. , Ozaki, G. T. & Yaksh, T. L. ( 1997 ) J. Neurosci. Methods 76 , 183191 .
                                                                                                                                                                                                                                         11. Yashpal, K. , Katz, J. & Coderre, T. J. ( 1996 ) Anesthesiology 84 , 11191128 .
                                                                                                                                                                                                                                         12. Buerkle, H. , Marsala, M. & Yaksh, T. L. ( 1998 ) Br. J. Anaesth . 80 , 348353 .
                                                                                                                                                                                                                                         13. Taylor, B. K. , Peterson, M. A. & Basbaum, A. I. ( 1995 ) J. Neurosci. 15 , 75757584 .
                                                                                                                                                                                                                                         14. Wilcox, G. L. & Seybold, V. ( 1997 ) in Anesthesia: Biologic Foundations , eds. Yaksh, T. L. , Lynch III, C. , Zapol, W. M. , Maze, M. , Biebuyck,
                                                                                                                                                                                                                                              J. F. & Saidman, L. J. ( Lippincott , Philadelphia ), pp. 557576 .
                                                                                                                                                                                                                                         15. Yaksh, T. L. & Rudy, T. A. ( 1976 ) Physiol. Behav. 17 , 10311036 .
                                                                                                                                                                                                                                         16. Yaksh, T. L. ( 1993 ) in Handbook of Experimental Pharmacology , ed. Herz A. ( Springer , Berlin ), Vol. 104/II , pp. 5390 .
                                                                                                                                                                                                                                         17. Yaksh, T. L. , Jage, J. & Takano, Y. ( 1993 ) Baillires Clin. Anaesthesiol. 7 , 597614 .
                                                                                                                                                                                                                                         18. Hua, X.-Y , Boublik, J. H. , Spicer, M. A. , Rivier, J. E. , Brown, M. R. & Yaksh, T. L. ( 1991 ) J. Pharmacol. Exp. Ther. 258 , 243258 .
the authoritative version for attribution.
                                                                                                                                                                                                                                         25. Chaplan, S. R. , Malmberg, A. B. & Yaksh, T. L. ( 1997 ) J. Pharmacol. Exp. Ther. 280 , 829838 .
                                                                                                                                                                                                                                         26. Yamamoto, T. & Yaksh, T. L. ( 1992 ) Anesthesiology 77 , 757763 .
                                                                                                                                                                                                                                         27. Marsala, M. , Malmberg, A. B. & Yaksh, T. L. ( 1995 ) J. Neurosci. Methods 62 , 4353 .
                                                                                                                                                                                                                                         28. Malmberg, A. B. & Yaksh, T. L. ( 1995 ) J. Neurosci. 15 , 768776 .
                                                                                                                                                                                                                                         29. Yang, L. C , Marsala, M. & Yaksh, T. L. ( 1996 ) Neuroscience 75 , 453461 .
                                                                                                                                                                                                                                         30. Coleman, R. A. , Smith, W. L. & Narumiya, S. ( 1994 ) Pharmacol. Rev. 46 , 205229 .
                                                                                                                                                                                                                                         31. Hingtgen, C. M. & Vasko, M. R. ( 1994 ) Brain Res. 655 , 5160 .
                                                                                                                                                                                                                                         32. Minami, T. , Uda, R. , Horiguchi, S. , Ito, S. , Hyodo, M. & Hayaishi, O. ( 1994 ) Pain 57 , 217223 .
                                                                                                                                                                                                                                         33. Malmberg, A. B. & Yaksh, T. L. ( 1992 ) J. Pharmacol. Exp. Ther. 263 , 264275 .
                                                                                                                                                                                                                                         34. Malmberg, A. B. , Rafferty, M. F. & Yaksh, T. L. ( 1994 ) Neurosci. Lett. 173 , 193196 .
                                                                                                                                                                                                                                         35. Seibert, K. , Zhang, Y. , Leahy, K. , Hauser, S. , Masferrer, J. , Perkins, W. , Lee, L. & Isakson, P. ( 1994 ) Proc. Natl. Acad. Sci. USA 91 , 12013
                                                                                                                                                                                                                                              12017 .
                                                                                                                                                                                                                                         36. Yamamoto, T. & Nozaki-Taguchi, N. ( 1997 ) NeuroReport 8 , 21792182 .
                                                                                                                                                                                                                                         37. Malmberg, A. B. & Yaksh, T. L. ( 1993 ) Pain 54 , 291300 .
                                                                                                                                                                                                                                         38. Ghosh, A. & Greenberg, M. E. ( 1995 ) Science 268 , 239247 .
                                                                                                                                                                                                                                         39. Malmberg, A. B. , Brandon, E. P. , Idzerda, R. L. , Liu, H. , McKnight, G. S. & Basbaum, A. I. ( 1997 ) J. Neurosci. 17 , 74627470 .
                                                                                                                                                                                                                                         40. Kocsis, J. D. , Rand, M. N. , Lankford, K. L. & Waxman, S. G. ( 1994 ) J. Neurobiol. 25 , 252264 .
                                                                                                                                                                                                                                         41. Roberts, R. E. & McLean, W. G. ( 1997 ) Brain Res. 754 , 147156 .
                                                                                                                                                                                                                                         42. Malmberg, A. B. , Chen, C. , Tonegawa, S. & Basbaum, A. I. ( 1997 ) Science 278 , 279283 .
                                                                                                                                                                                                                                         43. Leonard, A. S. & Hell, J. W. ( 1997 ) J. Biol. Chem. 272 , 12107 12115 .
                                                                                                                                                                                                                                         44. Munro, F. E. , Fleetwood-Walker, S. M. & Mitchell, R. ( 1994 ) Neurosci. Lett. 170 , 199202 .
                                                                                                                                                                                                                                         45. Sluka, K. A. & Willis, W. D. ( 1997 ) Pain 71 , 165178 .
                                                                                                                                                                                                                                         46. Cerne, R. , Rusin, K. I. & Randic, M. ( 1993 ) Neurosci. Lett. 161 , 124128 .
                                                                                                                                                                                                                                         47. Coderre, T. J. & Yashpal, K. ( 1994 ) Eur. J. Neurosci. 6 , 1328 1334 .
                                                                                                                                                                                                                                         48. Yashpal, K. , Pitcher, G. M. , Parent, A. , Quirion, R. & Coderre, T. J. ( 1995 ) J. Neurosci. 15 , 32633272 .
                                                                                                                                                                                                                                         49. Onofrio, B. M. & Yaksh, T. L. ( 1983 ) Lancet 1 , 13861387 .
                                                                                                                                                                                                                                         50. Eisenach, J. C. , De Kock, M. & Klimscha, W. ( 1996 ) Anesthesiology 85 , 655674 .
                                                                                                                                                                                                                                         51. Poon, A & Sawynok, J. ( 1998 ) Pain 74 , 235245 .
                                                                                                                                                                                                                                         52. Karlsten, R. & Gordh, T. , Jr. ( 1995 ) Anesth. Analg. 80 , 844847 .
                                                                                                                                                                                                                                         53. Rane, K. , Segerdahl, M. , Goiny, M. & Sollevi, A. ( 1998 ) Anesthesiology 5 , 11081115 .
                                                                                                                                                                                                                                         54. Dirig, D. M. & Yaksh, T. L. ( 1995 ) Pharmacol. Exp. Ther. 275 , 219227 .
                                                                                                                                                                                                                                         55. Porter, B. ( 1997 ) Br. J. Nursing 6 , 253260 .
                                                                                                                                                                                                                                         56. Partridge, B. J. , Chaplan, S. R. , Sakamoto, E. & Yaksh, T. L. ( 1998 ) Anesthesiology 88 , 196205 .
                                                                                                                                                                                                                                         57. Abdel-Ghaffar, M. E. , Abdulatif, M. A. , al-Ghamdi, A. , Mowafi, H. & Anwar, A. ( 1998 ) Can. J. Anaes. 45 , 103109 .
                                                                                                                                                                                                                                         58. Kristensen, J. D. , Svensson, B. & Gordh, T. , Jr. ( 1992 ) Pain 51 , 249253 .
                                                                                                                                                                                                                                         59. Brennan, T. J. ( 1998 ) Anesthesiology 89 , 10491051 .
                                                                                                                                                                                                                                         60. Fisher, K. & Coderre, T. J. ( 1996 ) Pain 68 , 255263 .
                                                                                                                                                                                                                                         61. Devoghel, J. C. ( 1983 ) J. Int. Med. Res. 11 , 9091 .
                                                                                                                                                                                                                                         62. Prado, W. A. & Goncalves, A. S. ( 1997 ) Braz. J. Med. Biol. Res. 30 , 12251231 .
                                                                                                                                                                                                                                         63. Naguib, M. & Yaksh, T. L. ( 1994 ) Anesthesiology 80 , 13381348 .
                                                                                                                                                                                                                                         64. Naguib, M. & Yaksh, T. L. ( 1997 ) Anesth. Analg. 85 , 847853 .
                                                                                                                                                                                                                                         65. Hood, D. D. , Eisenach, J. C. & Tuttle, R. ( 1995 ) Anesthesiology 82 , 331343 .
                                                                                                                                                                                                                                         66. Lauretti, G. R. & Lima, I. C. ( 1996 ) Anest. Analg. 82 , 617620 .
                                                                                                                                                                                                                                         67. Malmberg, A. B. & Yaksh, T. L. ( 1994 ) J. Neurosci. 14 , 4882 4890 .
                                                                                                                                                                                                                                         68. Brose, W. G. , Gutlove, D. P. , Luther, R. R. , Bowersox, S. S. & McGuire, D. ( 1997 ) Clin. J. Pain 13 , 256259 .
                                                                                                                                                                                                                                         69. Driessen, B. , Reimann, W. , Selve, N. , Friderichs, E. & Bultmann, R. ( 1994 ) Brain Res. 666 , 182188 .
                                                                                                                                                                                                                                         70. Young, M. R. , Blackburn-Munro, G. , Dickinson, T. , Johnson, M. J. , Anderson, H. , Nakalembe, I. & Fleetwood-Walker, S. M. ( 1998 ) J.
                                                                                                                                                                                                                                              Neurosci. 18 , 1018010188 .
                                                                                                                                                                                                                                         71. Levine, J. D. , Fields, H. L. & Basbaum, A. I. ( 1993 ) J. Neurosci. 13 , 22732286 .
                                                                                                                                                                                                                                         72. Michael, G. J. , Averill, S. , Nitkunan, A. , Rattray, M. , Bennett, D. L. , Yan, Q. & Priestley, J. V. ( 1997 ) J. Neurosci. 17 , 84768490 .
                                                                                                                                                                                                                                         73. Chapman, V. , Buritova, J. , Honore, P. & Besson, J. M. ( 1996 ) J. Neurophysiol. 76 , 18171827 .
                                                                                                                                                                                                                                         74. Kar, S. , Rees, R. G. & Quirion, R. ( 1994 ) Eur. J. Neurosci. 6 , 345354 .
                                                                                                                                                                                                                                         75. Beiche, F. , Scheuerer, S. , Brune, K. , Geisslinger, G. & Goppelt-Struebe, M. ( 1996 ) FEBS Lett. 390 , 165169 .
                                                                                                                                                                                                                                         76. Wu, J. , Lin, Q , Lu, Y. , Willis, W. D. & Westlund, K. N. ( 1998 ) Exp. Brain Res. 118 , 457465 .
                                                                                                                                                                                                                                         77. McQuay, H. J. ( 1995 ) Ann. Med. 27 , 249256 .
                                                                                                                                                                                                                                         78. Handwerker, H. O. & Kobal, G. ( 1993 ) Physiol. Rev. 73 , 639671 .
                                                                                                                                                                                                                                         79. OConnor, T. C. & Abram, S. E. ( 1997 ) in Anesthesia: Biologic Foundations, eds. Yaksh, T. L. , Lynch III, C. , Zapol, W. M. , Maze, M. ,
                                                                                                                                                                                                                                              Biebuyck, J. F. & Saidman, L. J. ( Lippincott , Philadelphia ), pp. 747758 .
                                                                                                                                                                                                                                         80. Silbert, B. S. , Osgoodm P. F. & Carr, D. B. ( 1997 ) in Anesthesia: Biologic Foundations , eds. Yaksh, T. L. , Lynch III, C. , Zapol, W. M. , Maze,
                                                                                                                                                                                                                                              M. , Biebuyck, J. F. & Saidman, L. J. ( Lippincott , Philadelphia ), pp. 759773 .
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                         display an abrupt pause in ongoing activity immediately before nociceptive reflexes and are proposed to contribute to inhibitory influences
                                                                                                                                                                                                                                         that descend from the RVM. On-cells display a burst of activity immediately before nociceptive reflexes and are proposed to contribute to
                                                                                                                                                                                                                                         facilitatory influences that descend from the RVM. Neutral cells show no nociception-related change in activity. Off-cells, on-cells, and
                                                                                                                                                                                                                                         neutral cells all project to the spinal dorsal horn ( 16 ), placing on-cell and off-cell terminals in appropriate laminae (I, II, and V) to
                                                                                                                                                                                                                                         modulate nociceptive transmission. That on- and off-cells mediate descending facilitatory and inhibitory influences from the RVM is
                                                                                                                                                                                                                                         supported by several reports demonstrating enhanced on- or off-cell activity during facilitation or inhibition of spinal nociceptive
                                                                                                                                                                                                                                         transmission, respectively ( 17  19 ).
                                                                                                                                                                                                                                               We hypothesize that there exists a spinobulbospinal circuit that contributes significantly to central sensitization and secondary
                                                                                                                                                                                                                                         hyperalgesia. Anatomically, this circuit is in place. Both the RVM and adjacent areas receive direct afferent input from the superficial
                                                                                                                                                                                                                                         spinal dorsal horn and in turn send descending projections through spinal funiculi that terminate in the superficial dorsal horn, completing
                                                                                                                                                                                                                                         a spinobulbospinal loop ( 20  23 ). We review below recent studies that document that spinal transection, or inactivation of supraspinal
                                                                                                                                                                                                                                         sites, prevents the expression of secondary hyperalgesia in a variety of animal models of persistent inflammatory, neurogenic, or
                                                                                                                                                                                                                                         neuropathic pain, thus providing the functional context in support of the anatomy (see Table 1 ).
                                                                                                                                                                                                                                               Inflammatory/Neurogenic Models of Hyperalgesia. Mustard oil. Mustard oil (allyl isothiocyanate) is a chemical irritant that
                                                                                                                                                                                                                                         produces a neurogenic inflammation and excites chemosensitive C-fibers, resulting in behavioral hyperalgesia and central sensitization (
                                                                                                                                                                                                                                         24 , 25 ). An involvement of supraspinal sites in mustard oil-induced sensitization was reported by Mansikka and Pertovaara ( 26 ), who
                                                                                                                                                                                                                                         found that tactile allodynia of the glabrous skin of the foot after topical application of mustard oil to the ankle was prevented in animals
                                                                                                                                                                                                                                         that had received spinal transection. Additionally, in spinally intact rats, the tactile allodynia was blocked after inactivation of the medial
                                                                                                                                                                                                                                         RVM by local lidocaine microinjection. The authors concluded that persistent nociceptor stimulation by topical mustard oil activates a
                                                                                                                                                                                                                                         positive feedback loop involving descending facilitatory influences from the RVM. In an electrophysiological study of spinal cord
                                                                                                                                                                                                                                         neurons, Pertovaara ( 27 ) subsequently reported that midthoracic spinal transection or lidocaine inactivation of the RVM blocked mustard
                                                                                                                                                                                                                                         oil-induced enhanced excitability of wide dynamic range neurons to mechanical stimulation. In these experiments, mustard oil was applied
                                                                                                                                                                                                                                         12 cm outside the border of the receptive field of the spinal neuron. Thus, in both studies, the allodynia/ hyperalgesia was tested at a site
                                                                                                                                                                                                                                         distant from the site of application of mustard oil (i.e., it was secondary in nature).
                                                                                                                                                                                                                                         Table 1. Summary of supraspinal contributions to hyperalgesia
                                                                                                                                                                                                                                         Model of hyperalgesia                  Nociceptive response                    Manipulation                           Effect        Ref.
                                                                                                                                                                                                                                         Inflammation/neurogenic
                                                                                                                                                                                                                                         Mustard oil (ankle)                    Tactile allodynia, foot                 Spinal transection Intra-RVM           Block         26
                                                                                                                                                                                                                                                                                                                        lidocaine
                                                                                                                                                                                                                                         Mustard oil (foot, outside receptive     Enhanced excitability of WDR          Spinal transection Intra-RVM           Block         27
                                                                                                                                                                                                                                         field)                                   dorsal horn neurons                   lidocaine
                                                                                                                                                                                                                                         Mustard oil (leg)                        Facilitation of the thermal tail-     Spinal transection Electrolytic        Block         28, 29
                                                                                                                                                                                                                                                                                  flick reflex                          RVM lesion Ibotenic acid RVM
                                                                                                                                                                                                                                                                                                                        lesion
                                                                                                                                                                                                                                         Carrageenan (knee joint)                 Enhanced C-fiber-mediated             Spinal transection                     Block         37
                                                                                                                                                                                                                                                                                  flexor motoneuron wind-up
                                                                                                                                                                                                                                         Carrageenan (knee joint)                 Facilitation of the thermal paw-      Intra-RVM lidocaine Ibotenic           Block         29
                                                                                                                                                                                                                                                                                  withdrawal response                   acid RVM lesion
                                                                                                                                                                                                                                         Carrageenan (plantar foot)               Facilitation of the thermal paw-      Intra-RVM lidocaine Ibotenic           No effect     29
                                                                                                                                                                                                                                                                                  withdrawal response                   acid RVM lesion
                                                                                                                                                                                                                                         Formalin (foot)                          Facilitation of the thermal tail-     Spinal transection Electrolytic        block         41, 42
                                                                                                                                                                                                                                                                                  flick reflex                          RVM lesion
                                                                                                                                                                                                                                         Neuropathic
                                                                                                                                                                                                                                         Spinal nerve ligation                    Tactile allodynia, foot               Intra-RVM lidocaine                    Block         44
                                                                                                                                                                                                                                         Spinal nerve ligation                    Tactile allodynia, foot               Spinal transection                     Block         45
                                                                                                                                                                                                                                                                                  Facilitation of the thermal paw-
                                                                                                                                                                                                                                                                                  withdrawal response
                                                                                                                                                                                                                                         Spinal nerve cut                         Tactile allodynia, foot               Spinal transection                     Block         46
                                                                                                                                                                                                                                         Illness
                                                                                                                                                                                                                                         LPS (intraperitoneal)                    Facilitation of the thermal tail-     Electrolytic RVM lesion                Block         42, 50
                                                                                                                                                                                                                                                                                  flick reflex                          Electrolytic NTS lesion
                                                                                                                                                                                                                                              In related studies, we documented a significant contribution of descending facilitatory influences in a model of thermal hyperalgesia
                                                                                                                                                                                                                                         involving topical application of mustard oil to the hind leg and measurement of the spinal nociceptive tail-flick reflex ( 28 ). It was found
                                                                                                                                                                                                                                         that midthoracic spinal transection or electrolytic lesion of the RVM prevented facilitation of the tail-flick reflex produced by mustard oil.
                                                                                                                                                                                                                                         To confirm an involvement of cells in the RVM in modulating this secondary thermal hyperalgesia, we found that RVM lesion using the
                                                                                                                                                                                                                                         soma-selective neurotoxin ibotenic acid resulted in a similar block of mustard oil-induced hyperalgesia ( 29 ).
                                                                                                                                                                                                                                              Active participation of descending facilitatory influences from the RVM in modulating mustard oil-induced hyperalgesia is supported
                                                                                                                                                                                                                                         further by evidence that NMDA and neurotensin receptors in the RVM modulate this secondary thermal hyperalgesia. As indicated above,
                                                                                                                                                                                                                                         neurotensin receptors ( 7 , 8 ) and NMDA receptors ( 30 , 31 ) in the RVM have been implicated in descending facilitation of spinal
                                                                                                                                                                                                                                         nociception. Selective blockage of these receptors should then modulate hyperalgesia. Indeed, intra-RVM injection of a selective
                                                                                                                                                                                                                                         neurotensin receptor antagonist (SR48692) or NMDA receptor antagonist [2-amino-5-phosphonovaleric acid (APV)] fully and dose
                                                                                                                                                                                                                                         dependently prevented mustard oil-induced facilitation of the tail-flick reflex ( 28 , 30 ). It is known that generation of nitric oxide (NO )
                                                                                                                                                                                                                                         is one downstream consequence of NMDA receptor activation ( 32 ). In complementary studies, we showed that intra-RVM administration
the authoritative version for attribution.
                                                                                                                                                                                                                                         N-nitro-L-arginine methyl ester (L-NAME), like the NMDA receptor antagonist APV, attenuated mustard oil-induced hyperalgesia (
                                                                                                                                                                                                                                         30 ). Conversely, microinjection of the NO donor GEA 5024 (or of NMDA itself) dose dependently facilitated the tail-flick reflex in
                                                                                                                                                                                                                                         nave rats. The involvement of NO in the RVM was further supported by a significant increase in the number of NADPHdiaphorase-
                                                                                                                                                                                                                                         labeled cells at the time of maximal mustard oil-induced hyperalgesia. Finally, in a model of visceral hyperalgesia in which the
                                                                                                                                                                                                                                         inflammogen zymosan is instilled into the colon, both APV and L-NAME given into the RVM 3 hr after colonic inflammation reversed
                                                                                                                                                                                                                                         the hyperalgesia for the duration of drug action, suggesting that the RVM plays a role in maintenance of the hyperalgesia ( 31 ). Similar to
                                                                                                                                                                                                                                         what was seen in the model of mustard oil hyperalgesia, both NADPHdiaphorase-labeled cell numbers and the number of cells
                                                                                                                                                                                                                                         immunostained for the neuronal isoform of NO -synthase were significantly increased in the RVM 3 hr after colonic inflammation. These
                                                                                                                                                                                                                                         results support a role for descending facilitatory influences in the maintenance of mustard oil-induced and visceral hyperalgesia involving
                                                                                                                                                                                                                                         activation of NMDA and neurotensin receptors in the RVM.
                                                                                                                                                                                                                                               Carrageenan. Several models of hyperalgesia involving subcutaneous injection of carrageenan have been characterized. Carrageenan
                                                                                                                                                                                                                                         is a water-extractable polysaccharide obtained from various seaweeds. Injection of lambda carrageenan (a hydrocolloid that does not form
                                                                                                                                                                                                                                         a gel) into the plantar foot, or intraarticular injection into the knee joint, results in a localized inflammation, decreased weight bearing,
                                                                                                                                                                                                                                         guarding of the affected limb, and hyperalgesia (e.g., refs. 33 and 34 ). Carrageenan-induced hyperalgesia is believed to occur as a
                                                                                                                                                                                                                                         consequence of sensitization of primary afferent nociceptors and neuron plasticity intrinsic to the spinal cord ( 35 , 36 ).
                                                                                                                                                                                                                                               Herrero and Cervero ( 37 ) first reported that the A- and C-fiber mediated wind-up of flexor motoneurons after intraarticular (knee)
                                                                                                                                                                                                                                         carrageenan injection was prevented by spinal transection. They concluded that supraspinal modulatory systems, either direct excitatory
                                                                                                                                                                                                                                         influences on spinal neurons or release of local inhibitory controls, are essential for wind-up. We examined a potential contribution of
                                                                                                                                                                                                                                         descending facilitatory influences from the RVM to enhanced behavioral nociceptive responses after intraplantar or intraarticular (knee)
                                                                                                                                                                                                                                         injection of carrageenan ( 29 ). Intraplantar injection of carrageenan and subsequent thermal stimulation of the plantar surface of the
                                                                                                                                                                                                                                         hindpaw is a model of primary hyperalgesia; intraarticular injection of carrageenan and subsequent thermal stimulation of the plantar
                                                                                                                                                                                                                                         surface of the hindpaw is a model of secondary hyperalgesia. Inactivation of the RVM by lidocaine microinjection reversed, and prior
                                                                                                                                                                                                                                         permanent inactivation of the RVM by ibotenic acid lesion completely blocked, facilitation of the thermal paw-withdrawal response after
                                                                                                                                                                                                                                         intraarticular carrageenan injection. RVM inactivation by either lidocaine or ibotenic acid was ineffective, however, in preventing thermal
                                                                                                                                                                                                                                         hyperalgesia after intraplantar carrageenan injection (i.e., model of primary hyperalgesia). These results suggest that these two models of
                                                                                                                                                                                                                                         carrageenan-induced thermal hyperalgesia are differentially modulated in the central nervous system. Additionally, similar to mustard oil-
                                                                                                                                                                                                                                         induced secondary hyperalgesia, intra-RVM injection of a selective neurotensin receptor antagonist (SR48692) or NMDA receptor
                                                                                                                                                                                                                                         antagonist (APV) was found to block facilitation of the thermal pawwithdrawal response after intraarticular, but not intraplantar,
                                                                                                                                                                                                                                         carrageenan injection ( Fig. 1 ). These results further support a contribution of descending facilitatory influences to secondary hyperalgesia
                                                                                                                                                                                                                                         that is mediated by neurotensin and NMDA receptors in the RVM.
                                                                                                                                                                                                                                               Formalin. Subcutaneous injection of formalin into the dorsum of the rodent hindpaw is a well characterized model in which animals
                                                                                                                                                                                                                                         exhibit spontaneous pain behaviors (shaking, licking of the injected hindpaw) as well as hyperalgesia ( 38 , 39 ). Additionally, formalin
                                                                                                                                                                                                                                         has been shown to produce secondary hyperalgesia after subcutaneous injection into either the hindpaw or tail ( 40 , 41 ). A significant
                                                                                                                                                                                                                                         contribution of supraspinal sites to formalin-produced secondary hyperalgesia was reported by Wiertelak et al. ( 41 ), who found that
                                                                                                                                                                                                                                         spinal transection prevented facilitation of the tail-flick reflex after formalin injection into the hindpaw. That activation of descending
                                                                                                                                                                                                                                         facilitatory influences from the RVM modulates this hyperalgesia was subsequently supported by the finding that electrolytic lesion of the
                                                                                                                                                                                                                                         RVM prevented facilitation of the tail-flick reflex after formalin injection ( 42 ).
                                                                                                                                                                                                                                               Neuropathic Models of Hyperalgesia. Animal models of neuropathic pain generally involve loose ligation of peripheral nerves,
                                                                                                                                                                                                                                         which results in spontaneous pain behaviors, enhanced responses of spinal-dorsal horn nociceptive neurons, and hyperalgesia (for review,
                                                                                                                                                                                                                                         see ref. 43 ). A contribution of supraspinal sites to neuropathic pain after spinal nerve ligation was initially reported by Pertovaara et al. (
                                                                                                                                                                                                                                         44 ). In that study, the tactile allodynia that develops after unilateral ligation of the L5 and L6 spinal nerves was found to be attenuated by
                                                                                                                                                                                                                                         inactivation of the RVM by lidocaine injection. The lidocaine effect was determined to be localized within the RVM and independent of
                                                                                                                                                                                                                                         an opioid mechanism, suggesting an inactivation of a descending facilitatory influence from the RVM. These results were supported in a
                                                                                                                                                                                                                                         subsequent study ( 45 ), in which spinal transection was found to abolish the tactile allodynia as well as thermal hyperalgesia produced by
                                                                                                                                                                                                                                         ligation of the L5 and L6 spinal nerves. Additionally, Kauppila ( 46 ) found spinal transection to block mechanical hyperalgesia observed
                                                                                                                                                                                                                                         after a chronic sciatic nerve cut. Thus, neuropathic pain after peripheral nerve injury appears to involve, at least in part, activation of
                                                                                                                                                                                                                                         descending facilitatory influences from supraspinal sites, including the RVM.  ropcapli9
                                                                                                                                                                                                                                               Illness-Induced Models of Hyperalgesia. The systemic administration of lipopolysaccharide (LPS) has been shown to produce a
                                                                                                                                                                                                                                         number of symptoms associated with illness, such as fever, lethargy, decreased food and water intake, and increased sleep (for review, see
                                                                                                                                                                                                                                         ref. 47 ). Additionally, administration of LPS produces hyperalgesia through the release of peripheral cytokines (e.g., IL-1) from immune
                                                                                                                                                                                                                                         cells ( 48 , 49 ). In a series of experiments, Watkins et al. ( 49 , 50 ) determined that facilitation of the tail-flick reflex after intraperitoneal
                                                                                                                                                                                                                                         injection of LPS does not involve primary afferent nociceptor input to the spinal dorsal horn. Instead, a novel circuit was proposed
                                                                                                                                                                                                                                         involving IL-1 activation of hepatic vagal afferent fibers that terminate in the nucleus tractus solitarius (NTS). Consistent with this
                                                                                                                                                                                                                                         proposal, electrolytic lesion of the NTS or RVM was found to block facilitation of the tail-flick reflex produced by intraperitoneal LPS.
                                                                                                                                                                                                                                         Because the NTS and RVM are reciprocally connected, direct afferent input to the RVM may mediate this effect, although Watkins et al. (
                                                                                                                                                                                                                                         50 ) implicated an unidentified site rostral to the midmesencephalon as an important relay. This interpretation is consistent with earlier
                                                                                                                                                                                                                                         studies of biphasic effects of electrical stimulation of vagal afferent fibers (see ref. 51 for review). In those experiments, low-intensity
                                                                                                                                                                                                                                         stimulation of vagal afferent fibers was documented to facilitate spinal nociceptive reflexes (tail-flick reflex) and spinal dorsal horn neuron
                                                                                                                                                                                                                                         responses to noxious stimuli. The facilitatory effect of vagal stimulation was abolished after midcollicular decerebration, implicating an
                                                                                                                                                                                                                                         NTSforebrain circuit in descending influences that ultimately exit the brainstem via the RVM. Although the tail-flick reflex is a spinally
                                                                                                                                                                                                                                         organized response, facilitation of this reflex after intraperitoneal LPS similarly appears to involve activation of descending facilitatory
                                                                                                                                                                                                                                         influences from the RVM.
                                                                                                                                                                                                                                               Primary vs. Secondary Hyperalgesia. We and others have studied the effects of spinal cord transection and of reversible (lidocaine)
                                                                                                                                                                                                                                         or permanent (ibotenic acid) inactivation of the RVM in models of primary and secondary hyperalgesia after peripheral tissue insult. The
                                                                                                                                                                                                                                         results reviewed above uniformly support the hypothesis that facilitatory influences from the
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         brainstem significantly contribute to secondary, but not primary, hyperalgesia. What has not yet been addressed specifically is whether the
                                                                                                                                                                                                                                         RVM is necessary and sufficient for development or for maintenance of secondary hyperalgesia. Intra-RVM injection of lidocaine
                                                                                                                                                                                                                                         reverses, in a time-limited fashion, already established secondary hyperalgesia, suggesting a clear role for the RVM in maintenance of
                                                                                                                                                                                                                                         secondary hyperalgesia. Other studies reveal that spinal-cord transection or soma-selective lesion of the RVM prevents development of
                                                                                                                                                                                                                                         secondary, but not primary, hyperalgesia. Accordingly, available evidence suggests that the RVM is important to both the development
                                                                                                                                                                                                                                         and maintenance of secondary hyperalgesia. The studies reviewed here all have examined behavioral consequences of peripheral tissue
                                                                                                                                                                                                                                         insult, and there are limited data available yet with respect to the direct influence of the RVM on spinal neuron plasticity (central
                                                                                                                                                                                                                                         sensitization).
                                                                                                                                                                                                                                           FIG. 1. Involvement of descending facilitatory influences from the RVM in models of secondary, but not primary, thermal
                                                                                                                                                                                                                                           hyperalgesia after peripheral inflammation. (A) RVM lesion produced by ibotenic acid prevented facilitation of the thermal paw-
                                                                                                                                                                                                                                           withdrawal response after intraarticular carrageenan/kaolin injection into the knee (t test, P < 0.05), but was ineffective in preventing
                                                                                                                                                                                                                                           facilitation of the thermal paw-withdrawal response after intraplantar carrageenan injection into the foot (model of primary
                                                                                                                                                                                                                                           hyperalgesia). (B) Intra-RVM microinjection of the NMDA receptor antagonist APV (1 pmol/1 l), or (C) intra-RVM microinjection
                                                                                                                                                                                                                                           of the neurotensin receptor antagonist SR48692 (3 nmol/1 l) attenuated secondary, but not primary, hyperalgesia (t test, P < 0.05).
                                                                                                                                                                                                                                           All data are represented as mean  SEM of the percent change in thermal paw-withdrawal latency (%) from the control response for
                                                                                                                                                                                                                                           the ipsilateral (inflamed) hindlimb. In experiments involving ibotenic acid RVM lesion, responses are represented at the time of
                                                                                                                                                                                                                                           maximal hyperalgesia (3 hr after carrageenan injection). Intra-RVM microinjection of APV or SR48692 was performed at the time of
                                                                                                                                                                                                                                           maximal hyperalgesia (3 hr), and responses are represented at the time of maximal drug effect after intra-RVM injection (10 min).
                                                                                                                                                                                                                                              Two studies have examined changes in spinal neuron behavior associated with peripheral tissue insult. Schaible et al. ( 52 )
                                                                                                                                                                                                                                         examined, in the cat, the effect of acute inflammation of the knee joint with a mixture of kaolin and carrageenan on spinal dorsal horn
                                                                                                                                                                                                                                         neurons. They documented that spontaneous activity and responses to both innocuous and noxious stimulation of the joint were increased
                                                                                                                                                                                                                                         progressively as the inflammation progressed. Neuron activity and responses to stimulation were increased further when spinal cord
                                                                                                                                                                                                                                         transmission was interrupted temporarily by cold block of the lower thoracic spinal cord. They concluded that spinal neuron
                                                                                                                                                                                                                                         hyperexcitability associated with a peripheral inflammation was counteracted by enhancement of descending inhibitory influences. Ren
                                                                                                                                                                                                                                         and Dubner ( 53 ) studied, in the rat, the effect of lidocaine injection into the midline RVM on spinal neuron responses to stimulation of a
                                                                                                                                                                                                                                         hindpaw inflamed with complete Freunds adjuvant. During the action of lidocaine, neuron spontaneous activity and responses to
                                                                                                                                                                                                                                         mechanical and thermal stimulation applied to the hindpaw were significantly increased, which was interpreted to indicate that peripheral
                                                                                                                                                                                                                                         inflammation leads to an enhanced descending inhibition. Both of these studies used models of primary hyperalgesia (stimuli were applied
                                                                                                                                                                                                                                         to the injured tissue). Both also noted, however, an increase in the size of neuron receptive fields, usually taken as an indication of
                                                                                                                                                                                                                                         secondary hyperalgesia. Although neither report directly addresses the hypothesis advanced here, both contribute relevant information.
                                                                                                                                                                                                                                         Both document an active modulation by the brainstem of spinal neuron excitability in the presence of tissue injury, confirming activation
                                                                                                                                                                                                                                         by peripheral noxious inputs of descending inhibition that can modulate further spinal nociceptive transmission.
                                                                                                                                                                                                                                              The generality of the present hypothesis remains to be established. Most of the studies done to date have examined secondary thermal
the authoritative version for attribution.
                                                                                                                                                                                                                                         hyperalgesia. Thermal hyperalgesia is widely used in studies with nonhuman animals, but secondary thermal hyperalgesia is not of
                                                                                                                                                                                                                                         significant consequence in most instances of tissue injury in humans. The extent to which secondary mechanical hyperalgesia is modulated
                                                                                                                                                                                                                                         by the RVM is unclear. The limited data available to date relate to tactile allodynia and mechanical hyperalgesia in models of neuropathic
                                                                                                                                                                                                                                         pain. Additional studies that use other models of hyperalgesia are necessary. Models of chemically produced hyperalgesia, which may
                                                                                                                                                                                                                                         involve more selective actions on different types of nociceptors, have not been studied extensively. Secondary thermal hyperalgesia
                                                                                                                                                                                                                                         produced by topical application of the C-fiber excitant mustard oil has been documented by several investigators to be influenced by the
                                                                                                                                                                                                                                         RVM. Whether secondary hyperalgesia produced by intradermal injection of capsaicin, which acts at the vanilloid-1 receptor, is similarly
                                                                                                                                                                                                                                         modulated by the RVM has not been reported.
                                                                                                                                                                                                                                              It is also unknown how blockage of central sensitization at the level of the spinal cord (by antagonism of the NMDA receptor, for
                                                                                                                                                                                                                                         example) influences the RVM. It may be that the spinal cord and RVM are both necessary and sufficient to development and maintenance
                                                                                                                                                                                                                                         of secondary hyperalgesia. Results reviewed here clearly indicate that central sensitization at the level of the spinal cord can be modulated
                                                                                                                                                                                                                                         by the RVM, even if the spinal cord is the portal of first entry of the relevant
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         input. Temporally, input to the spinal cord likely precedes receipt of similar input in the brain stem, but it may be that other avenues of
                                                                                                                                                                                                                                         input (e.g., via the vagus) provide an important (more important?) trigger for the RVM.
                                                                                                                                                                                                                                           FIG. 2. Summary diagram illustrating a significant supraspinal contribution to secondary, but not primary, thermal hyperalgesia after
                                                                                                                                                                                                                                           peripheral inflammation. Peripheral injury results in activation and sensitization of peripheral nociceptors and subsequent enhanced
                                                                                                                                                                                                                                           excitability of dorsal horn nociceptive neurons (central sensitization) that contributes to primary hyperalgesia (at site of injury) and
                                                                                                                                                                                                                                           secondary hyperalgesia (adjacent/distant from site of injury). Additionally, it is proposed that stimulation of nociceptors activates a
                                                                                                                                                                                                                                           spinobulbospinal loop, engaging a centrifugal descending nociceptive facilitatory influence from the RVM. Facilitatory influences
                                                                                                                                                                                                                                           are activated by NMDA receptors and NO , and neurotensin (NT) receptors in the RVM and descend to multiple spinal segments to
                                                                                                                                                                                                                                           contribute significantly to secondary hyperalgesia. In contrast, primary hyperalgesia does not involve descending facilitatory
                                                                                                                                                                                                                                           influences from supraspinal sites and is likely the direct result of peripheral nociceptor sensitization and neuroplasticity intrinsic to
                                                                                                                                                                                                                                           the spinal cord. For clarity, the afferent input to the spinal dorsal horn from the site of injury is illustrated as not entering the spinal
                                                                                                                                                                                                                                           cord (which it certainly does).
                                                                                                                                                                                                                                              Returning to the formulation advanced almost 50 years ago by Hardy et al. (1), we believe that a dominant active influence from the
                                                                                                                                                                                                                                         brainstem is necessary for the expression of secondary hyperalgesia (see Fig. 2 ). We acknowledge that there are likely multiple
                                                                                                                                                                                                                                         supraspinal sites involved in responding to peripheral tissue insult. Indeed, the limited data available suggest that forebrain sites can play
                                                                                                                                                                                                                                         an important role, even if the RVM is the final common pathway of facilitatory influences that mediate spinal neuron excitability.
                                                                                                                                                                                                                                              This work was supported by National Institutes of Health awards DA11431 (M.O.U.), NS19912 (G.F.G.), and DA02879 (G.F.G.).
                                                                                                                                                                                                                                         1. Hardy, J. D. , Wolff, H. G. & Goodell, H. ( 1950 ) J. Clin. Invest 29 , 115140 .
                                                                                                                                                                                                                                         2. Lewis, T. ( 1936 ) Clin. Sci. 2 , 373421 .
                                                                                                                                                                                                                                         3. Woolf, C. J. ( 1983 ) Nature (London) 306 , 686688 .
                                                                                                                                                                                                                                         4. LaMotte, R. H. , Shain, C. N. , Simone, D. A. & Tsai, E. F. P. ( 1991 ) J. Neurophysiol. 66 , 190211 .
                                                                                                                                                                                                                                         5. Woolf, C. J. ( 1992 ) in Hyperalgesia and Allodynia , ed. Willis, W. ( Raven , New York ) pp. 221243 .
                                                                                                                                                                                                                                         6. Urban, M. O. & Gebhart, G. F. ( 1998 ) Prog. Brain Res. 116 , 407420 .
                                                                                                                                                                                                                                         7. Urban, M. O. & Gebhart, G. F. ( 1997 ) J. Neurophysiol. 78 , 15501562 .
                                                                                                                                                                                                                                         8. Urban, M. O. & Smith, D. J. ( 1993 ) J. Pharmacol. Exp. Ther. 265 , 580586 .
                                                                                                                                                                                                                                         9. Zhuo, M. & Gebhart, G. F. ( 1992 ) J. Neurophysiol. 67 , 15991614 .
                                                                                                                                                                                                                                         10. Zhuo, M. & Gebhart, G. F. ( 1997 ) J. Neurophysiol. 78 , 746758 .
                                                                                                                                                                                                                                         11. Urban, M. O. , Smith, D. J. & Gebhart, G. F. ( 1996 ) J. Pharmacol. Exp. Ther. 278 , 9096 .
                                                                                                                                                                                                                                         12. Zhuo, M. & Gebhart, G. F. ( 1990 ) Brain Res. 535 , 6778 .
                                                                                                                                                                                                                                         13. Zhuo, M. & Gebhart, G. F. ( 1991 ) Brain Res. 550 , 3548 .
                                                                                                                                                                                                                                         14. Almeida, A. , Tjolsen, A. , Lima, D. , Coimbra, A. & Hole, K. ( 1996 ) Brain Res. Bull. 39 , 715 .
                                                                                                                                                                                                                                         15. Fields, H. L. , Bry, J. , Hentall, I. & Zorman, G. ( 1983 ) J. Neurosci. 3 , 25452552 .
                                                                                                                                                                                                                                         16. Fields, H. L. , Malick, A. & Burstein, R. ( 1995 ) J. Neurophysiol. 74 , 17421759 .
                                                                                                                                                                                                                                         17. Fields, H. L. , Vanegas, H. , Hentall, I. D. & Zorman, G. ( 1983 ) Nature (London) 306 , 684686 .
                                                                                                                                                                                                                                         18. Bederson, J. B. , Fields, H. L. & Barbaro, N. M. ( 1990 ) Somatosens. Res. 7 , 185203 .
                                                                                                                                                                                                                                         19. Morgan, M. M. & Fields, H. L. ( 1994 ) J. Neurophysiol. 72 , 11611170 .
                                                                                                                                                                                                                                         20. Almeida, A. , Tavares, L , Lima, D. & Coimbra, A. ( 1993 ) Neuroscience 55 , 10931106 .
                                                                                                                                                                                                                                         21. Basbaum, A. I. , Clanton, C. H. & Fields, H. L. ( 1978 ) J. Comp. Neurol. 178 , 209224 .
                                                                                                                                                                                                                                         22. Craig, A. D. ( 1995 ) J. Comp. Neurol. 361 , 225248 .
                                                                                                                                                                                                                                         23. Martin, G. F. , Vertes, R. P. & Waltzer, R. ( 1985 ) Exp. Brain Res. 58 , 154162 .
                                                                                                                                                                                                                                         24. Woolf, C. J. & Wall, P. D. ( 1986 ) J. Neurosci. 6 , 14331442 .
                                                                                                                                                                                                                                         25. Woolf, C. J. , Shortland, P. & Sivilotti, L. G. ( 1994 ) Pain 58 , 141155 .
the authoritative version for attribution.
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                         small-diameter nociceptive afferents in an isolated skin-nerve preparation to NGF applied directly to the receptive field. Individual small-
                                                                                                                                                                                                                                         diameter afferent fibers whose conduction velocity was established by the latency of their response to electrical stimulation in the
                                                                                                                                                                                                                                         receptive field were selected if they responded to high-intensity mechanical stimulation. The response to thermal stimulation was
                                                                                                                                                                                                                                         established. NGF then was applied directly to the receptive field for 20 min, and the mechanical and thermal responses were determined
                                                                                                                                                                                                                                         again. NGF was found to lower the threshold to thermal stimulation by about 2C, a change that was statistically significant. However, no
                                                                                                                                                                                                                                         change was noted in the mechanical threshold. Application of saline was found to elicit no effect. In later experiments, NT-3 applied to the
                                                                                                                                                                                                                                         receptive field also was determined not to alter the threshold to noxious heat ( 11 ). If the animals were pretreated with compound 48/80,
                                                                                                                                                                                                                                         NGF had no effect on the noxious heat threshold of individual nociceptive afferents. These experiments established that NGF changes the
                                                                                                                                                                                                                                         threshold of nociceptive afferents and that mast cells are involved. They also confirmed a peripheral locus for the sensitizing action of NGF.
                                                                                                                                                                                                                                               The experiments described so far show that NGF is sufficient to elicit hyperalgesia. A crucial question is whether it is a necessary
                                                                                                                                                                                                                                         intermediate. This issue has been explored by preventing the increase in NGF after an inflammatory injury induced experimentally by
                                                                                                                                                                                                                                         agents such as complete Freunds adjuvant. Antibodies to NGF ( 12 , 21 ) or the immunoadhesin molecule trkA-IgG ( 22 ) were used to
                                                                                                                                                                                                                                         prevent the increase of NGF levels. A uniform finding in these experiments was that hyperalgesia was abolished, suggesting that NGF is a
                                                                                                                                                                                                                                         necessary intermediate in inflammatory hyperalgesia induced by molecules such as complete Freunds adjuvant. It is presently believed
                                                                                                                                                                                                                                         that injury leads to release of cytokines, such as tumor necrosis factor- and IL-1, which cause the release of NGF from cells such as
                                                                                                                                                                                                                                         keratinocytes and fibroblasts ( 23 ). Such a release would initiate the degranulation of mast cells as illustrated in Fig. 1 .
                                                                                                                                                                                                                                               Direct Effects of NGF on Nociceptors. Although the experiments described thus far indicate an indirect role for NGF in peripheral
                                                                                                                                                                                                                                         sensitization via mast cells, the exact locus of action for NGF is not fixed by these findings. The reason for this uncertainty is that mast
                                                                                                                                                                                                                                         cells contain NGF, which would be released on degranulation. Thus, activation of mast cells by NGF might lead to the release of more
                                                                                                                                                                                                                                         NGF, and the presence of trkA receptors on sensory afferents could provide the ability for a direct effect on the peripheral threshold.
                                                                                                                                                                                                                                         Furthermore, repeated daily administration of NGF can eventually produce hyperalgesia despite maintained mast-cell degranulation ( 24 ),
                                                                                                                                                                                                                                         suggesting that, under some circumstances, mast cells can be bypassed. It is already known that NGF can affect the function of sensory
                                                                                                                                                                                                                                         neurons directly, because, both in culture ( 25 ) and in vivo ( 21 ), exogenous NGF administration leads to up-regulation of peptides such
                                                                                                                                                                                                                                         as substance P and calcitonin gene-related peptide in the cell body. However, this up-regulation is a relatively slow effect (hours to days)
                                                                                                                                                                                                                                         involving transcriptional mechanisms and would be much too slow to account for the rapid effect of NGF on sensory thresholds.
                                                                                                                                                                                                                                           FIG. 1. Schematic diagram outlining the relationship of mast cells, nociceptors, and NGF as well as how this system is activated as a
                                                                                                                                                                                                                                           consequence of peripheral injury. Skin injury leads to release of cytokines, such as tumor necrosis factor- and IL-1, which activate
                                                                                                                                                                                                                                           cells, such as keratinocytes and fibroblasts, to release NGF. The NGF can activate nociceptors directly but, in addition, can cause
                                                                                                                                                                                                                                           mast cells to degranulate their products, including 5-HT, histamine, and NGF. This endogenous source of NGF seems to be more
                                                                                                                                                                                                                                           potent than exogenous NGF in sensitizing nociceptors (see text for further details).
                                                                                                                                                                                                                                              We have initiated experiments to examine whether NGF can rapidly increase the threshold of sensory neurons directly. To
                                                                                                                                                                                                                                         accomplish this increase, it is necessary to provide a stimulus that excites these neurons and to determine whether NGF sensitizes the
                                                                                                                                                                                                                                         response. To determine whether this effect is direct, these experiments must be done in culture to avoid the potential actions of other cells
                                                                                                                                                                                                                                         such as mast cells.
                                                                                                                                                                                                                                              Both behavioral evidence and electrophysiological evidence indicate that peripheral nociceptors are very sensitive to capsaicin ( 26 ,
                                                                                                                                                                                                                                         27 ), an ingredient in hot peppers. When capsaicin is applied directly to a cell, it is depolarized as a consequence of a nonspecific increase
                                                                                                                                                                                                                                         in permeability to cations including Ca. Recently, the capsaicin receptor was cloned ( 28 ). This receptor, named VR1, when expressed in
                                                                                                                                                                                                                                         oocytes, is also sensitive to noxious heat, suggesting that the noxious-heat response of primary afferents is mediated via the VR1 receptor.
                                                                                                                                                                                                                                         However, the component of the VR1 receptor sensitive to heat and to capsaicin may be different. NGF has been shown to play some role
                                                                                                                                                                                                                                         in the expression of these receptors, because dorsal root ganglion (DRG) cells cultured for several days in the absence of NGF fail to
                                                                                                                                                                                                                                         display capsaicin sensitivity ( 29 ).
                                                                                                                                                                                                                                              Recently, we have shown that capsaicin responses are influenced acutely by NGF (X.-Q.S. and L.M.M., unpublished work).
                                                                                                                                                                                                                                         Capsaicin was pressure ejected on the somata of small dissociated (<30 m) DRG neurons that were recorded in whole-cell or perforated
                                                                                                                                                                                                                                         patch clamp. With the cell voltage clamped at 60 mV, an inward current was observed. A second identical capsaicin pulse 10 min later
                                                                                                                                                                                                                                         resulted uniformly in a substantially smaller current (Figs. 2 and 3 ). We found that bath-applying NGF during the 10-min interval (
                                                                                                                                                                                                                                         Fig. 2 ) often resulted in elimination of the tachyphylaxis and that the second response was often larger than the first ( Fig. 3 ), as much as
                                                                                                                                                                                                                                         twice as large. In some cells, tachyphylaxis after NGF
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 2. Protocol for testing NGF effects on capsaicin currents on DRG cells recorded in perforated patch clamp. Cells chosen for this
                                                                                                                                                                                                                                           analysis were <30 m in diameter. A microelectrode filled with 1 M capsaicin was placed close to the patched cell, and the
                                                                                                                                                                                                                                           capsaicin was ejected via a brief (400-ms) pressure pulse. The cell was clamped at 60 mV, and the inward capsaicin current lasting
                                                                                                                                                                                                                                           10 s is shown at the top. A second pressure pulse was delivered 10 min later. During the interval, the cell was exposed either to
                                                                                                                                                                                                                                           control saline solution or to 100 ng/ml of NGF. In controls, the response to the second capsaicin pulse was always smaller than the
                                                                                                                                                                                                                                           initial one.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
treatment was similar to that observed after saline treatment. We assume that these cells do not express trkA.
                                                                                                                                                                                                                                           FIG. 3. Examples of capsaicin responses in two DRG cells, the first conditioned by a 10-min exposure to saline (Upper) and the
                                                                                                                                                                                                                                           second conditioned by 100 ng/ml NGF (Lower). Note the smaller response to the second capsaicin pulse (tachyphylaxis) after saline
                                                                                                                                                                                                                                           treatment and the larger response when NGF is placed in the medium.
                                                                                                                                                                                                                                              If we assume that the heat response of nociceptors is mediated by peripheral capsaicin receptors ( 28 ), it is apparent that the
                                                                                                                                                                                                                                         sensitizing effect of NGF may be direct on the capsaicin receptor. It might involve phosphorylation of the capsaicin receptor that is
                                                                                                                                                                                                                                         dephosphorylated as a consequence of the initial capsaicin stimulus ( 30 ). However, the assumption that the heat response is sensitized in
                                                                                                                                                                                                                                         the same way as the response to capsaicin requires direct proof.
                                                                                                                                                                                                                                              These experiments were carried out on DRG cell bodies maintained in culture for up to 1 day. The question that inevitably arises is
                                                                                                                                                                                                                                         whether these conditions adequately test what might be occurring in the terminals. One problem is the presence of the nucleus in the
                                                                                                                                                                                                                                         cultured DRG cells, which would not be present in the terminals in vivo. However, the NGF effect on the capsaicin response in the present
                                                                                                                                                                                                                                         experiments was very rapid, within 10 minutes, which is too fast to require transcription. It follows that the influence of NGF on the
                                                                                                                                                                                                                                         capsaicin response does not involve transcription and probably occurs at a membrane and/or cytoplasmic level.
                                                                                                                                                                                                                                              The finding that NGF directly affects the capsaicin response of DRG cells suggests that NGF should be able to directly sensitize the
                                                                                                                                                                                                                                         response to noxious heat. If so, why should prior degranulation of mast cells be able to abolish the hyperalgesic effects of NGF? In other
                                                                                                                                                                                                                                         words, why should mast cells be necessary for NGF-induced hyperalgesia if NGF can directly sensitize nociceptive neurons as shown in
                                                                                                                                                                                                                                         Fig. 3 ? One possibility is that exogenous NGF is insufficient in amount or does not reach the terminal in large enough concentrations in
                                                                                                                                                                                                                                         the intact skin to influence nociceptive endings in vivo. However, if exogenous NGF degranulates mast cells, and these, in turn, release
                                                                                                                                                                                                                                         NGF, then the resulting positive feedback cycle would amplify the amount of NGF activating the sensory ending. In effect, this hypothesis
                                                                                                                                                                                                                                         suggests that exogenous NGF acts as a trigger that liberates NGF from mast cells. This latter source of NGF is postulated to be necessary
                                                                                                                                                                                                                                         because of its intimate relationship with nociceptive afferent terminals, which allows it to condition the nociceptive terminal. The second
                                                                                                                                                                                                                                         possibility is that other substances released from the mast cell, such as 5-HT or histamine, are necessary to enable NGF to have its full
                                                                                                                                                                                                                                         effect on nociceptive terminals.
                                                                                                                                                                                                                                              trkB Agonists and Hyperalgesia. A recent finding of interest in the context of neurotrophin involvement in hyperalgesia is that
                                                                                                                                                                                                                                         administration of exogenous NGF leads to up-regulation of the trkB agonist BDNF in trkA-expressing sensory neurons ( 31 , 32 ). There is
                                                                                                                                                                                                                                         evidence that BDNF can be transported anterogradely in intact axons to the periphery ( 33 ), raising the possibility that this neurotrophin
                                                                                                                                                                                                                                         plays some role in inflammation-triggered events. In support of this possibility, both trkB agonists (BDNF and NT-4) were found to evoke
                                                                                                                                                                                                                                         heat hyperalgesia when injected locally into the skin, and both of these agents were shown to elicit sensitization of individual nociceptive
                                                                                                                                                                                                                                         afferent fibers to noxious heat ( 11 , 20 ). Both the hyperalgesia ( 11 ) and the acute fiber sensitization ( 20 ) elicited by NT-4 were blocked
                                                                                                                                                                                                                                         in mast-cell-depleted preparations, suggesting the operation of a mechanism similar to the one mediating the hyperalgesic effects of NGF.
                                                                                                                                                                                                                                         The interpretation of these findings is still uncertain, because it is not known whether skin mast cells express functional trkB receptors.
                                                                                                                                                                                                                                         Intraperitoneal mast cells are known to express only trkA receptors ( 14 ), but blood mast cells, for example, express trkA, trkB, and trkC
                                                                                                                                                                                                                                         receptors ( 34 ).
                                                                                                                                                                                                                                              NT-3 did not elicit peripheral sensitization to noxious heat ( 11 ). This result might be interpreted as indicating that mast cells
                                                                                                                                                                                                                                         responsible for this effect do not express trkC receptors. However, the selectivity probably resides with the afferent fibers, because it is
                                                                                                                                                                                                                                         known that trkC receptors are expressed only on large-diameter afferents, whereas trkA is expressed only on small-diameter afferents ( 5 ).
                                                                                                                                                                                                                                         trkB is expressed on cells in the DRG with a wide range of sizes, with considerable numbers of cells coexpressing trkA and trkB.
                                                                                                                                                                                                                                         However, there is virtually no coexpression of trkA and trkC on sensory neurons ( 5 ).
                                                                                                                                                                                                                                                                                                         CONCLUSIONS
                                                                                                                                                                                                                                               If we consider capsaicin as a surrogate for noxious heat in activating the VR1 receptor ( 28 ), our findings suggest that NGF acts as a
                                                                                                                                                                                                                                         peripheral sensitizing agent, at least in part by sensitizing the response of the nociceptor to noxious heat directly. NGF is not the first such
                                                                                                                                                                                                                                         sensitizing agent to be described. It has been known that other sensitizing agents exist, including prostaglandins and bradykinin.
                                                                                                                                                                                                                                         Previously, prostaglandin E2 has been shown to sensitize sensory neurons to capsaicin ( 35 ). Acute exposure of cultured neonatal DRG
                                                                                                                                                                                                                                         cells to bradykinin can enhance their sensitivity to capsaicin and to low pH ( 36 ).
                                                                                                                                                                                                                                               These agents do not seem to function in isolation. For example, bradykinin activates postganglionic efferents in the skin, and these
                                                                                                                                                                                                                                         release prostaglandin E2 ( 37 ). Inactivation of postganglionic efferents reduces NGF-induced hyperalgesia (38), indicating interaction
                                                                                                                                                                                                                                         between NGF and prostaglandins in hyperalgesia. NGF also interacts with bradykinin, in part by stimulating the delayed up-regulation of
                                                                                                                                                                                                                                         BK1 receptors via release of kallikreins from mast cells (see ref. 13 for review). Blockade of BK1 receptors can transiently diminish NGF-
                                                                                                                                                                                                                                         induced hyperalgesia ( 13 ), further indication of the interrelationship of these agents in causing peripheral sensitization.
                                                                                                                                                                                                                                               Thus, NGF seems to be one of a number of sensitizing agents present in peripheral tissues. What is not clear at present is what the
                                                                                                                                                                                                                                         integrative function of these individual agents might be in causing pain associated with inflammatory injury. An answer might be found by
the authoritative version for attribution.
                                                                                                                                                                                                                                         considering the principal cellular events responsible for production of these different agents: cell breakdown for prostaglandin E2, clotting
                                                                                                                                                                                                                                         for bradykinin, and the immune reaction (mast cells) for NGF (see ref. 39 for review). Evidently, the hyperalgesia accompanying
                                                                                                                                                                                                                                         inflammation is sufficiently adaptive in terms of protection by immobilization of the affected body part that redundant mechanisms
                                                                                                                                                                                                                                         involving mast cells ( 12 ), sympathetic efferents ( 38 ), and neutrophils ( 19 ) have evolved. However, in some cases hyperalgesia
                                                                                                                                                                                                                                         becomes maladaptive, particularly if it elicits central sensitization ( 40 ) that causes the hyperalgesia to outlast the peripheral damage. It
                                                                                                                                                                                                                                         then becomes essential to minimize peripheral sensitization to reduce both the immediate nociceptive effects as well as the long-lasting
                                                                                                                                                                                                                                         ones produced by central sensitization ( 40 ).
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                              This work was supported by National Institutes of Health Grants NS-14899 and NS-32264. Partial support was furnished by the
                                                                                                                                                                                                                                         Javits Neuroscience Award NS-16996 (to L.M.M.). We thank Genentech for the NGF and NT-4/5 and Regeneron Pharmaceuticals
                                                                                                                                                                                                                                         (Tarrytown, NY) for the BDNF and NT-3 used in our studies reviewed in this paper.
                                                                                                                                                                                                                                         1. Davies, A. M. ( 1996 ) Philos. Trans. R. Soc. London B 351 , 389394 .
                                                                                                                                                                                                                                         2. Thoenen, H. ( 1991 ) Trends Neurosci. 14 , 165170 .
                                                                                                                                                                                                                                         3. Barbacid, M. ( 1994 ) J. Neurobiol. 2 , 13861403 .
                                                                                                                                                                                                                                         4. Lewin, G. R. , Ritter, A. M. & Mendell, L. M. ( 1992 ) J. Neurosci. 12 , 18961905 .
                                                                                                                                                                                                                                         5. McMahon, S. B. , Armanini, M. P. , Ling, L. H. & Phillips, H. S. ( 1994 ) Neuron 12 , 11611171 .
                                                                                                                                                                                                                                         6. Lindsay, R. M. ( 1996 ) Philos. Trans. R. Soc. London B 351 , 365373 .
                                                                                                                                                                                                                                         7. Lewin, G. R. & Mendell, L. M. ( 1993 ) Trends Neurosci. 16 , 353359 .
                                                                                                                                                                                                                                         8. Averill, S. , McMahon, S. B. , Clary, D. O. , Reichardt, L. F. & Priestley, J. V. ( 1995 ) Eur. J. Neurosci. 7 , 14841494 .
                                                                                                                                                                                                                                         9. Weskamp, G. & Otten, U. ( 1987 ) J. Neurochem. 48 , 17791786 .
                                                                                                                                                                                                                                         10. Lewin, G. R. , Ritter, A. M. & Mendell, L. M. ( 1993 ) J. Neurosci. 13 , 21362148 .
                                                                                                                                                                                                                                         11. Shu, X. Q. , Llinas, A. & Mendell, L. M. ( 1999 ) Pain 80 , 463470 .
                                                                                                                                                                                                                                         12. Lewin, G. M. , Rueff, A. & Mendell, L. M. ( 1994 ) Eur. J. Neurosci. 6 , 19031912 .
                                                                                                                                                                                                                                         13. Rueff, A. , Dawson, A. J. & Mendell L. M. ( 1996 ) Pain 66 , 359372 .
                                                                                                                                                                                                                                         14. Horigome, K. , Pryor, J. C , Bullock, E. D. & Johnson, E. M. , Jr. ( 1993 ) J. Biol. Chem. 268 , 1488114887 .
                                                                                                                                                                                                                                         15. Mazurek, N. , Weskamp, G. , Erne, P. & Otten, U. ( 1986 ) FEBS Lett. 198 , 315320 .
                                                                                                                                                                                                                                         16. Leon, A. , Buriani, A. , Dal Toso, R. , Fabris, M. , Romanello, S. , Aloe, L. & Levi-Montalcini, R. ( 1994 ) Proc. Natl. Acad. Sci. USA 91 , 3739
                                                                                                                                                                                                                                              3743 .
                                                                                                                                                                                                                                         17. Beck, P. W. & Handwerker, H. O. ( 1974 ) Pflgers Arch. 347 , 209222 .
                                                                                                                                                                                                                                         18. Rueff, A. & Dray, A. ( 1993 ) Agents Actions 38 , 1315 .
                                                                                                                                                                                                                                         19. Bennett, G. , al-Rasheed, S. , Hoult, J. R. S. & Brain, S. D. ( 1998 ) Pain 77 , 315322 .
                                                                                                                                                                                                                                         20. Rueff, A. & Mendell, L. M. ( 1996 ) J. Neurophysiol. 76 , 35933596 .
                                                                                                                                                                                                                                         21. Woolf, C. J. , Safieh-Garabedian, B. , Ma, Q.-P. , Crilly, P. & Winter, J. ( 1994 ) Neuroscience 62 , 327331 .
                                                                                                                                                                                                                                         22. McMahon, S. B. , Bennett, D. L. , Priestley, J. V. & Shelton, D. L. ( 1995 ) Nat. Med. 1 , 774780 .
                                                                                                                                                                                                                                         23. Woolf, C. J. , Allchorne, A. , Safieh-Garabedian, B. & Poole, S. ( 1997 ) Br. J. Pharmacol. 121 , 417424 .
                                                                                                                                                                                                                                         24. Amann, R. , Schuligoi, R. , Herzeg, G. & Donnerer, J. ( 1995 ) Pain 64 , 323329 .
                                                                                                                                                                                                                                         25. Lindsay, R. M. & Harmar, A. J. , ( 1989 ) Nature (London) 337 , 362364 .
                                                                                                                                                                                                                                         26. LaMotte, R. , Lundberg, L. E. & Torebjork, H. E. ( 1992 ) J. Physiol. (London) 448 , 749764 .
                                                                                                                                                                                                                                         27. Szolcsanyi, J. , Anton, R , Reeh, P. W. & Handwerker, H. O. ( 1988 ) Brain Res. 446 , 262268 .
                                                                                                                                                                                                                                         28. Caterina, M. J. , Schumacher, M. A. , Tominaga, M. , Rosen, T. A. , Levine, J. D. & Julius D. ( 1997 ) Nature (London) 389 , 816824 .
                                                                                                                                                                                                                                         29. Winter, J. , Forbes, C. A , Sternberg, J. & Lindsay, R. M. ( 1988 ) Neuron 1 , 973981 .
                                                                                                                                                                                                                                         30. Koplas, P. A. , Rosenberg, R. L. & Oxford , G. S. ( 1997 ) J. Neurosci. 17 , 35253537 .
                                                                                                                                                                                                                                         31. Apfel, S. C. , Wright, D. E. , Wiideman, A. M. , Dormia, C. , Snider, W. D. & Kessler, J. A. ( 1996 ) Mol. Cell. Neurosci. 7 , 134142 .
                                                                                                                                                                                                                                         32. Michael, G. J. , Averill, S. , Nitkunan, A. , Rattray, M. , Bennett, D. L. H. , Yan, Q. & Priestley, J. V. ( 1997 ) J. Neurosci. 17 , 84768490 .
                                                                                                                                                                                                                                         33. Tonra, J. R. , Curtis, R. , Wong, V. , differ, K. D. , Park, J. S. , Timmes, A. , Nguyen, T. , Lindsay, R. M. , Acheson, A. & DiStefano, P. S. ( 1998 ) J.
                                                                                                                                                                                                                                              Neurosci. 18 , 43744383 .
                                                                                                                                                                                                                                         34. Tam, S. Y. , Tsai, M. , Yamaguchi, M. , Yano, K. , Butterfield, J. H. & Galli, S. J. ( 1997 ) Blood 90 , 18071820 .
                                                                                                                                                                                                                                         35. Pitchford, S. & Levine, J. D. ( 1991 ) Neurosci. Lett. 132 , 105108 .
                                                                                                                                                                                                                                         36. Stucky, C. L. , Abrahams, L. G. & Seybold, V. S. ( 1998 ) Neuroscience 84 , 12571265 .
                                                                                                                                                                                                                                         37. Taiwo, Y. O. , Heller, P. H. & Levine, J. D. ( 1990 ) Neuroscience 39 , 523531 .
                                                                                                                                                                                                                                         38. Andreev, N. Y. , Dimitrieva, N. , Koltzenburg, M. & McMahon, S. B. ( 1995 ) Pain 63 , 109115 .
                                                                                                                                                                                                                                         39. Levine, J. & Taiwo, Y. ( 1994 ) in Textbook of Pain , eds. Wall, P. D. & Melzack, R. ( Churchill Livingstone , Edinburgh ), pp. 4556 .
                                                                                                                                                                                                                                         40. McMahon, S. B. , Lewin, G. R. & Wall, P. D. ( 1993 ) Curr. Opin. Neurobiol. 3 , 602610 .
the authoritative version for attribution.
                                                                                                                                                                                                                                         SRC, A MOLECULAR SWITCH GOVERNING GAIN CONTROL OF SYNAPTIC TRANSMISSION MEDIATED BY N-METHYL-D-                                          7697
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
ASPARTATE RECEPTORS
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
ASPARTATE RECEPTORS
                                                                                                                                                                                                                                         protein tyrosine kinase (PTK) or protein tyrosine phosphatase (PTP) enzymes are applied into neurons, the whole-cell currents through
                                                                                                                                                                                                                                         native NMDA receptors are increased by PTK and are decreased by PTP (e.g., Fig. 1 ). Conversely, applying PTK inhibitors has been
                                                                                                                                                                                                                                         found to decrease NMDA currents, whereas PTP inhibitors potentiate these currents ( 12 ), indicating that native NMDA receptors are
                                                                                                                                                                                                                                         controlled by the balance of PTK and PTP activity. The increase in the ensemble NMDA currents that are measured by using the whole-
                                                                                                                                                                                                                                         cell recording method was found to be caused by increased activity of individual NMDA channels and there is no change in the single-
                                                                                                                                                                                                                                         channel conductance ( 53 ). This increase in NMDA channel activity is produced through enhancing the gating of already active receptors
                                                                                                                                                                                                                                         rather than through recruiting previously inactive NMDA receptors ( 54 ).
                                                                                                                                                                                                                                               As it is known that NMDA receptor subunit proteins, in particular NR2A ( 55 ) and NR2B ( 56 ), are phosphorylated on tyrosine it is
                                                                                                                                                                                                                                         logical to ask whether the up-regulation of NMDA receptor function is due to phosphorylation of the subunit proteins themselves.
                                                                                                                                                                                                                                         Investigating this would appear to be technologically feasible, because in addition to increasing the function of native NMDA receptors,
                                                                                                                                                                                                                                         PTKs have been found to potentiate the function of recombinant NMDA receptors expressed heterologously ( 57 , 58 ). The number of
                                                                                                                                                                                                                                         tyrosine residues on presumed intracellular domains of NR2A and -2B is large ( 54 ), but some residues are better candidates than others,
                                                                                                                                                                                                                                         depending on the sequence of the surrounding amino acids, which confers selectively for particular PTKs or groups of PTKs ( 59 ).
                                                                                                                                                                                                                                         Recently, through mutagenesis of residues in the C-terminal region of NR2A, three tyrosine residues in this region were found to be
                                                                                                                                                                                                                                         necessary for the up-regulation of recombinant NR1/NR2A receptors expressed in HEK293 cells ( 60 ). This combination of receptor
                                                                                                                                                                                                                                         subunits was known to be especially sensitive to inhibition by Zn2+ ( 61 ), and evidence was found indicating that the enhancement of
                                                                                                                                                                                                                                         currents by tyrosine phosphorylation was caused by removal of this inhibition for NR1/NR2A, and also for NR1/NR2B, receptors. These
                                                                                                                                                                                                                                         findings are surprising and intriguing ( 62 ) because the site for inhibition by Zn2+ is on the extracellular region of the receptor, whereas
                                                                                                                                                                                                                                         tyrosine phosphorylation is presumed to occur at an intracellular site. Thus, there must be an unknown mechanism for transmitting the
                                                                                                                                                                                                                                         effect of phosphorylation from the inside of the membrane to the outside.
                                                                                                                                                                                                                                           FIG. 1. NMDA currents are up-regulated by protein tyrosine kinase and down-regulated by protein tyrosine phosphatase. NMDA
                                                                                                                                                                                                                                           receptor-mediated currents recorded by using the whole-cell patch configuration were evoked by pressure application of L-aspartate
                                                                                                                                                                                                                                           (200 M) at an interval of 1 min from a pressure pipette whose tip was positioned within 100 m from the recorded neuron. A shows
                                                                                                                                                                                                                                           individual whole-cell current traces taken before (Control) or 20 min after the start of recording with PTK (src; 30 units/ml) or PTP
                                                                                                                                                                                                                                           (truncated T cell PTP; 100 M) in the intracellular solution. B Normalized peak NMDA currents recorded with standard intracellular
                                                                                                                                                                                                                                           solution (Control; n = 11 neurons), or intracellular solution supplemented with PTK (n = 8) or PTP (n = 7). For each neuron peak,
                                                                                                                                                                                                                                           NMDA current is normalized by dividing the amplitude of current recorded at 20 min after the start of the recording (I20) by that of
                                                                                                                                                                                                                                           the initial current (I1). (Bar = 2 sec and 200 pA.)
                                                                                                                                                                                                                                              These observations foreshadow a new mechanism that has potential relevance to the general issue of the regulation of ion channels.
                                                                                                                                                                                                                                         However, whether this mechanism applies to native NMDA receptors is doubtful, because it has been found that NMDA channel function
                                                                                                                                                                                                                                         is up-regulated by tyrosine kinase activity even when Zn2+ is chelated ( 63 ), and NMDA channels with low single-channel conductance,
                                                                                                                                                                                                                                         characteristic of NMDA channels that are insensitive to Zn2+ (64), are also up-regulated by PTKs ( 54 ). Therefore, it appears that removal
                                                                                                                                                                                                                                         of Zn2+ inhibition is not the means by which the function of native NMDA receptors is up-regulated by tyrosine phosphorylation. Thus, for
                                                                                                                                                                                                                                         native NMDA channels, the question of whether phosphorylation at the sites implicated by mutagenesis is the means for up-regulating
                                                                                                                                                                                                                                         NMDA channel function remains open. It is alternatively possible that this up-regulation is through phosphorylation of other tyrosine
                                                                                                                                                                                                                                         residues in the NMDA receptor subunit proteins or by phosphorylation of an associated protein, such as one of many proteins already
                                                                                                                                                                                                                                         known to bind to the receptors ( 65  68 ).
                                                                                                                                                                                                                                              Src Is an Endogenous PTK That Up-Regulates NMDA Receptor Function. Once it had been determined that NMDA receptor
                                                                                                                                                                                                                                         function is regulated by PTKs and PTPs, a central question to be addressed was that of identifying the endogenous enzymes involved.
                                                                                                                                                                                                                                         Notionally, this is not a trivial task, because the mammalian genome is expected to encode more than a thousand PTKs ( 69 ) and nearly as
                                                                                                                                                                                                                                         many PTPs ( 70 ). Many of these enzymes are known to be expressed in the spinal cord and elsewhere in the CNS ( 20 , 71 ), providing
                                                                                                                                                                                                                                         numerous potential candidates for the endogenous enzymes. Nevertheless, an endogenous PTK regulating NMDA receptors has been
                                                                                                                                                                                                                                         identified, as described below, and there is preliminary evidence for a possible PTP ( 72 ).
                                                                                                                                                                                                                                              It is well known that PTKs fall into two main categories: receptor and nonreceptor kinases ( 73 , 74 ). Within each of these categories,
                                                                                                                                                                                                                                         there are numerous families with common features, in terms of primary sequence and domain structure. These common features have
                                                                                                                                                                                                                                         permitted the development of pharmacological tools, including peptides and antibodies with activity against particular families of
                                                                                                                                                                                                                                         enzymes, that have allowed the screening of broad groups of kinases. We took advantage of such reagents during our hunt to discover the
                                                                                                                                                                                                                                         PTK regulating NMDA receptors ( 17 , 54 ). As a first step, we used a reagent that activates PTKs in the Src family: the phosphopeptide,
                                                                                                                                                                                                                                         EPQ(pY)EEIPIA ( 75 ), which was found to enhance NMDA channel function. Conversely, channel function is depressed by an antibody,
                                                                                                                                                                                                                                         anti-cst1, which inhibits Src-family PTKs ( 76 ). These results indicated that the endogenous PTK was a member of the Src family.
                                                                                                                                                                                                                                              The family of Src kinases comprises a total of nine members, five of whichSrc, Fyn, Lyn, Lck, and Yesare known to be
                                                                                                                                                                                                                                         expressed in the CNS. All members of the Src family contain highly homologous regionsthe C-terminal, catalytic, Src homology 2, and
the authoritative version for attribution.
                                                                                                                                                                                                                                         Src homology 3 domains ( 77 ). The various members do, however, have substantial differences in a region of low sequence conservation
                                                                                                                                                                                                                                         near the N terminus known as the unique domain. Therefore, reagents directed against this domain may distinguish between the various
                                                                                                                                                                                                                                         Src family members.
                                                                                                                                                                                                                                              Src was identified as the specific member of the family that regulates NMDA channel function by means of testing one such reagent,
                                                                                                                                                                                                                                         the antibody, anti-src1, which selectively blocks the function of Src but not other members of the Src family ( 78 ). It was found that anti-
                                                                                                                                                                                                                                         src1 caused a decrease in NMDA channel activity when this antibody was applied to the cytoplasmic face of membrane patches containing
                                                                                                                                                                                                                                         NMDA channels (e.g., Fig. 2 A). In contrast, a control IgG had no effect. Moreover, anti-src1 prevented the enhancement of NMDA
                                                                                                                                                                                                                                         channel activity by EPQ(pY)EEIPIA, indicating that Src is
                                                                                                                                                                                                                                         SRC, A MOLECULAR SWITCH GOVERNING GAIN CONTROL OF SYNAPTIC TRANSMISSION MEDIATED BY N-METHYL-D-                                         7699
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
ASPARTATE RECEPTORS
                                                                                                                                                                                                                                         necessary for the effect of the activating peptide. As would be anticipated if Src indeed up-regulates NMDA receptors, we found that
                                                                                                                                                                                                                                         applying recombinant pp60c-src increased NMDA channel activity ( Fig. 2 B), an effect not produced by heat-inactivating the kinase just
                                                                                                                                                                                                                                         before use.
                                                                                                                                                                                                                                           FIG. 2. Regulation of NMDA receptor single-channel activity in inside-out patches by Src and overlapping distribution of Src and
                                                                                                                                                                                                                                           NMDA receptor subunit proteins. (A) A continuous record of NMDA channel-open probability (Po). Anti-src1 was applied to the
                                                                                                                                                                                                                                           cytoplasmic face of the patch during the period indicated. Po was calculated in bins of 10 sec. (B) A record of NMDA channel Po
                                                                                                                                                                                                                                           from a different inside-out patch with Src applied to the cytoplasmic side as indicated. (C) Confocal images show immunofluorescent
                                                                                                                                                                                                                                           labeling of a dorsal horn primary culture by antibodies recognizing NR2A/B subunit proteins (green; courtesy of R. Wenthold,
                                                                                                                                                                                                                                           National Institutes of Health, Bethesda, MD) or Src (red). Bottom shows the merged images; areas showing overlapping fluorescence
                                                                                                                                                                                                                                           are yellow. We found similar colocalization when anti-NR1 and anti-Src antibodies were used. Also, experiments without primary
                                                                                                                                                                                                                                           antibodies or with primary antibodies incubated with the respective immunogen peptides showed no labeling. (Bar = 10 m.)
                                                                                                                                                                                                                                              Anti-src1 does not bind to the catalytic domain of Src, and we were therefore curious as to its mechanism of action. Along this line, it
                                                                                                                                                                                                                                         was determined that applying a peptide, Src(4058), comprising the region in Src which the antibody recognizes, i.e., amino acids 4058,
                                                                                                                                                                                                                                         to the cytoplasmic side of membrane patches reduced NMDA channel activity. A control peptide with the same amino acid composition
                                                                                                                                                                                                                                         but in random order, scrambled Src(4058 ), had no effect on channel function. Because it was found that Src(4058) did not block in
                                                                                                                                                                                                                                         vitro phosphorylation of a small substrate peptide by recombinant Src, we concluded that amino acids in the region 4058 may interact
                                                                                                                                                                                                                                         with a component of the receptor complex and that this interaction is necessary for the effect of Src on NMDA channels. The region 4058
                                                                                                                                                                                                                                         is within the unique domain of Src, and these results implicated this domain as being functionally important in this well known enzyme.
                                                                                                                                                                                                                                              Through kinetic analysis of the NMDA channel activity in the patches, it was determined that the effect of Src is due to an increase in
                                                                                                                                                                                                                                         channel gating during single activations of the receptor. This is relevant because synaptic responses mediated by NMDA receptors are
                                                                                                                                                                                                                                         caused by single receptor activations ( 79 ). Thus, if NMDA receptors that are synaptically stimulated are affected similarly to the
                                                                                                                                                                                                                                         receptors in the patches, which are by necessity extrasynaptic, it was predicted that Src should increase synaptic NMDA responses. This
                                                                                                                                                                                                                                         was confirmed in studies of spontaneously occurring miniature excitatory postsynaptic currents (mEPSCs) ( 17 ). Consistent with these
                                                                                                                                                                                                                                         electrophysiological results, it was found by using immunocytochemistry that the distribution of Src within neurons overlaps with that of
                                                                                                                                                                                                                                         NMDA receptors ( Fig. 2 C) and that Src is localized at sites where NMDA receptors are highly enriched, presumably at synapses.
                                                                                                                                                                                                                                              Whether Src is physically associated with NMDA channels could not be determined from any of the previous experiments:
                                                                                                                                                                                                                                         membrane patches are large in comparison with the size of the proteins that comprise ion channels, and confocal microscopy does not
                                                                                                                                                                                                                                         have sufficient spatial resolution. While it was therefore possible that Src was separate from the NMDA receptor complex, we found that
                                                                                                                                                                                                                                         Src and NMDA receptor subunit proteins coprecipitate, demonstrating that Src is associated with the NMDA channel complex. The
                                                                                                                                                                                                                                         coprecipitation might be via a direct interaction between Src and an NMDA receptor subunit protein or, alternatively, it is possible that Src
                                                                                                                                                                                                                                         associates with NMDA channels by means of an intervening adaptor protein. Taking all of the information together, it was concluded that
                                                                                                                                                                                                                                         Src is physically associated with and up-regulates the function of NMDA receptors.
                                                                                                                                                                                                                                              Src is expressed at high levels within the CNS with a number of neuron-specific isoforms being generated by alternative splicing of
                                                                                                                                                                                                                                         one or more cassettes ( 19 , 80 , 81 ) after amino acid 114, which is in the SH3 domain. In the nervous system, Src has been found to be
                                                                                                                                                                                                                                         localized both pre- and postsynaptically ( 20 , 82 ). The postsynaptic localization is especially relevant to the modulation of NMDA
                                                                                                                                                                                                                                         receptor function because Src has been found in the postsynaptic density ( 82 , 83 ), which is the main structural component of excitatory
                                                                                                                                                                                                                                         synapses and is where glutamate receptors are concentrated.
                                                                                                                                                                                                                                              Src Up-Regulation of NMDA Receptors in Hippocampal LTP. The studies described above implicating Src in the up-regulation of
                                                                                                                                                                                                                                         NMDA receptor function were focused primarily on neurons from the spinal cord dorsal horn. Because NMDA receptors and Src are
                                                                                                                                                                                                                                         widely expressed in the nervous system, there is the possibility that by regulating the activity of
the authoritative version for attribution.
                                                                                                                                                                                                                                         SRC, A MOLECULAR SWITCH GOVERNING GAIN CONTROL OF SYNAPTIC TRANSMISSION MEDIATED BY N-METHYL-D-                                        7700
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
ASPARTATE RECEPTORS
                                                                                                                                                                                                                                         postsynaptic NMDA receptors, tyrosine phosphorylation/ dephosphorylation might modulate the efficacy of synaptic transmission in many
                                                                                                                                                                                                                                         regions of the CNS. One region where Src is highly expressed is the CA1 region of the hippocampus ( 20 ). In this region, a lasting
                                                                                                                                                                                                                                         enhancement of the efficacy of synaptic transmission, LTP, is induced by tetanic stimulation of the Schaffer collateral inputs to CA1
                                                                                                                                                                                                                                         neurons ( 84 , 85 ). It has been established that LTP in the CA1 region is induced by a sequence of biochemical steps occurring in the
                                                                                                                                                                                                                                         pyramidal neurons ( 86 , 87 ). Both PTK function ( 88 ) and NMDA receptor activation ( 89 ) have been found to be necessary for LTP
                                                                                                                                                                                                                                         induction in these neurons. Therefore, we used the reagents characterized previously to determine whether Src participates in LTP in CA1.
                                                                                                                                                                                                                                               It was found that administering anti-src1 or Src(4058) into CA1 neurons prevented the induction of LTP in an acute hippocampal
                                                                                                                                                                                                                                         slice preparation ( 21 ). On the other hand, administering recombinant Src or activating Src by means of the EPQ(pY)EEIPIA peptide
                                                                                                                                                                                                                                         induced a long-lasting enhancement of synaptic responses. This enhancement occluded the induction of LTP and vice versa, implying that
                                                                                                                                                                                                                                         Src activation is sufficient to cause LTP. In addition, by measuring Src activity biochemically in vitro by using an immune-complex kinase
                                                                                                                                                                                                                                         assay, it was found that tetanic stimulation, which produced LTP, caused an increase in the activity of Src within 1 min of the stimulation.
                                                                                                                                                                                                                                         Thus, Src is up-regulated very rapidly as a consequence of tetanic stimulation. Because inhibiting Src did not affect basal synaptic
                                                                                                                                                                                                                                         responses, it was concluded that rapid up-regulation of Src activity is necessary, as well as sufficient, for the induction of LTP.
                                                                                                                                                                                                                                               The principal means by which LTP is expressed in CA1 neurons is enhancement of the -amino-3-hydroxy-5-methyl-4-
                                                                                                                                                                                                                                         isoxazolepropionic acid (AMPA) component of synaptic responses and therefore, our conclusion seemed at odds with other findings ( 17 )
                                                                                                                                                                                                                                         that the AMPA receptor-mediated synaptic response is not potentiated when Src is activated in dorsal horn neurons. However, in our
                                                                                                                                                                                                                                         studies on dorsal horn neurons, intracellular Ca2+ was highly buffered, whereas in the experiments in hippocampal slices low intracellular
                                                                                                                                                                                                                                         Ca2+ buffering was used, because this is required to induce LTP. When Ca2+ buffering was increased in the hippocampal neurons, Src no
                                                                                                                                                                                                                                         longer potentiated the synaptic AMPA responses, but synaptic NMDA responses were still potentiated by Src, as was the case in the dorsal
                                                                                                                                                                                                                                         horn neurons. Thus, enhancement of AMPA responses produced by activating Src depends on a rise in intracellular Ca2+. Because Src is
                                                                                                                                                                                                                                         not a Ca2+-dependent enzyme ( 90 ), these results indicated that Src does not up-regulate AMPA receptors directly but rather does so
                                                                                                                                                                                                                                         indirectly through one or more Ca2+-dependent steps.
                                                                                                                                                                                                                                               In other experiments, it was determined that blocking NMDA receptors prevents but does not reverse Src-induced potentiation of
                                                                                                                                                                                                                                         AMPA responses. Thus, like LTP induced by tetanus, NMDA receptors are necessary to produce, but not to maintain, the potentiation of
                                                                                                                                                                                                                                         AMPA responses induced by activating Src directly. Together, these findings required the development of a new model where activation
                                                                                                                                                                                                                                         of Src appears to be a biochemical mechanism gating the induction of LTP in CA1 neurons ( 54 ). It is hypothesized that during induction
                                                                                                                                                                                                                                         of LTP, Src is rapidly activated, leading to enhanced NMDA receptor function, which boosts the entry of Ca2+ sufficiently to trigger the
                                                                                                                                                                                                                                         downstream signaling cascade.
                                                                                                                                                                                                                                               A key question opened up by this work is, what is the mechanism causing Src activation upon tetanic stimulation? Src has a number
                                                                                                                                                                                                                                         of regulatory sites ( 18 ), and there are numerous biochemical pathways that converge to activate ( 91 , 92 ) or to inhibit ( 93 , 94 ) this
                                                                                                                                                                                                                                         kinase. Determining whether the increase in Src activity is produced by stimulating an activating pathway or by blocking an inhibiting one
                                                                                                                                                                                                                                         and identifying the specific biochemical steps are central goals of future work in this area.
                                                                                                                                                                                                                                               Because the role of Src in LTP induction appears to be to enhance NMDA receptor function, one potential mechanism is
                                                                                                                                                                                                                                         phosphorylation of one of the NMDA receptor subunit proteins, as discussed above. Indeed, it has been found that the level of tyrosine
                                                                                                                                                                                                                                         phosphorylation of the NR2B NMDA receptor subunit is increased after induction of LTP in the dentate gyrus in the hippocampus ( 95 ,
                                                                                                                                                                                                                                         96 ). Like CA1, the dentate is a region where LTP induction is NMDA receptor-dependent and is blocked by inhibitors of PTKs ( 97 ).
                                                                                                                                                                                                                                         Another region where NMDA receptor-dependent synaptic plasticity has been associated with tyrosine phosphorylation of NMDA
                                                                                                                                                                                                                                         receptors is the insular cortex, where it has been found that taste aversion conditioning causes an increase in phosphorylation of NR2B (
                                                                                                                                                                                                                                         98 ). However, whether phosphorylation of NR2B mediates the plasticity in the dentate gyrus or in the insular cortex, and if so by what
                                                                                                                                                                                                                                         mechanism, remains to be determined ( 54 ).
                                                                                                                                                                                                                                               Tyrosine kinases were first implicated in the induction of LTP from experiments showing that LTP is blocked by bath-applied
                                                                                                                                                                                                                                         tyrosine kinase inhibitors ( 88 ). It was found subsequently that mutant mice with targeted deletion of the src gene showed LTP in CA1,
                                                                                                                                                                                                                                         which is a genetic argument against the absolute requirement for Src in the induction of LTP. Also, it was reported that in mice lacking the
                                                                                                                                                                                                                                         Src-family kinase fyn, LTP is blunted but not abolished ( 99 ). The impairment in LTP is age-dependent in fyn / mice, with young fyn
                                                                                                                                                                                                                                         / animals showing LTP comparable to that in wild-type animals ( 100 ). The developmental time at which LTP becomes impaired in fyn
                                                                                                                                                                                                                                         / mice correlates with a large decline in the level of Src expression. Src and Fyn are known to substitute for each other in various
                                                                                                                                                                                                                                         processes ( 101 ). Thus, from our evidence together with that from experiments using genetic manipulation, it appears that in wild-type
                                                                                                                                                                                                                                         animals, Src is a required mediator of LTP induction, whereas in animals that develop without src, another member of the src family,
                                                                                                                                                                                                                                         likely fyn, may substitute. It is possible that, for example, by being upstream of Src activation, Fyn might also be necessary for LTP
                                                                                                                                                                                                                                         induction in the wild-type animal, and this possibility needs to be examined in acute experiments by using Fyn-specific manipulations.
                                                                                                                                                                                                                                               Intracellular Sodium Regulates NMDA Receptors. During high levels of neuronal discharge activity, such as occur during the
                                                                                                                                                                                                                                         induction of lasting changes in synaptic efficacy, there is a large influx of Na+, leading to substantial increases in the intracellular
                                                                                                                                                                                                                                         concentration of Na+ ([Na+]i) ( 102 ). It has been found that during such activity, the level of [Na+]i may increase by 1520 mM in the
                                                                                                                                                                                                                                         neuronal soma ( 103 ) and likely even more in the dendrites ( 102 ). While there can be no doubt that Na+ is the major carrier of electrical
                                                                                                                                                                                                                                         charge responsible for producing action potentials and excitatory postsynaptic potentials, the possibility that raising [Na+]i may act as a
                                                                                                                                                                                                                                         signaling factor in neurons had been virtually ignored. Thus, we recently set out to determine whether Na+ might regulate synaptic
                                                                                                                                                                                                                                         function in postsynaptic neurons ( 22 ).
                                                                                                                                                                                                                                               We first examined the effects of raising [Na+]i on whole-cell NMDA currents evoked by exogenously applied NMDA. Perfusing the
                                                                                                                                                                                                                                         neurons with an elevated [Na+]i solution produced an increase in the amplitude of the NMDA currents of nearly 40% ( 22 ). In this case,
                                                                                                                                                                                                                                         the intracellular perfusion contained [Na+]i of 50 mM, but we only achieved an increase in [Na+]i of about 30 mM, as determined by using
                                                                                                                                                                                                                                         the fluorescent Na+-sensitive dye sodium-binding benzofuran isophthalate (SBFI), presumably because of the very active Na+ pumping in
                                                                                                                                                                                                                                         the neurons. The increase in NMDA currents was not reproduced by perfusing Cs+ at 50 mM, indicating that not all monovalent cations
                                                                                                                                                                                                                                         were able to cause the potentiation and, equally importantly, that the potentiation was not because of lowering intracellular [K+], which
                                                                                                                                                                                                                                         was required to maintain proper osmolarity.
                                                                                                                                                                                                                                               To characterize the effects of intracellular Na+ on NMDA channel gating, single-channel currents were recorded by using cell-
the authoritative version for attribution.
ASPARTATE RECEPTORS
                                                                                                                                                                                                                                         the Na+-ionophore monensin ( 104 ). Ratiometric measurement of [Na+]i was done under similar conditions so that single-channel activity
                                                                                                                                                                                                                                         could be correlated to the actual change in [Na+]i. It was found that NMDA channel activity followed the level of [Na+]iincreasing when
                                                                                                                                                                                                                                         Na+ was raised and falling when [Na+]i was reduced ( Fig. 3 ). We found no change in the single-channel conductance of the NMDA
                                                                                                                                                                                                                                         channels, indicating that the increase in whole-cell current when Na+ was raised could be accounted for by increased NMDA channel
                                                                                                                                                                                                                                         gating.
                                                                                                                                                                                                                                              Neurons express a diversity of Na+-permeable channels, e.g., ionotropic glutamate receptors and voltage-gated Na+ channels, and
                                                                                                                                                                                                                                         therefore we wondered whether Na+ influx through these various channels could affect NMDA channel function. This was investigated by
                                                                                                                                                                                                                                         examining single-channel activity by enclosing single channels within a patch pipette attached to the cell and stimulating surrounding
                                                                                                                                                                                                                                         channels in the cell by bath-applying activators. An important consideration in these experiments was to avoid the known voltage
                                                                                                                                                                                                                                         dependence of NMDA channel gating ( 105 ), and this was done by adjusting the transmembrane potential of the patch so that it was at a
                                                                                                                                                                                                                                         constant level with respect to the channel reversal potential. We thus found that bath-applying agonists to activate NMDA or non-NMDA
                                                                                                                                                                                                                                         receptors outside the patch led to an increase in activity of the NMDA channels within the patch. This increase in activity was prevented
                                                                                                                                                                                                                                         when Na+ was removed from the bath solution. Importantly, removal of Ca2+ from the bath did not alter the effect of stimulating the extra-
                                                                                                                                                                                                                                         patch NMDA receptors. Also, depolarizing the cells by bath-applying a high-K+ solution to mimic the depolarization caused by applying
                                                                                                                                                                                                                                         the agonists did not affect NMDA channel function. Thus, we concluded that NMDA receptor function may be up-regulated by Na+ influx
                                                                                                                                                                                                                                         through neighboring glutamate receptors.
                                                                                                                                                                                                                                           FIG. 3. Increases in [Na+]i by application of monensin potentiate single NMDA channel activity recorded in the cell-attached
                                                                                                                                                                                                                                           configuration. A shows the recording configuration. (B) A continuous record of NMDA channel Po from neurons bathed with
                                                                                                                                                                                                                                           extracellular solution containing 50 mM Na+. (C) shows representative single-channel currents before and during monensin
                                                                                                                                                                                                                                           application. D Changes in Po and mean open time (to) versus [Na+]i during monensin application. For each Na+ concentration, six
                                                                                                                                                                                                                                           patches were tested. *, P < 0.05; **, P < 0.01, MannWhitney U test when compared with the channel activity at 0 mM Na+.
                                                                                                                                                                                                                                              Another main route for Na+ entry into neurons is via voltage-gated channels permeable to Na+, such as those responsible for the
                                                                                                                                                                                                                                         generation of action potentials. To produce a consistent activity of voltage-gated Na+ channels and thereby attain a stable membrane
                                                                                                                                                                                                                                         potential of the cell, as required to accurately record channel function in the cell-attached patches, we bath-applied the alkaloid veratridine
                                                                                                                                                                                                                                         ( 106 ). Veratridine caused a significant increase in NMDA channel activity, and the effects of veratridine were prevented when it was
                                                                                                                                                                                                                                         applied in the presence of tetrodotoxin. Thus, by using veratridine as a surrogate for evoking action potentials, we concluded that influx of
                                                                                                                                                                                                                                         Na+ through tetrodotoxin-sensitive voltage-gated Na+ channels is sufficient to increase NMDA channel activity.
                                                                                                                                                                                                                                              The NMDA receptors studied by using the cell-attached recordings are of necessity localized extrasynaptically. To determine whether
                                                                                                                                                                                                                                         synaptic NMDA receptors are affected by changing [Na+]i, we studied spontaneous mEPSCs. We found that applying Na+ into neurons via
                                                                                                                                                                                                                                         the patch pipette significantly increased the NMDA receptor-mediated component of the mEPSCs. By contrast, the non-NMDA receptor
                                                                                                                                                                                                                                         component of the mEPSCs was not altered by raising [Na+]i. Applying Cs+ into the cell did not affect either the NMDA or the non-NMDA
                                                                                                                                                                                                                                         receptor-mediated components of the mEPSCs. Thus, increasing [Na+]i selectively amplifies synaptic responses mediated by NMDA but
                                                                                                                                                                                                                                         not non-NMDA receptors. Taking into account the effects of varying Na+ levels on NMDA channel activity in cell-attached patches
                                                                                                                                                                                                                                         described above, it appears that the efficacy of synaptic transmission through NMDA receptors tracks the level of Na+ in the postsynaptic
                                                                                                                                                                                                                                         neuron.
                                                                                                                                                                                                                                              A Src Kinase Controls the Enhancement of NMDA Channel Function by [Na+]i. To lay the foundation for understanding the
                                                                                                                                                                                                                                         mechanisms and molecules by which intracellular Na+ alters NMDA channel function, we examined the effects of Na+ applied directly to
the authoritative version for attribution.
                                                                                                                                                                                                                                         the cytoplasmic side of the membrane in inside-out patches. In contrast to the effects found in cell-attached recordings from intact neurons,
                                                                                                                                                                                                                                         applying 50 mM Na+ to the cytoplasmic face of inside-out patches did not change NMDA channel activity. We reasoned that the action of
                                                                                                                                                                                                                                         Na+ on NMDA receptors may depend on a molecule(s) lost from the excised patches or a biochemical process that had been disrupted.
                                                                                                                                                                                                                                         Because regulation of NMDA receptors by protein phosphorylation is well established, we considered the possibility that phosphorylation
                                                                                                                                                                                                                                         may be involved in the effects of intracellular Na+. To examine this, we used a broad-spectrum inhibitor of protein kinases, staurosporine,
                                                                                                                                                                                                                                         which was found to abolish the increase in NMDA channel activity caused by raising Na+ through bath-applying monensin during cell-
                                                                                                                                                                                                                                         attached patch experiments. Importantly, staurosporine did not prevent the rise in [Na+]i produced by the application of monensin. Thus, it
                                                                                                                                                                                                                                         was concluded that protein kinase activity may be required for the up-regulation of NMDA receptor activity by raising intracellular Na+.
                                                                                                                                                                                                                                              From these results, two mechanistic possibilities emerged: that the effect of Na+ is mediated by activating a kinase (or inhibiting a
                                                                                                                                                                                                                                         phosphatase) that is present in the cell but is lost from the patches, or alternatively, but not mutually exclusively, that the activity of a
                                                                                                                                                                                                                                         kinase is required for the effect of Na+ but is not directly a mediator. Because it had been established that endogenous Src could be up-
                                                                                                                                                                                                                                         regulated in the excised patches, we wondered whether this up-regulation would permit an effect of Na+. Indeed, this was found to be the
                                                                                                                                                                                                                                         case, because
                                                                                                                                                                                                                                         SRC, A MOLECULAR SWITCH GOVERNING GAIN CONTROL OF SYNAPTIC TRANSMISSION MEDIATED BY N-METHYL-D-                                            7702
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
ASPARTATE RECEPTORS
                                                                                                                                                                                                                                         applying Na+ to the cytoplasmic face increased NMDA channel activity in patches treated with the Src family-activating phosphopeptide
                                                                                                                                                                                                                                         EPQ(pY)EEIPIA ( Fig. 4 ). In contrast, the nonphosphorylated form of this peptide, which does not activate Src kinases, did not permit the
                                                                                                                                                                                                                                         effect of Na+. We also found that at concentrations well above those expected under physiological conditions, Na+ increased NMDA
                                                                                                                                                                                                                                         channel activity in the untreated inside-out patches ( 22 ). Thus, the Src-activating peptide shifted the concentration-response curve for Na+
                                                                                                                                                                                                                                         to the left.
                                                                                                                                                                                                                                           FIG. 4. NMDA channel activity is increased by raising the Na+ concentration on the cytoplasmic side of the membrane in inside-out
                                                                                                                                                                                                                                           patches during activation of Src-family kinases. A shows the inside-out recording configuration. (B) A continuous record of NMDA
                                                                                                                                                                                                                                           channel Po. The peptide EPQ(pY)EEIPIA was applied 35 min before the recording and throughout the recording period. Na+ (50
                                                                                                                                                                                                                                           mM) was applied as indicated. (C) Left, changes in Po versus [Na+] on the cytoplasmic side in the presence of the peptide EPQ(pY)
                                                                                                                                                                                                                                           EEIPIA; Right, effects of 50 mM Na+ on NMDA channels in control experiments and in the presence of the nonphosphorylated
                                                                                                                                                                                                                                           peptide EPQYEEIPIA. *, P < 0.05, Wilcoxon test or paired t test.
                                                                                                                                                                                                                                           FIG. 5. Diagram illustrating a working model for the regulation of NMDA channel gating by Src and Na +. See text for details.
                                                                                                                                                                                                                                              Therefore, from the convergence of evidence described above, we have developed a working model that the sensitivity of NMDA
                                                                                                                                                                                                                                         channels to intracellular Na+ is controlled by a channel-associated Src kinase. This model is represented diagrammatically in Fig. 5 . For
                                                                                                                                                                                                                                         simplicity of presentation, we represent the entire complex of NMDA channel subunits and associated proteins together as one pair of
                                                                                                                                                                                                                                         ovals forming the ion channel. This is not to imply that either the target of phosphorylation or the site of action of Na+ is necessarily one of
                                                                                                                                                                                                                                         the NMDA channel subunit proteins. Rather, as alluded to above, the target of phosphorylation remains to be determined, as does the site
                                                                                                                                                                                                                                         of action of Na+. Also, because the Src-activating peptide stimulates all Src-family kinases, it needs to be established whether the
                                                                                                                                                                                                                                         enhanced sensitivity to Na+ is caused by Src itself or another member of the family.
                                                                                                                                                                                                                                              Changing Src activity from the lowest to the highest level produces an 3- to 4-fold increase in NMDA channel function. Raising Na+
                                                                                                                                                                                                                                         from 0 to 40 mM produces an 1.5- to 2-fold change in NMDA channel activity. The change produced by Na+ is over and above that
                                                                                                                                                                                                                                         produced by Src and therefore, we estimate that the total range over which NMDA channel activity is regulated by Src and Na+ is about 5-
                                                                                                                                                                                                                                         to 8-fold. This degree of change would represent a dramatic alteration in synaptic efficacy and would be expected to have sizable effects
the authoritative version for attribution.
                                                                                                                                                                                                                                         on synaptic integration of individual neurons and on the behavior of neural networks. Moreover, because the influx of Ca2+ through
                                                                                                                                                                                                                                         NMDA receptors follows the amplitude of the currents (e.g., ref. 12 ), then a 5-fold increase in current would result in a 5-fold increase in
                                                                                                                                                                                                                                         Ca2+ influx. This boost in Ca2+ influx may then allow the resultant rise in the level of intracellular Ca2+ to be large enough to engage Ca2+-
                                                                                                                                                                                                                                         activated signaling pathways in the cell.
                                                                                                                                                                                                                                              We expect that the boost in NMDA channel function by coincidence of Src activation and raising [Na+]i may be relevant to the
                                                                                                                                                                                                                                         induction of lasting enhancement of synaptic efficacy in phenomena such as LTP. The role of Src in this process has already been
                                                                                                                                                                                                                                         established, but what about a role of Na+? High levels of action potential discharge, similar to those that have been shown to cause rises in
                                                                                                                                                                                                                                         [Na+]i, occur as a result of tetanic stimulation. Action potentials initiated in the soma are known to propagate into the dendrites ( 107 
                                                                                                                                                                                                                                         109 ), and this action potential backpropagation may participate in the induction of LTP ( 110 , 111 ). It is thought that the
                                                                                                                                                                                                                                         backpropagation of action potentials works by promoting Ca2+ entry
                                                                                                                                                                                                                                         SRC, A MOLECULAR SWITCH GOVERNING GAIN CONTROL OF SYNAPTIC TRANSMISSION MEDIATED BY N-METHYL-D-                                                      7703
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
ASPARTATE RECEPTORS
                                                                                                                                                                                                                                         through voltage-gated Ca2+ channels and by producing depolarization that relieves the Mg2+ block of NMDA channels. Our recent results,
                                                                                                                                                                                                                                         however, open up a new possibility, i.e., that it is the rise in intracellular Na+ that is the important event. This rise in Na+, when coincident
                                                                                                                                                                                                                                         with postsynaptic activation of Src, may lead to a large amplification of NMDA receptor function sufficiently large to set off the rest of the
                                                                                                                                                                                                                                         intracellular signaling cascade.
                                                                                                                                                                                                                                               What about implications for pain? Excitatory synaptic transmission in nociceptive pathways in the spinal cord is facilitated by
                                                                                                                                                                                                                                         stimulation of C fiber nociceptors ( 112  114 ), apparently as a result of increased responsiveness of NMDA receptors ( 6 , 11 ). It is not
                                                                                                                                                                                                                                         yet known whether Src is activated under such circumstances, but it seems likely given that Src can be activated through a number of
                                                                                                                                                                                                                                         signaling pathways, such as stimulating G protein-coupled receptors and receptor tyrosine kinases ( 91 , 92 ), and these types of pathways
                                                                                                                                                                                                                                         have been implicated in enhancement of nociceptive transmission ( 115 , 116 ). Moreover, the discharge rate of central nociceptive
                                                                                                                                                                                                                                         neurons responding to noxious inputs is sufficiently high as to be expected to produce dramatic rises in [Na+]i. Therefore, it is possible that
                                                                                                                                                                                                                                         the up-regulation of NMDA receptors by Src and sodium is involved in the central up-regulation of transmission in nociceptive pathways.
                                                                                                                                                                                                                                         This may be relevant to human pain states because this central up-regulation appears to be sufficient to produce hyperalgesia and allodynia
                                                                                                                                                                                                                                         (e.g., ref. 117 ).
                                                                                                                                                                                                                                                                                                      CONCLUSIONS
                                                                                                                                                                                                                                               The gene encoding Src was found over 20 years ago as the first protooncogene, with particular Src mutations causing cancer ( 118 ).
                                                                                                                                                                                                                                         Since then, this enzyme has been found to have many roles in cell signaling in a variety of cell types. In neurons, Src up-regulates the
                                                                                                                                                                                                                                         function of NMDA receptors and thereby gates the induction of a lasting enhancement of synaptic transmission, LTP in the CA1 region of
                                                                                                                                                                                                                                         the hippocampus. Src also sensitizes NMDA channels to up-regulation by intracellular Na+. Thus, the coincidence of Src activation and a
                                                                                                                                                                                                                                         rise in [Na+]i may be important for boosting synaptic NMDA receptor function and initiating the intracellular signaling cascades that
                                                                                                                                                                                                                                         produce persistent alterations in synaptic function. Because NMDA receptors are implicated in a variety of pathophysiological conditions
                                                                                                                                                                                                                                         in the CNS, the regulation by Src and Na+ represent potential targets for developing new types of therapeutic intervention in a variety of
                                                                                                                                                                                                                                         CNS disorders.
                                                                                                                                                                                                                                               We thank Yueqiao Huang and Jeff Gingrich for helpful comments on the manuscript. Work of the authors is supported by the
                                                                                                                                                                                                                                         Medical Research Council of Canada and the Nicole Fealdman Memorial Fund. M.W.S. in an MRC Scientist and X.M.Y. is an MRC
                                                                                                                                                                                                                                         Scholar.
                                                                                                                                                                                                                                         1. Dubner, R. & Ruda, M. A. ( 1992 ) Trends Neurosci. 15 , 96103 .
                                                                                                                                                                                                                                         2. Levine, J. D. ( 1998 ) Neuron 20 , 649654 .
                                                                                                                                                                                                                                         3. Woolf, C. J. & Doubell, T. P. ( 1994 ) Curr. Opin. Neurobiol. 4 , 525534 .
                                                                                                                                                                                                                                         4. Chiang, C. Y. , Park, S. J. , Kwan, C. L. , Hu, J. W. & Sessle, B. J. ( 1998 ) J. Neurophysiol. 80 , 26212631 .
                                                                                                                                                                                                                                         5. Yu, X. M. , Sessle, B. J. , Haas, D. A. , Izzo, A. , Vernon, H. & Hu, J. W. ( 1996 ) Pain 68 , 169178 .
                                                                                                                                                                                                                                         6. Woolf, C. J. & Thompson, S. W. N. ( 1991 ) Pain 44 , 293299 .
                                                                                                                                                                                                                                         7. Neugebauer, V. , Lucke, T. & Schaible, H. G. ( 1993 ) J. Neurophysiol. 70 , 13651377 .
                                                                                                                                                                                                                                         8. Dickenson, A. H. , Chapman, V. & Green, G. M. ( 1997 ) Gen. Pharmacol. 28 , 633638 .
                                                                                                                                                                                                                                         9. Dickenson, A. H. & Sullivan, A. F. ( 1987 ) Neuropharmacology 26 , 12351238 .
                                                                                                                                                                                                                                         10. Ma, Q. P. & Woolf, C. J. ( 1995 ) Pain 61 , 383390 .
                                                                                                                                                                                                                                         11. Liu, X. G. & Sandkuhler, J. ( 1995 ) Neurosci. Lett. 191 , 4346 .
                                                                                                                                                                                                                                         12. Wang, Y. T. & Salter, M. W. ( 1994 ) Nature (London) 369 , 233235 .
                                                                                                                                                                                                                                         13. Fadool, D. A. , Holmes, T. C. , Berman, K. , Dagan, D. & Levitan, I. B. ( 1997 ) J. Neurophysiol. 78 , 15631573 .
                                                                                                                                                                                                                                         14. Cataldi, M. , Taglialatela, M. , Guerriero, S. , Amoroso, S. , Lombardi, G. , Di Renzo, G. & Annunziato, L. ( 1996 ) J. Biol. Chem 271 , 94419446 .
                                                                                                                                                                                                                                         15. Moss, S. J. , Gorrie, G. H. , Amato, A. & Smart, T. G. ( 1995 ) Nature (London) 377 , 344348 .
                                                                                                                                                                                                                                         16. Lev, S. , Moreno, H. , Martinez, R. , Canoll, P. , Peles, E. , Musacchio, J. M. , Plowman, G. D. , Rudy, B. & Schlessinger, J. ( 1995 ) Nature
                                                                                                                                                                                                                                              (London) 376 , 737745 .
                                                                                                                                                                                                                                         17. Yu, X. M. , Askalan, R. , Keil, G. J. & Salter, M. W. ( 1997 ) Science 275 , 674678 .
                                                                                                                                                                                                                                         18. Brown, M. T. & Cooper, J. A. ( 1996 ) Biochim. Biophys. Acta 1287 , 121149 .
                                                                                                                                                                                                                                         19. Brugge, J. S. , Cotton, P. C. , Queral, A. E. , Barrett, J. N. , Nonner, D. & Keane, R. W. ( 1985 ) Nature (London) 316 , 554557 .
                                                                                                                                                                                                                                         20. Sugrue, M. M. , Brugge, J. S. , Marshak, D. R. , Greengard, P. & Gustafson, E. L. ( 1990 ) J. Neurosci. 10 , 25132527 .
                                                                                                                                                                                                                                         21. Lu, Y. M. , Roder, J. C. , Davidow, J. & Salter, M. W. ( 1998 ) Science 279 , 13631368 .
                                                                                                                                                                                                                                         22. Yu, X.-M. & Salter, M. W. ( 1998 ) Nature (London) 396 , 469474 .
                                                                                                                                                                                                                                         23. McBain, C. J. & Mayer, M. L. ( 1994 ) Physiol. Rev. 74 , 723760 .
                                                                                                                                                                                                                                         24. Gasic, G. P. & Hollmann, M. ( 1992 ) Annu. Rev. Physiol. 54 , 507536 .
                                                                                                                                                                                                                                         25. Seeburg, P. H. ( 1993 ) Trends Neurosci. 16 , 359365 .
                                                                                                                                                                                                                                         26. Raymond, L. A. , Blackstone, C. D. & Huganir, R. L. ( 1993 ) Trends Neurosci. 16 , 147153 .
                                                                                                                                                                                                                                         27. Benveniste, M. & Mayer, M. L. ( 1991 ) Biophys. J. 59 , 560573 .
                                                                                                                                                                                                                                         28. Johnson, J. W. & Ascher, P. ( 1987 ) Nature (London) 325 , 529531 .
                                                                                                                                                                                                                                         29. Hirai, H. , Kirsch, J. , Laube, B. , Betz, H. & Kuhse, J. ( 1996 ) Proc. Natl. Acad. Sci. USA 93 , 60316036 .
                                                                                                                                                                                                                                         30. Ivanovic, A. , Reilander, H. , Laube, B. & Kuhse, J. ( 1998 ) J. Biol. Chem. 273 , 1993319937 .
                                                                                                                                                                                                                                         31. Laube, B. , Hirai, H. , Sturgess, M. , Betz, H. & Kuhse, J. ( 1997 ) Neuron 18 , 493503 .
                                                                                                                                                                                                                                         32. Sucher, N. J. , Akbarian, S. , Chi, C. L. , Leclerc, C. L. , Awobuluyi, M. , Deitcher, D. L. , Wu, M. K. , Yuan, J. P. , Jones, E. G. & Lipton, S. A. (
                                                                                                                                                                                                                                              1995 ) J. Neurosci. 15 , 65096520 .
                                                                                                                                                                                                                                         33. Ciabarra, A. M. , Sullivan, J. M. , Gahn, L. G. , Pecht, G. , Heinemann, S. & Sevarino, K. A. ( 1995 ) J. Neurosci. 15 , 64986508 .
                                                                                                                                                                                                                                         34. Mayer, M. L. & Westbrook, G. L. ( 1987 ) J. Physiol. (London) 394 , 501527 .
                                                                                                                                                                                                                                         35. Ascher, P. & Nowak, L. ( 1988 ) J. Physiol. (London) 399 , 247266 .
                                                                                                                                                                                                                                         36. Traynelis, S. F. & Cull-Candy, S. G. ( 1990 ) Nature (London) 345 , 347350 .
                                                                                                                                                                                                                                         37. Christine, C. W. & Choi, D. W. ( 1990 ) J. Neurosci. 10 , 108116 .
                                                                                                                                                                                                                                         38. Hollmann, M. , Boulter, J. , Maron, C. , Beasley, L. , Sullivan, J. , Pecht, G. & Heinemann, S. ( 1993 ) Neuron 10 , 943954 .
                                                                                                                                                                                                                                         39. McGurk, J. F. , Bennett, M. V. L. & Zukin, R. S. ( 1990 ) Proc. Natl. Acad. Sci. USA 87 , 99719974 .
                                                                                                                                                                                                                                         40. Aizenman, E. , Lipton, S. A. & Loring, R. H. ( 1989 ) Neuron 2 , 12571263 .
                                                                                                                                                                                                                                         41. Rosenmund, C. & Westbrook, G. L. ( 1993 ) Neuron 10 , 805814 .
the authoritative version for attribution.
                                                                                                                                                                                                                                         42. MacDonald, J. F. , Mody, I. & Salter, M. W. ( 1989 ) J. Physiol. (London) 414 , 1734 .
                                                                                                                                                                                                                                         43. Chen, L. & Huang, L.-Y. M. ( 1991 ) Neuron 7 , 319326 .
                                                                                                                                                                                                                                         44. Chen, L. & Huang, L. Y. M. ( 1992 ) Nature (London) 356 , 521523 .
                                                                                                                                                                                                                                         45. Kelso, S. R. , Nelson, T. E. & Leonard, J. P. ( 1992 ) J. Physiol. (London) 449 , 705718 .
                                                                                                                                                                                                                                         46. Durand, G. M. , Gregor, P. , Zheng, X. , Bennett, M. V. L. , Uhl, G. R. & Zukin, R. S. ( 1992 ) Proc. Natl. Acad. Sci. USA 89 , 93599363 .
                                                                                                                                                                                                                                         47. Tingley, W. G. , Roche, K. W. , Thompson, A. K. & Huganir, R. L. ( 1993 ) Nature (London) 364 , 7073 .
                                                                                                                                                                                                                                         48. Sigel, E. , Baur, R. & Malherbe, P. ( 1994 ) J. Biol. Chem. 269 , 82048208 .
                                                                                                                                                                                                                                         49. Wang, L.-Y. , Orser, B. A. , Brautigan, D. L. & MacDonald, J. F. ( 1994 ) Nature (London) 369 , 230232 .
                                                                                                                                                                                                                                         50. Raman, I. M. , Tong, G. & Jahr, C. E. ( 1996 ) Neuron 16 , 415421 .
                                                                                                                                                                                                                                         51. Lieberman, D. N. & Mody, I. ( 1994 ) Nature (London) 369 , 235239 .
                                                                                                                                                                                                                                         SRC, A MOLECULAR SWITCH GOVERNING GAIN CONTROL OF SYNAPTIC TRANSMISSION MEDIATED BY N-METHYL-D-                                                       7704
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
ASPARTATE RECEPTORS
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                         differential results include (i) influences of cognitive modulation in S1 activity; (ii) averaging-related degradation of the signal because of
                                                                                                                                                                                                                                         variability of sulcal anatomy; (iii) a possible combination of excitatory and inhibitory effects of nociceptive input to S1; and (iv)
                                                                                                                                                                                                                                         differences in statistical analyses and power.
                                                                                                                                                                                                                                         Table 1. Methods and results of brain imaging studies
                                                                                                                                                                                                                                         Study              Ref.     Modality        Subject                 n     Stimulation device         Stimulation                S1
                                                                                                                                                                                                                                         Talbot et al.      12       PET H215O       Healthy                 8     Thermode 1 cm2             42, 4748C                Yes
                                                                                                                                                                                                                                         Jones et al.       13       PET 15CO2       Healthy                 6     Thermode 2.5 cm           36.1, 41.3, and 46.4C     No
                                                                                                                                                                                                                                                                                                                   5.0 cm
                                                                                                                                                                                                                                         Apkarian et al.      14      SPECT            Healthy                    Water bath                 Moderate heat pain         Inhibition
                                                                                                                                                                                                                                         Crawford et al.      24      PET 131Xe        Healthy               11    Tourniquet                 Ischemia                   Yes
                                                                                                                                                                                                                                         Coghill et al.       25      PET H215O        Healthy               9     Thermode, 1 cm2            34, 4748C                Yes
                                                                                                                                                                                                                                         Di Piero et al.      26      SPET 133Xn       Healthy               7     Water bath                 Cold pressor test          Yes
                                                                                                                                                                                                                                         Derbyshire et al.    27      PET H215O        Facial pain           6     Thermode                   Ramp, 2543C              No
                                                                                                                                                                                                                                                                                       Healthy               7
                                                                                                                                                                                                                                         Rosen et al.         28      PET H215O        Angina pectoris       12    Dobutamine infusion        Angina                     No
                                                                                                                                                                                                                                         Hsieh et al.         29      PET H215O        Neuropathic           8     None                       Spontaneous pain           No
                                                                                                                                                                                                                                         Hsieh et al.         30      PET H215O        Healthy               4     Intracutaneous             Ethanol                    Yes
                                                                                                                                                                                                                                                                                                                   injection
                                                                                                                                                                                                                                         Davis et al.         31      fMRI             Healthy               9     Electrical nerve           Median nerve, 50 Hz        Yes
                                                                                                                                                                                                                                                                                                                   stimulator
                                                                                                                                                                                                                                         Weiller et al.       32      PET H215O        Migraine patients     9     None                       Spontaneous migraine       No
                                                                                                                                                                                                                                         Howland et al.       33      MEG              Healthy               5     Electrical nerve           Electric finger shock      Yes
                                                                                                                                                                                                                                                                                                                   stimulator
                                                                                                                                                                                                                                         Kitamura et al.      34      MEG              Healthy               5     Electrical nerve           Electric finger shock      Yes
                                                                                                                                                                                                                                                                                                                   stimulator
                                                                                                                                                                                                                                         Craig et al.         35      PET H215O        Healthy               11    Thermal grill              Bars 20 and 40C           Yes
                                                                                                                                                                                                                                         Casey et al.         36      PET H215O        Healthy               9     Thermode                   40 and 50C                Yes
                                                                                                                                                                                                                                         Casey et al.         36      PET H215O        Healthy               9     Thermode                   6, 20C                    Yes
                                                                                                                                                                                                                                         Hsieh et al.         37      PET H215O        Cluster headache      7     Sublinguial                Headache                   No
                                                                                                                                                                                                                                                                                                                   nitroglycerin
                                                                                                                                                                                                                                         Andersson et al.     21      PET H215O        Healthy               6     Capsaicin                  Intracutaneous             Yes
                                                                                                                                                                                                                                         Antognini et al.     38      fMRI             Healthy               5     Electrical stimulator      Electric hand shock        Yes
                                                                                                                                                                                                                                         Aziz et al.          39      PET H215O        Healthy               8     Balloon                    Esophageal distension      Yes
                                                                                                                                                                                                                                                                                       Cluster headache      7
                                                                                                                                                                                                                                         DiPiero et al.       40      SPECT            Healthy               12    Water bath                 Cold pressor               Yes
                                                                                                                                                                                                                                         Rainville et al.     17      PET H215O        Healthy               11    Water bath                 35, 47C                  Yes
                                                                                                                                                                                                                                         Silverman et al.     41      PET H215O        Healthy               6     Balloon                    Rectal distension          No
                                                                                                                                                                                                                                         Svensson et al.      42      PET H215O        Healthy               11    CO2 laser                  Cutaneous                  Yes, ns
                                                                                                                                                                                                                                         Svensson et al.      43      PET H215O        Healthy               11    Electrical stimulator      Intramuscular              Yes, ns
                                                                                                                                                                                                                                         Xu et al.            44      PET H215O        Healthy               6     CO2 laser                  Cutaneous                  Yes
                                                                                                                                                                                                                                         Binkofski et al.     45      fMRI             Healthy               5     Balloon                    Esophageal distension      Yes
                                                                                                                                                                                                                                         Derbyshire et al.    46      PET H215O        Healthy               6     Thermode                   Ramp, 2543C              No
                                                                                                                                                                                                                                         Iadarola et al.      47      PET H215O        Healthy                    Capsaicin                  Subcutaneous arm           No
                                                                                                                                                                                                                                         May et al.           48      PET H215O        Healthy               7     Capsaicin                  Subcutaneous               No
                                                                                                                                                                                                                                                                                                                                              forehead
                                                                                                                                                                                                                                         Oshiro et al.        49      fMRI             Healthy               6     Electrical stimulator      Finger, 8 Hz               Yes
                                                                                                                                                                                                                                         Paulson et al.       50      PET H215O        Healthy               10    Thermode, 254 mm2          40 & 50C                  No
                                                                                                                                                                                                                                         ns, not significant; SPECT, single photon-emission computed tomography; fMRI, functional MRI; MEG, magnetoencephalographic
                                                                                                                                                                                                                                         imaging.
                                                                                                                                                                                                                                               Cognitive Modulation of S1 Activity. As proposed by Jones and colleagues ( 15 , 16 ), S1 pain-related activation is highly
                                                                                                                                                                                                                                         modulated by cognitive factors that alter pain perception, including attention and previous experience. In our laboratory, we have shown
                                                                                                                                                                                                                                         that when the subjects attention is directed away from a painful stimulus, the activity of S1 cortex is dramatically reduced (B.C., P.
                                                                                                                                                                                                                                         Rainville, T. Paus, G.H.D., and M.C.B., unpublished observations). Fig. 1 shows the results of this study, in which we used PET H215O
                                                                                                                                                                                                                                         bolus methods to measure regional cerebral blood flow (rCBF) in nine subjects while they discriminated changes in thermal intensity or
                                                                                                                                                                                                                                         auditory frequency. During all scans, concurrent sequences of tones and contact heat stimuli (pain, 46.548.5C or warm, 3238C on the
                                                                                                                                                                                                                                         left arm) were presented. After each scan, subjects used a 100-mm visual analogue scale to rate the perceived level of pain associated with
                                                                                                                                                                                                                                         the thermal stimuli. Statistical brain maps of pain-related activity, i.e., rCBF during the pain condition minus rCBF during the nonpainful
                                                                                                                                                                                                                                         warm condition, were obtained. Pain intensity was rated higher in the thermal than in the auditory task (50.4 vs. 41.4, P = 0.01), indicating
                                                                                                                                                                                                                                         that pain perception was modulated by the attentional demands of the discrimination tasks. Likewise, whereas in the thermal task there
                                                                                                                                                                                                                                         was a significant pain-related rCBF increase within S1 (t = 4.42, P < 0.01), there was no significant change in pain-related rCBF within S1
                                                                                                                                                                                                                                         during the auditory task ( Fig. 1 ). A direct comparison of pain-related S1 activity during the pain and auditory tasks showed that pain-
                                                                                                                                                                                                                                         evoked rCBF was significantly larger in the thermal than in the auditory task (t = 3.92; P < 0.01). In this experiment, the behavioral task
the authoritative version for attribution.
                                                                                                                                                                                                                                         used to direct attention toward the thermal stimuli involved the detection of a small change in the intensity of the heat stimulus. This task
                                                                                                                                                                                                                                         probably served to specifically direct the subjects attention to sensory aspects of the pain, rather than to the unpleasantness or suffering.
                                                                                                                                                                                                                                               Other data from our laboratory also support the idea that attention to sensory aspects of the pain experience can alter S1 activity. By
                                                                                                                                                                                                                                         using hypnosis, we found that suggestions specifically directed toward increasing or decreasing the perceived intensity of the burning pain
                                                                                                                                                                                                                                         sensation produced by submerging a subjects hand in painfully hot water modulated pain-related activity in S1 (R.K.H., P. Rainville,
                                                                                                                                                                                                                                         G.H.D., and M.C.B., unpublished observations). In contrast, suggestions
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         directed toward changing the unpleasantness of the pain had no effect on pain-related activity in S1, but produced instead a robust
                                                                                                                                                                                                                                         modulation of activity in anterior cingulate cortex directly correlated with the subjects perception of unpleasantness (ANCOVA, P =
                                                                                                                                                                                                                                         0.005) ( 17 ).
                                                                                                                                                                                                                                           FIG. 1. Pain-related activity when attention is directed to the painful heat stimulus (Left) or to an auditory stimulus (Center) is
                                                                                                                                                                                                                                           revealed by subtracting PET data recorded when a warm stimulus (3238C) was presented from those recorded when a painfully hot
                                                                                                                                                                                                                                           stimulus (46.548.5C) was presented during each attentional state. Differences in pain-related activity during the two attentional
                                                                                                                                                                                                                                           conditions are revealed (Right) by subtracting PET data recorded during the auditory task from that recorded during the heat-
                                                                                                                                                                                                                                           discrimination task (using only painful stimulus trials46.548.5C). PET data, averaged across nine subjects, are illustrated against
                                                                                                                                                                                                                                           an MRI from one subject. Horizontal and coronal slices through S1 are centered at the activation peaks. Red circles surround the
                                                                                                                                                                                                                                           region of S1. Whereas there was a significant activation of S1 when subjects attended to the painful stimulus (Left), there was no
                                                                                                                                                                                                                                           significant activation when subjects attended to the auditory stimulus (Center). However, there was a subsignificant activation in S1
                                                                                                                                                                                                                                           during the auditory task, as shown in the Inset. The direct comparison of pain in the two attentional conditions (Right) shows a
                                                                                                                                                                                                                                           significant difference in pain-related S1 activity during the two attentional states.
                                                                                                                                                                                                                                               In these hypnosis experiments, we also found evidence that experience with the hypnotic suggestions may have produced long-term
                                                                                                                                                                                                                                         changes in the subjects neural processing of pain. At least a week before participating in a PET scanning session, all subjects received the
                                                                                                                                                                                                                                         same hypnotic induction, suggestions, and painful stimuli that were to be used during the scanning experiment. In subsequent PET
                                                                                                                                                                                                                                         sessions, two scans using the painful heat and two using the nonpainful warm control stimulus were performed before the subjects
                                                                                                                                                                                                                                         underwent hypnotic induction and suggestions. During these four scans, the subjects were simply instructed to relax and attend to the
                                                                                                                                                                                                                                         thermal stimulusa control situation for identifying regions that could be examined for modulation related to hypnotic suggestions given
                                                                                                                                                                                                                                         in subsequent scans. Although subjects in the two hypnosis experiments produced similar ratings of pain intensity and unpleasantness
                                                                                                                                                                                                                                         during these control scans, those previously trained to attend to the intensity of the painful stimuli showed substantially greater pain-
                                                                                                                                                                                                                                         related activity in S1 than did those who had been trained to attend to the unpleasantness of those stimuli [ Fig. 2 Upper (t = 5.05); Lower
                                                                                                                                                                                                                                         (17) (t = 3.01)].
                                                                                                                                                                                                                                               Attentional modulation within S1 cortex is not restricted to pain-related activity. Other investigators have found that rCBF in S1,
                                                                                                                                                                                                                                         evoked by tactile stimuli, is reduced when subjects attend to another stimulus modality ( 18 ). Similarly, neuronal recordings in S1 cortex
                                                                                                                                                                                                                                         of trained monkeys reveal low-threshold neurons whose activity is enhanced by attention to the tactile stimulus ( 19 , 20 ). Despite the
                                                                                                                                                                                                                                         extensive nature of attentional modulation of S1 activity, there is little evidence that attention activates S1 neurons without the concurrent
                                                                                                                                                                                                                                         presence of sensory-evoked activation. Anticipation of a painful stimulus has been shown to produce decreases in S1 rCBF ( 51 ) rather
                                                                                                                                                                                                                                         than increases in rCBF that would reflect excitatory neuronal activity.
                                                                                                                                                                                                                                               Variability of S1 Sulcal Anatomy. Both monkey and human data indicate that nociceptive activity in S1 cortex is somatotopically
                                                                                                                                                                                                                                         organized. Neuronal recording studies in monkeys show a somatotopic organization for nociceptive neurons similar to that observed for
                                                                                                                                                                                                                                         low-threshold cells ( 3 ). Similarly, by using PET to measure rCBF and capsaicin as a specific nociceptive stimulus, Andersson et al. ( 21 )
                                                                                                                                                                                                                                         found distinct activation sites related to foot and hand pain, consistent with the known topographic organization of cutaneous receptive
                                                                                                                                                                                                                                         fields within S1. The somatotopic organization of S1 cortex probably results in a small focal activation evoked by a localized pain
                                                                                                                                                                                                                                         stimulus. Such focal activation is more easily observed with single-subject functional MRI studies than in PET studies involving a number
                                                                                                                                                                                                                                         of subjects and data smoothing. Fig. 3 shows the focal activation produced in S1 cortex of three individual subjects when the leg was
                                                                                                                                                                                                                                         stimulated with noxious heat (B.H., J.-I.C, B. Pike, G.H.D., and M.C.B., unpublished observations). The images show regions activated
                                                                                                                                                                                                                                         during stimulation with painful heat, as compared with that observed during nonpainful warm. Fig. 3 also shows that the pain-related S1
                                                                                                                                                                                                                                         activation sites, although on the posterior bank of the central sulcus in all subjects, varied in terms of their stereotaxic coordinates,
                                                                                                                                                                                                                                         suggesting small intersubject differences in the localization of pain-related activity. This anatomical variability, although not of a large
                                                                                                                                                                                                                                         magnitude, degrades a focal signal when data are averaged across subjects. Thus, because of the somatotopically organized focal
                                                                                                                                                                                                                                         activation observed in S1, the rCBF signal arising from this area may be particularly susceptible to degradation when averaging data.
                                                                                                                                                                                                                                               Inhibitory Effects of Noxious Stimuli in S1 activity. Tommerdahl et al. ( 11 ) found in monkey S1 cortex that the presence of
the authoritative version for attribution.
                                                                                                                                                                                                                                         noxious heat reduced the intrinsic optical-imaging signal evoked by low-threshold mechanical stimulation of the skin.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         These data are consistent with the findings of Apkarian et al. ( 14 ), which showed a decrease in blood flow to S1 cortex in human subjects
                                                                                                                                                                                                                                         during the presentation of a tonic heat stimulus. Consonant with the idea that noxious stimulation produces inhibition of tactile sensitivity
                                                                                                                                                                                                                                         in S1 cortex are psychophysical data showing that the presence of pain reduces tactile perception ( 22 ).
                                                                                                                                                                                                                                           FIG. 2. Changes in pain-related activity associated with previous hypnotic training by using suggestions for modulating pain
                                                                                                                                                                                                                                           sensation (Upper) or pain unpleasantness (Lower). Both images represent data from control scans, in which no hypnotic suggestions
                                                                                                                                                                                                                                           were given. Each image represents the subtraction of PET data recorded when the hand was submerged in thermally neutral water
                                                                                                                                                                                                                                           (35C) from data recorded when the hand was submerged in painfully hot water (47C). PET data were averaged across 10
                                                                                                                                                                                                                                           experimental sessions in the sensory study (Upper) and, in a different group of subjects, 11 experimental sessions in the affective
                                                                                                                                                                                                                                           study (Lower). The PET data are illustrated against the average MRI for that subject group. Coronal slices through S1 are centered at
                                                                                                                                                                                                                                           the activation peaks, and red circles surround the region of S1.
                                                                                                                                                                                                                                              Other evidence suggests that the inhibition of S1 tactile activity by noxious stimuli may take place at lower levels of the neuraxis
                                                                                                                                                                                                                                         rather than through a direct inhibitory influence at the level of S1. In awake monkeys, the spontaneous activity of low-threshold neurons in
                                                                                                                                                                                                                                         the ventroposterior thalamus is inhibited by topical application of capsaicin, which specifically excites C fibers (C.-C. Chen and M.C.B.,
                                                                                                                                                                                                                                         unpublished observations). Similarly, capsaicin sometimes reduces the responses of spinothalamic tract neurons to noxious heat
                                                                                                                                                                                                                                         stimulation ( 23 ). Thus, when a painful stimulus is presented in a human brain imaging study, the net effect of exciting some neurons and
                                                                                                                                                                                                                                         inhibiting the spontaneous activity of others could have different effects on rCBF (as measured by PET) or on venous blood oxygenation
                                                                                                                                                                                                                                         (as measured by functional MRI), depending on such variables as timing, duration, location, and intensity of the painful stimulus.
                                                                                                                                                                                                                                              Procedural and Analytical Differences in Studies. In human brain imaging studies, many procedural variables can influence the
                                                                                                                                                                                                                                         resultant data. Analytical techniques are not standardized across laboratories or across imaging methods. For example, different
                                                                                                                                                                                                                                         approaches are used to compare stimulation conditions, including subtraction and regression comparisons across scans. Instructions to the
                                                                                                                                                                                                                                         subjects, which can influence the cognitive state, vary among studies, as does the timing of stimulus variables. The statistical analyses,
                                                                                                                                                                                                                                         including methods for calculating variance and assumptions about the nature of the data, also differ among laboratories. Although all
                                                                                                                                                                                                                                         analyses rely on some type of statistical determination of significance, the method of accounting for multiple comparisons varies, and thus
                                                                                                                                                                                                                                         the criteria for identifying an activation as significant are not uniform across studies. Finally, the power of any statistical test is influenced
                                                                                                                                                                                                                                         by the number of subjects studied, which is another factor that varies greatly among studies.
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 3. Functional MRI data from three subjects, using a 1.5-T scanner and standard head coil. Each horizontal and coronal image
                                                                                                                                                                                                                                           represents the anatomical and functional data from a single subject during one session, which included a high-resolution anatomical
                                                                                                                                                                                                                                           scan and five to eight runs of 120 whole-brain functional MRI scans ( 10  13 ) 7-mm slices acquired at 3-sec intervals. Thermal
                                                                                                                                                                                                                                           stimuli were applied to the left calf on separate runs. Thermal runs consisted of 9 s alternating cycles of rest, painful (4546C), rest,
                                                                                                                                                                                                                                           and neutral (35 36C) stimulation by using a 9 cm2 thermode. Activation maps were generated by using Spearmans rank order
                                                                                                                                                                                                                                           correlation, comparing painful to neutral heat. The coronal and horizontal slices through S1 are centered at the activation peaks, and
                                                                                                                                                                                                                                           red circles surround the region of S1.
                                                                                                                                                                                                                                              As with any statistical test, the interpretation of negative results must be performed with caution. Thus, it would appear more fruitful
                                                                                                                                                                                                                                         to identify regions that show activation across a
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         number of pain studies than to rely on the data of any one study in isolation. Despite the wide methodological and analytical variation in
                                                                                                                                                                                                                                         human brain imaging studies of pain, there is surprising consistency in the activation of a number of brain regions, including anterior
                                                                                                                                                                                                                                         cingulate and insular cortices. Although the observation of pain-related activation in S1 is somewhat less consistent, the fact that at least
                                                                                                                                                                                                                                         half of the human brain imaging studies have identified significant activation of this region when subjects perceive pain suggests that S1
                                                                                                                                                                                                                                         has a significant role in nociceptive processing.
                                                                                                                                                                                                                                               What Is the Role of S1 in Pain Processing? Anatomical, neurophysiological, and imaging data confirm a role of S1 cortex in pain
                                                                                                                                                                                                                                         processing. Overall, the findings support the traditional view that S1 is primarily involved in discriminative aspects of somatic sensation
                                                                                                                                                                                                                                         and extends this view to include discriminative aspects of somatic stimulation that is potentially tissue-damaging, e.g., painful. Single
                                                                                                                                                                                                                                         neurons in monkey S1 code stimulus intensity, location, and duration, and their activity correlates with human perception. Human imaging
                                                                                                                                                                                                                                         studies show activation of S1 by a range of noxious stimuli, including capsaicin, which selectively activates C fibers. These studies also
                                                                                                                                                                                                                                         confirm the somatotopic organization of S1 pain responses, thus supporting the role of S1 in pain localization. Other imaging data that
                                                                                                                                                                                                                                         implicate S1 in the sensory aspect of pain perception are findings that S1 activation is modulated by cognitive manipulations that alter
                                                                                                                                                                                                                                         perceived pain intensity but not by manipulations that alter unpleasantness, independent of pain intensity. Nevertheless, despite the
                                                                                                                                                                                                                                         probable role of S1 in the encoding of the various sensory features of pain, a considerable amount of evidence suggests that nociceptive
                                                                                                                                                                                                                                         input to S1 may also serve to modulate tactile perception, described by Apkarian et al. as a touch gate ( 22 ). Thus, S1 cortex may be
                                                                                                                                                                                                                                         involved in both perception and modulation of both painful and nonpainful somatosensory sensations.
                                                                                                                                                                                                                                               We wish to express our appreciation to Ms. Francine Blanger for her help in preparing this manuscript and to Dr. Pierre Rainville for
                                                                                                                                                                                                                                         his intellectual and technical contributions to these studies. Imaging studies were performed at the Montreal Neurological Institute with the
                                                                                                                                                                                                                                         help and expertise of the its staff. This research is supported by operating grants from the Canadian Medical Research Council awarded to
                                                                                                                                                                                                                                         M.C.B. and to G.H.D.
                                                                                                                                                                                                                                         1. Head, H. & Holmes, G. ( 1911 ) Brain 34 , 102254 .
                                                                                                                                                                                                                                         2. Penfield, W. & Boldrey, E. ( 1937 ) Brain 60 , 389443 .
                                                                                                                                                                                                                                         3. Kenshalo, D. R., Jr. , & Isensee, O. ( 1983 ) J. Neurophysiol. 50 , 14791496 .
                                                                                                                                                                                                                                         4. Kenshalo, D. R., Jr. , Chudler, E. H. , Anton, F. & Dubner, R. ( 1988 ) Brain Res. 454 , 378382 .
                                                                                                                                                                                                                                         5. White, J. C. & Sweet, W. H. ( 1969 ) Pain and the Neurosurgeon: A Forty-Year Experience ( Thomas , Springfield, IL ).
                                                                                                                                                                                                                                         6. Young, G. B. & Blume, W. T. ( 1983 ) Brain 106 , 537554 .
                                                                                                                                                                                                                                         7. Gingold, S. I. , Greenspan, J. D. & Apkarian, A. V. ( 1991 ) J. Comp. Neurol. 308 , 467490 .
                                                                                                                                                                                                                                         8. Kenshalo, D. R., Jr. , Giesler, G. J. , Leonard, R. B. & Willis, W. D. ( 1980 ) J. Neurophysiol. 43 , 15941614 .
                                                                                                                                                                                                                                         9. Rausell, E. & Jones, E. G. ( 1991 ) J. Neurosci. 11 , 210225 .
                                                                                                                                                                                                                                         10. Chudler, E. H. , Anton, F. , Dubner, R. & Kenshalo, D. R. , Jr. ( 1990 ) J. Neurophysiol. 63 , 559569 .
                                                                                                                                                                                                                                         11. Tommerdahl, M. , Delemos, K. A , Vierck, C. J., Jr. , Favorov, O. V. & Whitsel, B. L. ( 1996 ) J. Neurophysiol. 75 , 26622670 .
                                                                                                                                                                                                                                         12. Talbot, J. D. , Marrett, S. , Evans, A. C. , Meyer, E. , Bushnell, M. C. & Duncan, G. H. ( 1991 ) Science 251 , 13551358 .
                                                                                                                                                                                                                                         13. Jones, A K. P. , Brown, W. D. , Friston, K. J. , Qi, L. Y. & Frackowiak, R. S. J. ( 1991 ) Proc. R. Soc. London Ser. B 244 , 3944 .
                                                                                                                                                                                                                                         14. Apkarian A. V. , Stea R. A , Manglos S. H. , Szeverenyi N. M. , King R. B. & Thomas F. D. ( 1992 ) Neurosci. Lett. 140 , 141147 .
                                                                                                                                                                                                                                         15. Jones, A. K. P. , Friston, K. & Frackowiak, R. S. J. ( 1992 ) Science 255 , 215215 .
                                                                                                                                                                                                                                         16. Jones, A. K. P. & Derbyshire, S. W. G. ( 1996 ) Ann. Rheum. Dis. 55 , 411420 .
                                                                                                                                                                                                                                         17. Rainville, P. , Duncan, G. H. , Price, D. D. , Carrier, B. , and Bushnell, M. C. ( 1997 ) Science 227 , 968971 .
                                                                                                                                                                                                                                         18. Meyer, E. , Ferguson, S. G. , Zatorre, R. J. , Alivisatos, B. , Marrett, S. , Evans, A. C. & Hakim, A. M. ( 1991 ) Ann. Neurol. 29 , 440443 .
                                                                                                                                                                                                                                         19. Hyvrinen, J. , Poranen, A. & Jokinen, Y. ( 1980 ) J. Neurophysiol. 43 , 870882 .
                                                                                                                                                                                                                                         20. Poranen, A. & Hyvrinen, J. ( 1982 ) Electroencephalogr. Clin. Neurophysiol. 53 , 535537 .
                                                                                                                                                                                                                                         21. Andersson, J. L. R. , Lilja, A , Hartvig, P. , Lngstrm, B. , Gordh, T. , Handwerker, H. & Torebjrk, E. ( 1997 ) Exp. Brain Res. 117 , 192199 .
                                                                                                                                                                                                                                         22. Apkarian, A. V. , Stea, R. A. & Bolanowski, S. J. ( 1994 ) Somatosens. Mot. Res. 11 , 259267 .
                                                                                                                                                                                                                                         23. Dougherty, P. M. , Schwartz, A. & Lenz, F. A. ( 1998 ) Neuroscience , 90 , 13771392 .
                                                                                                                                                                                                                                         24. Crawford, H. J. , Gur, R. C. , Skolnick, B. , Gur, R. E. & Benson, D. M. ( 1993 ) Int. J. Psychophysiol. 15 , 181195 .
                                                                                                                                                                                                                                         25. Coghill, R. C. , Talbot, J. D. , Evans, A. C. , Meyer, E. , Gjedde, A , Bushnell, M. C. & Duncan, G. H. ( 1994 ) J. Neurosci. 14 , 40954108 .
                                                                                                                                                                                                                                         26. Di Piero, V. , Ferracuti, S. , Sabatini, U. , Pantano, P. , Cruccu, G. & Lenzi, G. L. ( 1994 ) Pain 56 , 167173 .
                                                                                                                                                                                                                                         27. Derbyshire, S. W. G. , Jones, A. K. P. , Devani, P. , Friston, K. J. , Feinmann, C. , Harris, M. , Pearce, S. , Watson, J. D. G. & Frackowiak, R. S. J. (
                                                                                                                                                                                                                                              1994 ) J. Neurol. Neurosurg. Psychiatry 57 , 11661172 .
                                                                                                                                                                                                                                         28. Rosen, S. D. , Paulesu, E. , Frith, C. D. , Frackowiak, R. S. J. , Davies, G. J. , Jones, T. & Camici, P. G. ( 1994 ) Lancet 344 , 147150 .
                                                                                                                                                                                                                                         29. Hsieh, J. C. , Belfrage, M. , Stone-Elander, S. , Hansson, P. & Ingvar, M. ( 1995 ) Pain 63 , 225236 .
                                                                                                                                                                                                                                         30. Hsieh, J. C. , Sthle-Bckdahl, M. , Hgermark , Stone-Elander, S. , Rosenquist, G. & Ingvar, M. ( 1995 ) Pain 64 , 303314 .
                                                                                                                                                                                                                                         31. Davis, K. D. , Wood, M. L. , Crawley, A. P. & Mikulis, D. J. ( 1995 ) NeuroReport 7 , 321325 .
                                                                                                                                                                                                                                         32. Weiller, C. , May, A , Limmroth, V. , Juptner, M. , Kaube, H. , Schayck, R. v. , Coenen, H. H. & Diener, H. C. ( 1995 ) Nat. Med. 1 , 658660 .
                                                                                                                                                                                                                                         33. Howland, E. W. , Wakai, R. T. , Mjaanes, B. A. , Balog, J. P. & Cleeland, C. S. ( 1995 ) Cognit. Brain Res. 2 , 165172 .
                                                                                                                                                                                                                                         34. Kitamura, Y. , Kakigi, R. , Hoshiyama, M. , Koyama, S. , Shimojo, M. & Watanabe, S. ( 1995 ) Electroencephalogr. Clin. Neurophysiol. 95 , 463
                                                                                                                                                                                                                                              474 .
                                                                                                                                                                                                                                         35. Craig, A. D. , Reiman, E. M. , Evans, A. C. & Bushnell, M. C. ( 1996 ) Nature (London) 384 , 258260 .
                                                                                                                                                                                                                                         36. Casey K. L. , Minoshima, S. , Morrow T. J. & Koeppe, R. A. ( 1996 ) Neurophysiology 76 , 571581 .
                                                                                                                                                                                                                                         37. Hsieh, J. C. , Hannerz, J. & Ingvar, M. ( 1996 ) Pain 67 , 5968 .
                                                                                                                                                                                                                                         38. Antognini, J. F. , Buonocore, M. H. , Disbrow, E. A. & Carstens, E. ( 1997 ) Life Sci. 61 , 349354 .
                                                                                                                                                                                                                                         39. Aziz, Q. , Andersson, J. L. R. , Valind, S. , Sundin, A. , Hamdy, S. , Jones, A. K. P. , Foster, E. R. , Langstrom, B. & Thompson, D. G. ( 1997 )
                                                                                                                                                                                                                                              Gastroenterology 113 , 5059 .
                                                                                                                                                                                                                                         40. Di Piero, V. , Fiacco, F. , Tombari, D. & Pantano, P. ( 1997 ) Pain 70 , 185191 .
                                                                                                                                                                                                                                         41. Silverman, D. H. S. , Munakata, J. A , Ennes, H. , Mandelkern, M. A. , Hoh, C. K. & Mayer, E. A ( 1997 ) Gastroenterology 112 , 6472 .
                                                                                                                                                                                                                                         42. Svensson, P. , Rosenberg, B. , Beydoun, A. , Morrow, T. J. , and Casey, K. L. ( 1997 ) Somatosens. Mot. Res. 14 , 113118 .
                                                                                                                                                                                                                                         43. Svensson, P. , Beydoun, A. , Morrow, T. J. & Casey, K. L. ( 1997 ) Exp. Brain Res. 114 , 390392 .
                                                                                                                                                                                                                                         44. Xu, X. P. , Fukuyama, H. , Yazawa, S. , Mima, T. , Hanakawa, T. , Magata, Y. , Kanda, M. , Fujiwara, N. , Shindo, K. , Nagamine, T. & Shibasaki,
                                                                                                                                                                                                                                              H. ( 1997 ) NeuroReport 8 , 555559 .
                                                                                                                                                                                                                                         45. Binkofski, F. , Schnitzler, A , Enck, P. , Frieling, T. , Posse, S. , Seitz, R. J. & Freund, H.-J. ( 1998 ) Ann. Neurol. 44 , 811815 .
                                                                                                                                                                                                                                         46. Derbyshire, S. W. G. , Vogt, B. A. & Jones, A. K. P. ( 1998 ) Exp. Brain Res. 118 , 5260 .
the authoritative version for attribution.
                                                                                                                                                                                                                                         47. Iadarola, M. J. , Berman, K. F. , Zeffiro, T. A , Byas-Smith, M. G. , Gracely, R. H. , Max, M. B. & Bennett, G. J. ( 1998 ) Brain 121 , 931947 .
                                                                                                                                                                                                                                         48. May, A , Kaube, H. , Bchel, C. , Eichten, C. , Rijntjes, M. , Jptner, M. , Weiller, C. & Diener, H. C. ( 1998 ) Pain 74 , 6166 .
                                                                                                                                                                                                                                         49. Oshiro, Y. , Fuijita, N. , Tanaka, H. , Hirabuki, N. , Nakamura, H. & Yoshiya, I. ( 1998 ) NeuroReport 9 , 22852289 .
                                                                                                                                                                                                                                         50. Paulson P. E. , Monoshima, S. , Morrow, T. J. & Casey, K. L. ( 1998 ) Pain 76 , 223229 .
                                                                                                                                                                                                                                         51. Hsieh, J.-C . ( 1995 ) Ph.D. thesis ( Karolinska Institute , Stockholm ).
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                         to-brain communication must occur because only the brain can orchestrate such a pervasive array of changes.
                                                                                                                                                                                                                                              The triggers for initiation of the sickness response are substances released by immune cells activated by the foreign entity. As a
                                                                                                                                                                                                                                         group, these proteins are referred to as proinflammatory cytokines ( 3 ). This name reflects the fact that these proteins orchestrate and
                                                                                                                                                                                                                                         augment inflammatory responses. Proinflammatory cytokines include IL-1, IL-6, and tumor necrosis factor. These proteins may be
                                                                                                                                                                                                                                         necessary and sufficient for sickness, because preventing their actions (using receptor antagonists, etc.) block sickness responses, whereas
                                                                                                                                                                                                                                         exogenous administration of these proteins can create sickness responses ( 1 , 5 ).
                                                                                                                                                                                                                                                                  PAIN AS A NATURAL OUTCOME OF IMMUNE-TO-BRAIN COMMUNICATION
                                                                                                                                                                                                                                               The constellation of responses reviewed above constitute the classic view of sickness. Although changes in pain responsivity have not
                                                                                                                                                                                                                                         been considered in this classic view, it would be reasonable to suspect that hyperalgesia might be a natural part of this response profile
                                                                                                                                                                                                                                         because recuperative behaviors supportive of healing would be produced by enhanced pain ( 6 ). Furthermore, recuperative behaviors
                                                                                                                                                                                                                                         would be expected to decrease activity and so decrease energy expenditure, again in keeping with the view that sickness responses serve to
                                                                                                                                                                                                                                         save energy for use in the production of fever ( 5 ).
                                                                                                                                                                                                                                               If such an argument has merit, then one would expect that hyperalgesia should occur after administration of agents known to induce
                                                                                                                                                                                                                                         sickness. In fact, hyperalgesia is produced both by i.p. administration of the cell walls of Gram-negative bacteria (endotoxin; also called
                                                                                                                                                                                                                                         lipopolysaccharide) ( 7 ) and by i.p. live bacteria ( 8 ), both of which are known to elicit the release of proinflammatory cytokines from a
                                                                                                                                                                                                                                         variety of immune cells. Hyperalgesia also can be elicited simply by administering either IL-1 or tumor necrosis factor alone ( 9 , 10 ). The
                                                                                                                                                                                                                                         key importance of proinflammatory cytokines in sickness hyperalgesia is clear from the fact that it can be blocked by either an IL-1
                                                                                                                                                                                                                                         receptor antagonist or tumor necrosis factor binding protein ( 9 , 11 ). Thus, these cytokines can be both necessary and sufficient for
                                                                                                                                                                                                                                         sickness-induced hyperalgesia to occur.
                                                                                                                                                                                                                                               The mechanism(s) by which proinflammatory cytokines activate the central nervous system is a matter of lively controversy. Both
                                                                                                                                                                                                                                         blood-borne and peripheral nerve-generated signals have been proposed ( 12 ). For localized tissue infection/inflammation and localized
                                                                                                                                                                                                                                         proinflammatory cytokine administration, at least, activation of peripheral nerves appears to be the most likely route ( 12 14 ). By using
                                                                                                                                                                                                                                         i.p. administration of sickness agents as the example, signals to the brain appear to be carried via the subdiaphragmatic vagus because: (a)
                                                                                                                                                                                                                                         sickness agents activate sensory vagal neurons, as supported by increases in cFos expression in these cells ( 15 ), and (b) subdiaphragmatic
                                                                                                                                                                                                                                         vagotomy blocks hyperalgesia induced by endotoxin, IL-1, and tumor necrosis factor ( 9 , 10 ). Activation of the sensory vagus may be
                                                                                                                                                                                                                                         secondary to activation of specialized sensory structures called paraganglia ( 16 ). These sensory cells are thought to be chemoreceptors,
                                                                                                                                                                                                                                         based on their anatomy. They express IL-1 binding sites and are known to form afferent synapses onto sensory vagal fibers. Intriguingly,
                                                                                                                                                                                                                                         these IL-1 binding paraganglia are in close physical proximity to dense populations of immune cells that can create and release IL-1 ( 17 ).
                                                                                                                                                                                                                                         These accumulations of macrophages, mast cells, and dendritic cells embedded in connective tissue near paraganglia have been referred to
                                                                                                                                                                                                                                         as nerve-associated lymphoid cells (NALC) ( 17 ). The location of these immune cells and their ability to express IL-1 suggest that this
                                                                                                                                                                                                                                         NALC may rapidly recognize infection and signal the brain via IL-1 release onto neighboring paraganglia. In support of this notion, we
                                                                                                                                                                                                                                         have found that levels of IL-1 in the NALC associated with the paraganglia rapidly and dramatically increase after i.p. illness agents ( 17 ).
                                                                                                                                                                                                                                         Thus, this arrangement of peripheral cells may underlie immune-to-brain communication arising from the abdomen.
                                                                                                                                                                                                                                               The pathway by which abdominal sickness signals elicit hyperalgesia has been at least partially mapped in the central nervous
                                                                                                                                                                                                                                         system, by using a combination of discrete lesions and expression of cFos, an immediate-early gene product used as a neuronal activation
                                                                                                                                                                                                                                         marker ( 18 ). From this work, the neural circuitry underlying sickness hyperalgesia was found to involve a nucleus tractus solitarius
                                                                                                                                                                                                                                         nucleus raphe magnusspinal cord dorsolateral funiculus circuit. The involvement of the nucleus tractus solitarius is notable, in that this
                                                                                                                                                                                                                                         appears to be a common hub within the brain for creating sickness responses ( 19 ).
                                                                                                                                                                                                                                               Whether hyperalgesia induced by peripheral inflammation is mediated by the same general pathway is not yet clear. Although s.c.
                                                                                                                                                                                                                                         inflammation (formalin) hyperalgesia is not affected by subdiaphragmatic vagotomy ( 10 ), it does require a brain-to-spinal cord circuit
                                                                                                                                                                                                                                         that, like sickness hyperalgesia, involves the nucleus raphe magnus ( 20 ). Whether the nucleus tractus solitarius is involved is uncertain.
                                                                                                                                                                                                                                         Although unilateral lesions of the nucleus tractus solitarius failed to affect s.c. formalin hyperalgesia ( 21 ), this may well be because
                                                                                                                                                                                                                                         dorsal horn laminae responsive to s.c. formalin send bilateral projections to this medullary structure ( 22 ). Bilateral lesions could not be
                                                                                                                                                                                                                                         tested given the survival problems such animals face. In addition to at least partial overlap in the neurocircuitry of inflammation- and
                                                                                                                                                                                                                                         sickness-induced hyperalgesias, there are similarities in their neurochemistry as well. Both depend on nitric oxide, excitatory amino acids,
                                                                                                                                                                                                                                         and substance P at the level of the spinal cord ( 20 ).
                                                                                                                                                                                                                                                                                          Role of Spinal Cord Glia in Exaggerated Pain
                                                                                                                                                                                                                                               From the discussion above, it is clear that peripheral infection/ inflammation leads to activation of a brain-to-spinal cord pathway,
                                                                                                                                                                                                                                         culminating in the creation of hyperalgesia. However, an intriguing aspect of this spinal circuitry is that it critically depends on activation
                                                                                                                                                                                                                                         of spinal cord microglia and astrocytes. Indeed, hyperalgesia produced either by s.c. inflammation ( 23 ) or i.p. bacterial infection ( 8 ) can
                                                                                                                                                                                                                                         be blocked by spinal administration of drugs that disrupt glial function. Further, anatomical examination of astrocytes and microglia show
                                                                                                                                                                                                                                         them to be clearly activated by peripheral infection/inflammation, as evidenced immunohistochemically by increased expression of glia-
                                                                                                                                                                                                                                         specific activation markers ( 8 ). Lastly, i.p. endotoxin at the same dose that elicits hyperalgesia rapidly increases dorsal spinal cord levels
                                                                                                                                                                                                                                         of IL-1, a product of glial activation ( 24 ).
                                                                                                                                                                                                                                               So how are these glia activated and what role do they play in exaggerated pain states? Regarding activation, the glia may be activated
                                                                                                                                                                                                                                         by neurotransmitter(s) released by spinal projections of the nucleus raphe magnus. Candidate neurotransmitters to serve this role are
                                                                                                                                                                                                                                         substance P and glutamate, because: (a) as noted above, both sickness-induced hyperalgesia and s.c. formalin hyperalgesia are mediated,
                                                                                                                                                                                                                                         in part, by spinal cord substance P and excitatory amino acids, (b) the nucleus raphe magnus-to-spinal cord pathway mediating both
                                                                                                                                                                                                                                         sickness-induced hyperalgesia and s.c. formalin hyperalgesia contains substance P and glutamate as neurotransmitters ( 25 ), (c) microglia
                                                                                                                                                                                                                                         and astrocytes express substance P and glutamate receptors ( 26 ), and (d) glia are activated by substance P and glutamate in vitro ( 27 ).
                                                                                                                                                                                                                                         Once activated, astrocytes and microglia form a positive feedback circuit whereby substances released from microglia activate astrocytes
                                                                                                                                                                                                                                         to release substances that further stimulate microglia, and so forth ( 28 ). Many of the substances that can be released from microglia and
                                                                                                                                                                                                                                         astrocytes are known to be key mediators of hyperalgesia, including nitric oxide, excitatory amino acids [both N-methyl-D-aspartate
the authoritative version for attribution.
                                                                                                                                                                                                                                         taglandins, and nerve growth factor ( 28 ). Thus, once spinal cord microglia and astrocytes are activated in a perseverative positive
                                                                                                                                                                                                                                         feedback manner, the neuroexcitatory substances they release could drive exaggerated pain states.
                                                                                                                                                                                                                                               However, astrocytes and microglia have not generally been viewed as cells whose major function is activation in response to
                                                                                                                                                                                                                                         centrifugal hyperalgesia circuitry. Rather, astrocytes and microglia are immunocompetent cells and thus can respond like immune cells
                                                                                                                                                                                                                                         within the central nervous system. Astrocytes and microglia express specific receptors for various bacteria and viruses and are activated on
                                                                                                                                                                                                                                         binding to these infectious agents. An example of a neurotropic virus (that is, a virus that can home to the brain and spinal cord) is
                                                                                                                                                                                                                                         HIV-1, which causes AIDS. HIV-1 invades the brain and spinal cord early in, and continuing throughout, disease progression ( 29 ) and
                                                                                                                                                                                                                                         this invasion leads to the activation of microglia and astrocytes ( 30 ). One reason for the prolonged microglial and astrocyte activation by
                                                                                                                                                                                                                                         HIV-1 is that currently available drugs used to treat AIDS do not readily penetrate the blood-brain barrier, so HIV-1 within the brain and
                                                                                                                                                                                                                                         spinal cord are not disrupted by such treatments.  Once within nervous tissue, HIV-1 binds to receptors on microglia and astrocytes that
                                                                                                                                                                                                                                         recognize one specific portion of the virus, namely a glycoprotein (called gp120) expressed on the outer surface of the viral coat ( 31 ).
                                                                                                                                                                                                                                               The arguments developed above predict that intrathecal delivery of gp120 should be sufficient to produce hyperalgesia, if immune
                                                                                                                                                                                                                                         activation of glia initiates the same sort of positive feedback cascade as occurs for sickness-induced hyperalgesia. In our initial series of
                                                                                                                                                                                                                                         studies of this issue, we found that gp120 delivered over lumbosacral cord caused dose-dependent hyperalgesia as measured by the tail-
                                                                                                                                                                                                                                         flick test. Because the receptors identified on microglia and astrocytes require the complex three-dimensional conformational structure of
                                                                                                                                                                                                                                         native gp120 for receptor activation to occur ( 32 ), it follows that irreversible heat denaturation of gp120s natural conformation should
                                                                                                                                                                                                                                         disrupt the ability of this protein to create hyperalgesia, if microglia and astrocytes function as we hypothesize. In fact, such disruption of
                                                                                                                                                                                                                                         the normal three-dimensional structure of gp120 dose abolish the effects of this viral protein on pain.
                                                                                                                                                                                                                                               Results using the tail-flick test can, at times, be confounded by drug-induced alterations in tail skin temperature ( 33 ). We have
                                                                                                                                                                                                                                         examined this issue by using two independent approaches. First, we monitored superficial tail temperature throughout tail-flick testing and
                                                                                                                                                                                                                                         found that there was no correlation between superficial tail temperature and tail-flick latency ( 24 ). Second, we examined the effect of
                                                                                                                                                                                                                                         intrathecal gp120 on withdrawal latency of the plantar surface of the hind paws in response to a radiant heat stimulus. This experiment was
                                                                                                                                                                                                                                         important because the paws, unlike the tail, are not used to regulate the organisms temperature. Therefore, paw withdrawal latencies are
                                                                                                                                                                                                                                         not subject to the skin temperature confounds inherent in the tail-flick test. Here again, robust hyperalgesia was observed, supporting the
                                                                                                                                                                                                                                         conclusion that intrathecal gp120 produces thermal hyperalgesia ( 34 ).
                                                                                                                                                                                                                                               Intrathecal gp120 produces mechanical allodynia as well as thermal hyperalgesia. The first indication of this effect came from pilot
                                                                                                                                                                                                                                         studies in which we observed marked increases in vocalization of gp120-injected rats to light touch, compared with vehicle controls. These
                                                                                                                                                                                                                                         initial observations were followed by experiments that used two standardized tests. First, we used calibrated VonFrey monofilaments to
                                                                                                                                                                                                                                         examine withdrawal responses elicited from the plantar surface of the hind paws in response to low-threshold mechanical stimuli. This
                                                                                                                                                                                                                                         procedure clearly demonstrated that gp120 induces allodynia ( 34 ). Second, we used light touch stimuli to the fur of the hindquarters and
                                                                                                                                                                                                                                         found that gp120 induced touch-evoked agitation as well ( 34 ).
                                                                                                                                                                                                                                               Importantly, activation of spinal cord microglia and astrocytes appear to be critical for the hyperalgesic and allodynic effects of
                                                                                                                                                                                                                                         gp120. Pilot studies using immunohistochemistry suggest activation of glia in spinal cord after gp120. Furthermore, pretreatment of the
                                                                                                                                                                                                                                         rats with a drug that disrupts glial function prevented both gp120-induced thermal hyperalgesia and mechanical allodynia ( 24 , 34 ).
                                                                                                                                                                                                                                               The mechanisms underlying these effects are at present unknown. We are actively investigating this issue and predict that the effects
                                                                                                                                                                                                                                         will be similar to those previously defined for sickness-induced hyperalgesia. Recall that IL-1 was previously noted to rapidly increase in
                                                                                                                                                                                                                                         dorsal spinal cord after i.p. endotoxin, and that spinal cord IL-1 is a key mediator of hyperalgesia induced by peripheral inflammation.
                                                                                                                                                                                                                                         Thus, our initial pilot studies have focused on potential gp120-induced changes in spinal cord IL-1. To date, these data indicate that
                                                                                                                                                                                                                                         intrathecal gp120 produces a rapid and dramatic increase in dorsal spinal cord IL-1 mRNA and IL-1 protein ( 34 ). This gp120-induced
                                                                                                                                                                                                                                         increase in IL-1 protein does not simply reflect intracellular content, but rather is indicative of increased IL-1 release into extracellular
                                                                                                                                                                                                                                         space, because levels of IL-1 in lumbosacral cerebrospinal fluid dramatically increase as well ( 34 ). Although we have not yet directly
                                                                                                                                                                                                                                         tested the effect of IL-1 receptor antagonist on gp120-induced effects, these preliminary data are certainly consistent with the view that
                                                                                                                                                                                                                                         gp120-induced alterations in glial function are likely to be important for hyperalgesia and/or allodynia.
                                                                                                                                                                                                                                                                                            CONCLUSIONS AND IMPLICATIONS
                                                                                                                                                                                                                                              The evidence reviewed above places exaggerated pain in a new framework. This view conceptualizes hyperalgesia not as an entity in
                                                                                                                                                                                                                                         and of itself, but rather as a single element in a much larger constellation of physiological, behavioral, and hormonal changes, orchestrated
                                                                                                                                                                                                                                         by the central nervous system in response to bodily infection and inflammation. This sickness response enhances survival of the organism
                                                                                                                                                                                                                                         in the face of immune challenges. Hyperalgesia, like all of the other components of the sickness response, is triggered by proinflammatory
                                                                                                                                                                                                                                         cytokines released by activated macrophages and other immune cells. For localized immune challenges, at least, this proinflammatory
                                                                                                                                                                                                                                         cytokine release leads to activation of peripheral nerves that signal the brain. Activation of a centrifugal pathway then occurs, resulting in
                                                                                                                                                                                                                                         the activation of microglia and astrocytes within the spinal cord dorsal horn. Neurotransmitters released by the centrifugal pathway
                                                                                                                                                                                                                                         combined with neuroexcitatory substances released by astrocytes and microglia create exaggerated pain responses. Thus, for this form of
                                                                                                                                                                                                                                         hyperalgesia at least, glia assume a new, pivotal role in the generation of exaggerated pain.
                                                                                                                                                                                                                                              The importance of microglia and astrocytes in spinal cord hyperalgesia mechanisms has potentially important implications. Most
                                                                                                                                                                                                                                         important of these is that glia may exaggerate pain when these immune-like cells respond to infection/ inflammation within the central
                                                                                                                                                                                                                                         nervous system. Clearly, this is the case after intrathecal administration of the HIV-1 envelope protein, gp120. Given that a number of
                                                                                                                                                                                                                                         viruses and bacteria can invade the central nervous system, such observations suggest that glia may have a far more important role in pain
                                                                                                                                                                                                                                         modulation than previously recognized.
                                                                                                                                                                                                                                              This work was supported by National Institutes of Health Grants MH55283, MH01558, MH00314, and MH45045 and the Council of
                                                                                                                                                                                                                                         Research and Creative Works of the University of Colorado at Boulder.
the authoritative version for attribution.
                                                                                                                                                                                                                                            Sham, H., Kempf, D., Molla, A., Marsh, K., Betebenner, D., Chen, X., Rosenbrook, W., Wideburg, N., Chen, C, Kati, W., et al.,
                                                                                                                                                                                                                                         Fourth Conference on Retroviruses and Opportunistic Infections, Jan. 22 26, 1997, Washington, DC, abstr. 396.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
SPINAL CORD
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
SPINAL CORD
                                                                                                                                                                                                                                         abundant in intrinsic spinal-cord neurons nor does it coexist and 9 ; see Fig. 1 ). Notably, BDNF immunoreactivity is not within the
                                                                                                                                                                                                                                         primary afferent terminals or cell bodies marked by binding of the lectin IB4. This population of sensory neurons does not constitutively
                                                                                                                                                                                                                                         express neuropeptides but expresses GDNF receptor components and is responsive to this factor ( 10 ). BDNF immunoreactivity is present
                                                                                                                                                                                                                                         only sparingly within deeper laminae. Thus, most of the BDNF protein in spinal cord seems to be localized in the terminals of primary
                                                                                                                                                                                                                                         sensory nociceptors, specifically those that express trkA and are known to be sensitive to NGF ( 11 ). In these sensory neurons, BDNF is
                                                                                                                                                                                                                                         located in dense core vesicles ( 12 ), suggesting that it is likely to be released with activity in nociceptors. The superficial laminae of the
                                                                                                                                                                                                                                         spinal cord are of course, important sites for integration of nociceptive information. Full-length trkB receptors are present in the cells in
                                                                                                                                                                                                                                         this region of the spinal cord ( 13 ).
                                                                                                                                                                                                                                           FIG. 1. BDNF expression within the DRG and spinal dorsal horn, (a and c) Under normal circumstances, modest BDNF
                                                                                                                                                                                                                                           immunoreactivity is observed within lumbar DRG and superficial dorsal horn, (e, bold arrow) Constitutively expressed BDNF
                                                                                                                                                                                                                                           colocalizes with the neuropeptide calcitonin-gene-related peptide (CGRP) and is present mainly within small-diameter sensory
                                                                                                                                                                                                                                           neurons. These neurons mostly do not express trkB. BDNF expression is regulated by NGF. (b and d) After systemic NGF treatment,
                                                                                                                                                                                                                                           BDNF immunoreactivity is enhanced within lumbar ganglia and dorsal horn, (e, light arrow) After NGF treatment, 8090% of trkA-
                                                                                                                                                                                                                                           expressing sensory neurons contain BDNF immunoreactivity. These neurons coexpress CGRP.
                                                                                                                                                                                                                                               The possible interaction of BDNF with nociceptive pathways is highlighted further by the fact that the levels of BDNF are regulated
                                                                                                                                                                                                                                         by another neurotrophin, NGF ( Fig. 1 ). Several recent studies have shown that BDNF levels within sensory neurons may be increased by
                                                                                                                                                                                                                                         exogenous administration of NGF ( 6 , 8 ): NGF increases the number of DRG cells showing BDNF immunoreactivity and mRNA.
                                                                                                                                                                                                                                         Although there is some debate as to which cells express BDNF after NGF treatment, it is apparent that considerable numbers of these
                                                                                                                                                                                                                                         neurons express trkA (and are thus directly responsive to NGF). In one study ( 8 ) for instance, 8090% of those sensory neurons
                                                                                                                                                                                                                                         expressing BDNF immunoreactivity after intrathecal NGF belonged to the trkA group.
                                                                                                                                                                                                                                               The peripheral synthesis of NGF is known to be increased in a variety of inflammatory conditions, particularly after injury or
                                                                                                                                                                                                                                         experimentally induced inflammation. Under such pathophysiological states BDNF may be up-regulated in nociceptors. Cho et al. ( 7 )
                                                                                                                                                                                                                                         showed that intraplantar Freunds adjuvant injection was associated with a significant increase in BDNF mRNA with a time course
                                                                                                                                                                                                                                         consistent with the known increase of NGF in this model. Moreover, the increase in BDNF mRNA was prevented by coinjection of NGF
                                                                                                                                                                                                                                         antibody, suggesting that BDNF expression depends on peripheral NGF production. Because there is good evidence that some of the
                                                                                                                                                                                                                                         sensory abnormalities associated with inflammation depend on NGF production, this finding in consistent with the notion that BDNF itself
                                                                                                                                                                                                                                         contributes to altered sensory processing.
                                                                                                                                                                                                                                               The expression of BDNF is altered in an interesting manner in another pathophysiological state, that associated with peripheral nerve
                                                                                                                                                                                                                                         injury. After sciatic axotomy, BDNF is up-regulated in large-diameter DRG cells, and anterograde transport to the dorsal horn of the spinal
                                                                                                                                                                                                                                         cord increases ( 14 ). Thus, with peripheral nerve injury, BDNF expression decreases in nociceptors but increases in large-diameter
                                                                                                                                                                                                                                         sensory neurons (these large-diameter neurons are generally considered to subserve transmission of innocuous sensations from the
                                                                                                                                                                                                                                         periphery). The signal for the expression in large cells is not yet known, although reduced retrograde transport of neurotrophin-3 by large-
                                                                                                                                                                                                                                         diameter sensory neurons is one obvious possibility. The BDNF in damaged large neurons is also transported to the central terminals of
                                                                                                                                                                                                                                         these fibers. It is tempting to speculate that this BDNF also may have similar postsynaptic effects to those described below. Of course, if
                                                                                                                                                                                                                                         so, such effects are likely to be found in deeper dorsal horn laminae, the known normal termination sites of large afferents.
                                                                                                                                                                                                                                               BDNF Has Postsynaptic Actions Within the Spinal Dorsal Horn. Modulation of spinal reflex excitability. Although there is
                                                                                                                                                                                                                                         evidence for the activity-dependent secretion of neurotrophins from hippocampal slices ( 15 ), there is, as yet, no such evidence that BDNF
                                                                                                                                                                                                                                         is released within the spinal cord after afferent fiber activation. The localization of BDNF and its injury-associated regulation detailed
                                                                                                                                                                                                                                         above, however, are strongly consistent with a possible neuromodulatory role. Recently, we tested this hypothesis further by examining
                                                                                                                                                                                                                                         the effect of exogenous BDNF delivery on spinal reflex excitability.
                                                                                                                                                                                                                                               Because neurotrophins do not penetrate the spinal cord readily after systemic delivery, we used an in vitro spinal-cord preparation to
                                                                                                                                                                                                                                         study BDNFs actions. The in vitro hemisected spinal cord is used routinely in our and other laboratories to monitor alterations in synaptic
                                                                                                                                                                                                                                         excitability. Reflex responses to C fiber inputs may be recorded from the ventral root, close to the motoneuron cell bodies, with close-
                                                                                                                                                                                                                                         fitting glass suction electrodes, and these reflex responses appear as prolonged ventral root potentials (VRPs). These extracellularly
                                                                                                                                                                                                                                         recorded potentials are a good measure of spinal excitability ( 16 ) and, under control conditions, will remain stable for several hours (
                                                                                                                                                                                                                                         Fig. 2 a). These preparations were superfused the with BDNF at 2- 1,000 ng/ml for 30 min. The C fiber-evoked responses were recorded
the authoritative version for attribution.
                                                                                                                                                                                                                                         before BDNF exposure and for several hours after cessation of BDNF superfusion. A significant and sustained increase in the C fiber-
                                                                                                                                                                                                                                         evoked reflex response was observed repeatedly ( Fig. 2 b; refs. 17 and 18 ). This
                                                                                                                                                                                                                                         BRAIN-DERIVED NEUROTROPHIC FACTOR IS AN ENDOGENOUS MODULATOR OF NOCICEPTIVE RESPONSES IN THE                                          7716
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
SPINAL CORD
                                                                                                                                                                                                                                         enhancement of C fiber-evoked excitability was sustained, outlasting the duration of BDNF administration, and was dosedependent. The
                                                                                                                                                                                                                                         threshold dose to elicit an increased spinal excitability was in the range 100200 ng/ml BDNF, which suggests a specific and receptor-
                                                                                                                                                                                                                                         mediated event. Coadministration of the BDNF-sequestering molecule trkB-IgG to spinal-cord preparations eliminates the facilitatory
                                                                                                                                                                                                                                         effect of BDNF and further indicates that the effect of BDNF is likely to be mediated by interaction at the trkB receptor. Supervision of
                                                                                                                                                                                                                                         spinal-cord preparations with NGF (200 ng/ml) does not produce similar effects on C fiber-evoked reflex excitability.
                                                                                                                                                                                                                                           FIG. 2. Exogenous BDNF enhances postsynaptic activity in the spinal cord and endogenous BDNF contributes to reflex excitability,
                                                                                                                                                                                                                                           (a) Spinal reflex excitability was assessed from the VRP evoked after electrical stimulation of C fibers in the dorsal root of an
                                                                                                                                                                                                                                           isolated hemisected rat spinal-cord preparation. Under normal circumstances, the VRP evoked by C fiber activation is characterized
                                                                                                                                                                                                                                           by a prolonged, slowly decaying potential, and this potential remains stable from trial to trial, (b) After a 30-min superfusion with
                                                                                                                                                                                                                                           BDNF (200 ng/ml), the amplitude and duration of the C fiber-evoked VRP increased significantly relative to pretreatment activity
                                                                                                                                                                                                                                           (arrow, control trace; arrowhead, trace after BDNF superfusion). (c) The contribution of endogenous BDNF to reflex excitability was
                                                                                                                                                                                                                                           assessed after a 30-min superfusion of spinal-cord preparations with trkB-IgG (500 ng/ml). Under these circumstances C fiber-
                                                                                                                                                                                                                                           evoked activity was reduced significantly compared with pretreatment control responses (arrow, control response; arrowhead,
                                                                                                                                                                                                                                           response after trkB-IgG superfusion). (b and c; bars = 0.5 mV and 5.0 s.)
                                                                                                                                                                                                                                               The mechanism of action of BDNF within the spinal cord is, at present, unknown. There is evidence from primary cultures of
                                                                                                                                                                                                                                         hippocampal neurons that BDNF enhances spontaneous release of glutamate and acetylcholine, suggesting that BDNF functions there as a
                                                                                                                                                                                                                                         retrograde modulator of presynaptic transmitter release ( 19 ). It is unlikely that such a mechanism will operate within the spinal cord,
                                                                                                                                                                                                                                         because those sensory neurons that express BDNF do not in general display the trkB receptor and are therefore likely to be insensitive to
                                                                                                                                                                                                                                         the synaptically released neurotrophin. There is also good evidence, again from the hippocampus, that postsynaptic injection of protein
                                                                                                                                                                                                                                         kinase inhibitors will block the effect of BDNF on synaptic enhancement, suggesting that phosphorylation of postsynaptic receptors is a
                                                                                                                                                                                                                                         critical step in the BDNF effect. Recently, it has been shown that BDNF rapidly and specifically enhances phosphorylation of the
                                                                                                                                                                                                                                         postsynaptic N-methyl-D-aspartic acid (NMDA) receptor ( 20 ). Again in the hippocampus, BDNF will potentiate NMDA responses via a
                                                                                                                                                                                                                                         3-fold increase in NMDA receptor open time ( 20 ). In cultured rat neurons in the central nervous system, BDNF will also induce changes
                                                                                                                                                                                                                                         in NR2A and NR2B NMDA receptor subunit expression ( 21 ). NR2 NMDA receptor subunits define the pharmacological and biophysical
                                                                                                                                                                                                                                         properties of this receptor ( 22 ). In the spinal cord, NMDA receptor activation plays a central role in the induction and maintenance of
                                                                                                                                                                                                                                         central sensitization. In this well studied phenomenon, the recruitment of the NMDA receptor seems to
                                                                                                                                                                                                                                           FIG. 3. BDNF induces alterations in immediate early gene expression within the spinal cord. (a) A large increase in c-fos
                                                                                                                                                                                                                                           immunoreactivity is observed within the superficial dorsal horn 2 h after intrathecal administration of BDNF (5 g). (b) This increase
                                                                                                                                                                                                                                           in immunoreactivity is not observed after intrathecal administration of NGF or saline.
the authoritative version for attribution.
                                                                                                                                                                                                                                         BRAIN-DERIVED NEUROTROPHIC FACTOR IS AN ENDOGENOUS MODULATOR OF NOCICEPTIVE RESPONSES IN THE                                               7717
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
SPINAL CORD
be the pivotal event in increasing the sensitivity of nociceptive spinal circuits to sensory inputs ( 23 ).
                                                                                                                                                                                                                                           FIG. 4. Endogenous BDNF release contributes to nociceptive behavioral responses in vivo. Animals were pretreated with NGF to
                                                                                                                                                                                                                                           maximize nociceptor BDNF levels. Intraplantar formalin [50 l; 2% (vol/vol)] induces a characteristic two-phase nocifensor paw-
                                                                                                                                                                                                                                           withdrawal behavior. Intrathecal administration of trkB-IgG (but not saline) 30 min before formalin injection significantly reduces
                                                                                                                                                                                                                                           both phases of the formalin response.
                                                                                                                                                                                                                                               Central sensitization is believed to underlie some aspects of altered sensibility found in chronic pain states. Thus, again, the available
                                                                                                                                                                                                                                         data are consistent with the idea that BDNF may be a key mediator of central sensitization within the spinal cord via an interaction at the
                                                                                                                                                                                                                                         NMDA receptor site. Several other molecules have been implicated in this process of central sensitization, most notably the tachykinin
                                                                                                                                                                                                                                         substance P ( 23 ). It is possible that BDNF is simply one of a series of modulators capable of triggering postsynaptic changeperhaps via
                                                                                                                                                                                                                                         a common mechanism of NMDA receptor phosphorylation. It is interesting that substance P is itself regulated in a very similar fashion to
                                                                                                                                                                                                                                         that described above for BDNF. That is, it is up-regulated in trkA-expressing cells by exogenous NGF or by inflammatory stimuli in an
                                                                                                                                                                                                                                         NGF-dependent fashion. Moreover, its expression falls in these cells after peripheral axotomy, but the same stimulus apparently can
                                                                                                                                                                                                                                         induce expression in some large neurons ( 24 ). The clear effects of trkB-IgG coupled with the rather modest phenotype of substance P and
                                                                                                                                                                                                                                         NK1 null-mutant animals ( 25 ) may indicate that BDNF is a more important mediator of these central changes, and antagonism of this
                                                                                                                                                                                                                                         molecule may provide a novel therapeutic target.
                                                                                                                                                                                                                                               Modulation of gene expression. In addition to the effect of BDNF on short-term modulation of the NMDA receptor, there is a strong
                                                                                                                                                                                                                                         possibility that trk receptor activation and phosphorylation may underlie more prolonged changes in synaptic excitability. Kang and
                                                                                                                                                                                                                                         Schuman ( 26 ) showed that some effects of BDNF on synaptic plasticity in the hippocampus may occur in the presence of the NMDA
                                                                                                                                                                                                                                         receptor antagonist AP-5, suggesting that alternative mechanisms may exist. There is now evidence that BDNF will interact with signal
                                                                                                                                                                                                                                         transduction pathways that underlie longer-lasting forms of synaptic plasticity ( 27 ). We have examined the ability of BDNF to induce
                                                                                                                                                                                                                                         alterations in immediate early genes expressed in dorsal horn neurons in the spinal cord ( 18 ). Intrathecal administration of BDNF to adult
                                                                                                                                                                                                                                         rats induces a large increase in c-fos immunoreactivity in the superficial dorsal horn ( Fig. 3 ), whereas NGF does not. In the spinal cord, it
                                                                                                                                                                                                                                         is likely therefore that BDNF interacts with signal transduction pathways to induce long-term changes in synaptic excitability that depend
                                                                                                                                                                                                                                         on gene transcription and protein synthesis. It is well recognized that activity in primary sensory neurons, and specifically in nociceptive
                                                                                                                                                                                                                                         sensory neurons, is capable of inducing c-fos in spinal
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 5. Potential mechanisms by which BDNF may influence spinal nociceptive processing in normal and inflammatory conditions.
                                                                                                                                                                                                                                           See text for discussion. mem. pot., membrane potential; AMPA,  - amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid.
                                                                                                                                                                                                                                         BRAIN-DERIVED NEUROTROPHIC FACTOR IS AN ENDOGENOUS MODULATOR OF NOCICEPTIVE RESPONSES IN THE                                                          7718
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
SPINAL CORD
                                                                                                                                                                                                                                         neurons. However, it is not yet clear whether the c-fos induced by BDNF represents some nociceptive action or it relates to some other
                                                                                                                                                                                                                                         function.
                                                                                                                                                                                                                                               Endogenous BDNF Contributes to Nociceptive Responses. A key question for the hypothesis put forward here is whether
                                                                                                                                                                                                                                         endogenous BDNF contributes to behavioral responses after peripheral injury. To address this important question, we attempted to
                                                                                                                                                                                                                                         neutralize synaptically released BDNF with the sequestering antibody trkB-IgG. This molecule is a recombinantly produced fusion protein
                                                                                                                                                                                                                                         based on the extracellular domain of the trkB receptor. It is soluble and has the ability to bind with high affinity and thereby sequester
                                                                                                                                                                                                                                         endogenous BDNF (as well as the other trkB ligand, neurotrophin-4/5). It effectively serves as an antagonist of BDNF.
                                                                                                                                                                                                                                               We have examined the effects of trkB-IgG on nociceptive afferent-evoked reflex responses in the spinal cord. We have again used the
                                                                                                                                                                                                                                         in vitro hemisected spinal-cord preparation and maximized levels of BDNF within the spinal cord by pretreating animals with NGF. Under
                                                                                                                                                                                                                                         such circumstances, the afferentevoked response is reduced significantly after superfusion with trkB-IgG ( Fig. 2 c), strongly suggesting
                                                                                                                                                                                                                                         that BDNF is released from afferent fibers under these conditions.
                                                                                                                                                                                                                                               To date, most of the work that has examined the effects of BDNF on synaptic transmission has been undertaken on isolated
                                                                                                                                                                                                                                         preparations in vitro. There is evidence, howeveragain from work on the hippocampusthat BDNF will induce long-lasting
                                                                                                                                                                                                                                         enhancement of synaptic transmission in vivo ( 28 ). We have recently tested this idea more stringently by assessing the effect of trkB-IgG
                                                                                                                                                                                                                                         on nociceptive behavioral responses in adult rats. Behavioral nociceptive responses evoked after subcutaneous injection of dilute formalin
                                                                                                                                                                                                                                         into one hind paw were significantly reduced by prior intrathecal administration of trkB-IgG ( Fig. 4 ). Both phases of the formalin
                                                                                                                                                                                                                                         response were affected, although the second phase, known to depend in part on the induction of central sensitization, was reduced to a
                                                                                                                                                                                                                                         greater degree.
                                                                                                                                                                                                                                               Thus, these findings show that under some conditions, specifically those that model persistent pain states (e.g., inflammation), BDNF
                                                                                                                                                                                                                                         is released with activity from nociceptive afferent terminals and contributes to the postsynaptic responses in spinal-cord neurons.
                                                                                                                                                                                                                                                                                                       CONCLUSIONS
                                                                                                                                                                                                                                              As we review here, BDNF meets many of the criteria necessary to establish it as a neurotransmitter/neuromodulator in small-diameter
                                                                                                                                                                                                                                         nociceptive neurons. Thus, it is synthesized by these neurons and packaged in dense core vesicles. The BDNF-expressing nociceptive
                                                                                                                                                                                                                                         afferents terminate mostly in the superficial dorsal horn, and the postsynaptic cells in this region express fall-length trkB receptors.
                                                                                                                                                                                                                                         However, the synaptic apposition of BDNF containing afferent profiles on neurons with trkB has not yet been established. Spinal neurons
                                                                                                                                                                                                                                         are responsive to exogenous BDNF, as evidenced both histochemically (by the induction of c-fos) and electrophysiologically (by an
                                                                                                                                                                                                                                         increased excitability to nociceptive inputs). BDNF is thus sufficient to elicit changes in postsynaptic excitability. Although the release of
                                                                                                                                                                                                                                         BDNF from nociceptive fibers with activity has not been shown, there are both electrophysiological and behavioral data indicating that
                                                                                                                                                                                                                                         antagonism of BDNF at least partially prevents some aspects of central sensitization. Thus, BDNF is also necessary for the full expression
                                                                                                                                                                                                                                         of this phenomenon.
                                                                                                                                                                                                                                              Currently, the mechanism of action of BDNF is not well understood. In Fig. 5 , we illustrate several events that may contribute to the
                                                                                                                                                                                                                                         central modulatory role of BDNF. BDNF is constitutively expressed in some nociceptors. In normal animals, some BDNF is likely to
                                                                                                                                                                                                                                         released with noxious stimulation; however, such release may have limited postsynaptic effects, because much of the trkB receptor protein
                                                                                                                                                                                                                                         may not be present at the postsynaptic cell surface. Glutamate, also released with activity in nociceptors, may also have only restricted
                                                                                                                                                                                                                                         postsynaptic actions because of the limited responsiveness of NMDA receptors in these normal states. With peripheral inflammation,
                                                                                                                                                                                                                                         however, BDNF may play a much greater role. The expression is markedly up-regulated in trkA-expressing nociceptors in an NGF-
                                                                                                                                                                                                                                         dependent fashion. More BDNF is likely to be released with activity. The increased activity of postsynaptic neurons in these states may
                                                                                                                                                                                                                                         also lead to translocation of trkB receptors to the cell surface. The increased activation of trkB receptors in turn may lead to
                                                                                                                                                                                                                                         phosphorylation and increased responsiveness of NMDA receptors, perhaps independently of induction of immediate early genes.
                                                                                                                                                                                                                                         Together, these actions potentiate the efficacy of synaptic inputs from nociceptors and thus may contribute to the abnormal pain sensitivity
                                                                                                                                                                                                                                         found in these inflammatory conditions.
                                                                                                                                                                                                                                              We would like to thank Matt Ramer for useful comments regarding the manuscript. Some of the work described in this article was
                                                                                                                                                                                                                                         supported by grants from the Medical Research Council of the United Kingdom, the Special Trustees of St. Thomas Hospital, and the
                                                                                                                                                                                                                                         Physiological Society (London).
                                                                                                                                                                                                                                         1. Snider, W. D. & McMahon, S. B. ( 1998 ) Neuron 20 , 629632 .
                                                                                                                                                                                                                                         2. McMahon, S. B. , Bennett, D. L. H. , Michael, G. J. & Priestley J. V. ( 1997 ) in Neurotrophic Factors and Pain , eds. Jensen, T. S. , Turner, J. A. &
                                                                                                                                                                                                                                              Wiesenfeld-Hallin, Z. ( IASP , Seattle ), pp. 353379 .
                                                                                                                                                                                                                                         3. Carroll, P. , Lewin, G. R. , Koltzenburg, M. , Toyka, K. V. & Thoenen, H. ( 1998 ) Nat. Neurosci. 1 , 4246 .
                                                                                                                                                                                                                                         4. DiStefano, P. S. , Friedman, B. , Radziejewski, C. , Alexander, C. , Boland, P. , Schick, C. M. , Lindsay, R. M. & Wiegand, S. J. ( 1992 ) Neuron 8 ,
                                                                                                                                                                                                                                              983993 .
                                                                                                                                                                                                                                         5. Ernfors, P. , Wetmore, C. , Olson, L. & Persson, H. ( 1990 ) Neuron 5 , 511526 .
                                                                                                                                                                                                                                         6. Apfel, S. C. , Wright, D. E. , Wiideman, A. M. , Dormia, C. , Snider, W. D. & Kessler, J. A. ( 1996 ) Mol Cell Neurosci. 7 , 134142 .
                                                                                                                                                                                                                                         7. Cho, H. J. , Kim, J. K. , Zhou, X. F. & Rush, R. A. ( 1997 ) Brain Res. 764 , 269272 .
                                                                                                                                                                                                                                         8. Michael, G. J. , Averill, S. , Nitkunan, A. , Rattray, M. , Bennett, D. L. , Yan, Q. & Priestley, J. V. ( 1997 ) J. Neurosci. 17 , 84768490 .
                                                                                                                                                                                                                                         9. Zhou, X. F. & Rush, R. A. ( 1996 ) Neuroscience 74 , 945951 .
                                                                                                                                                                                                                                         10. Bennett, D. L. H. , Michael, G. J. , Ramachandran, N. , Munson, J. B. , Averill, S. , Yan, Q. , McMahon, S. B. & Priestley, J. V. ( 1998 ) J. Neurosci.
                                                                                                                                                                                                                                              18 , 30593072 .
                                                                                                                                                                                                                                         11. McMahon, S. B. & Bennett, D. L. H. ( 1999 ) in Text Book of Pain , eds. Wall, P. D. & Dubner, R. ( Churchill Livingston , Edinburgh ), 4th Ed. , in
                                                                                                                                                                                                                                              press.
                                                                                                                                                                                                                                         12. Michael, G. J. , Averill, S. , Nitkunan, A. , Rattray, M. , Bennett, D. L. H. , Yan, Q. & Priestley, J. V. ( 1997 ) J. Neurosci. 17 , 84768490 .
                                                                                                                                                                                                                                         13. Bradbury, E. J. , King, V. , Simmons, L. J. , Priestley, J. V. & McMahon, S. B. ( 1998 ) Eur. J. Neurosci. 10 , 30583068 .
                                                                                                                                                                                                                                         14. Tonra, J. R. , Curtis, R. , Wong, V. , Cliffer, K. D. , Park, J. S. , Timmes, A. , Nguyen, T. , Lindsay, R. M. , Acheson, A. & DiStefano, P. S. ( 1998 )
                                                                                                                                                                                                                                              J. Neurosci. 18 , 43744383 .
                                                                                                                                                                                                                                         15. Blochl, A. & Thoenen, H. ( 1995 ) Eur. J. Neurosci. 7 , 12201228 .
                                                                                                                                                                                                                                         16. Thompson, S. W. N. , King, A. E. & Woolf, C. J. ( 1990 ) Eur. J. Neurosci. 2 , 638649 .
                                                                                                                                                                                                                                         17. Thompson, S. W. N. ( 1998 ) Eur. J. Neurosci. 10 , Suppl. 10, 48.03 (abstr.) .
the authoritative version for attribution.
                                                                                                                                                                                                                                         18. Kerr, B. J. , Bradbury, E. J. , Bennett, D. L. H. , Trivedi, P. M. , Dassan, P. , French, J. , Shelton, D. B. , McMahon, S. B. & Thompson, S. W. N. (
                                                                                                                                                                                                                                              1999 ) J. Neurosci. 15 , in press.
                                                                                                                                                                                                                                         19. Lessmann, V. , Gottmann, K. & Heumann, R. ( 1994 ) Neuroreport 6 , 2125 .
                                                                                                                                                                                                                                         20. Levine, E. S. , Crozier, R. A. , Black, I. B. & Plummer, M. R. ( 1998 ) Proc. Natl. Acad. Sci. USA 95 , 1023510238 .
                                                                                                                                                                                                                                         21. Small, D. L. , Murray, C. L. , Mealing, G. E. A. , Poulter, M. O. , Buchan, A. M. & Morley, P. ( 1998 ) Neurosci. Lett. 252 , 211214 .
                                                                                                                                                                                                                                         22. Sucher, N. J. , Awobuluyi, M. , Choi, Y. B. & Lipton, S. A. ( 1996 ) Trends Pharmacol Sci. 17 , 348355 .
                                                                                                                                                                                                                                         23. Urban, L. , Thompson, S. W. N. & Dray, A , ( 1994 ) Trends Neurosci. 17 , 432438 .
                                                                                                                                                                                                                                         24. Noguchi, K. , Dubner, R. , De Leon, M. , Senba, E. & Ruda, M. A. ( 1994 ) J. Neurosci. Res. 37 , 596603 .
                                                                                                                                                                                                                                         25. Woolf, C. J. , Mannion, R. J. & Neumann, S. ( 1998 ) Neuron 20 , 10631066 .
                                                                                                                                                                                                                                         26. Kang, H. & Schuman, E. M. ( 1995 ) Science 267 , 16581662 .
                                                                                                                                                                                                                                         27. Kang, H. , Welcher, A. A. , Shelton, D. & Schuman, E. M. ( 1997 ) Neuron 19 , 653664 .
                                                                                                                                                                                                                                         28. Messaoudi, E. , Bardsen, K. , Srebo, B. & Bramham, C. R. ( 1998 ) J. Neurophysiol. 79 , 496499 .
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                         P3, the mean latency of the A-fiber-evoked response is 33.1  2.78 ms (n = 22), compared with 19.1  1.32 ms (n = 65) at P6, 13.5  0.8
                                                                                                                                                                                                                                         ms (n = 53) at P10, and 7.3  0.3 ms (n = 35) at P21. Furthermore, the variation in the A-fiber latencies within the population of recorded
                                                                                                                                                                                                                                         cells decreases with age ( 28 ).
                                                                                                                                                                                                                                               In contrast to responses to A-fiber input, no long-latency C-fiber-evoked (15 mA, 500 s) spike responses are evoked in dorsal horn
                                                                                                                                                                                                                                         cells in vivo in the first postnatal week ( 28 , 29 ). At P10, only 7 of 53 cells (13%) have a C-fiber input with a mean latency of 97.65 
                                                                                                                                                                                                                                         4.44 ms (n = 7), and at P21 the value is 32% with a mean latency of 107.0  10.12 ms (n = 10). These results do not, of course, provide
                                                                                                                                                                                                                                         information about subthreshold C-fiber-evoked responses at this time.
                                                                                                                                                                                                                                               Convergence of Afferent Inputs in the Postnatal Dorsal Horn Cells. Low-threshold inputs can also be seen to dominate the
                                                                                                                                                                                                                                         newborn dorsal horn when the responses to natural stimulation are examined. Background activity is generally absent when neonatal cells
                                                                                                                                                                                                                                         are initially isolated for recording, but strong responses can be evoked by mechanical stimulation of the skin of the receptive field. Some
                                                                                                                                                                                                                                         cells respond to both innocuous brushing and noxious pinching of the skin, but the convergence of input to dorsal horn cells changes over
                                                                                                                                                                                                                                         the postnatal period ( Table 1 ). The responses recorded from cells in the younger animals are elicited mainly by low-threshold
                                                                                                                                                                                                                                         mechanoreceptors, and there are few cells with convergent input in the first week of life. This population gradually increases so that by
                                                                                                                                                                                                                                         P21 the percentage of neurones with convergent primary afferent input is similar to that seen in the adult.
                                                                                                                                                                                                                                               Receptive Fields of Postnatal Dorsal Horn Cells. The size of dorsal horn cell peripheral cutaneous receptive field decreases with
                                                                                                                                                                                                                                         age ( 30 ). We have studied this in more detail using those cells with mechanoreceptive fields on the plantar surface of the hindpaw.
                                                                                                                                                                                                                                         Receptive field areas were scanned into a computer, converted into pixels by using Adobe PHOTOSHOP software (Adobe Systems,
                                                                                                                                                                                                                                         Mountain View, CA) and expressed as a percentage of the entire plantar surface of the hindpaw. At P3, the mean (SE) peripheral
                                                                                                                                                                                                                                         receptive field occupies 50  5.6% of the plantar hindpaw. This value drops to 36  2.9% at P6, 20  1.9% at P10, and 15%  1.6 at P21.
                                                                                                                                                                                                                                         The biggest change, therefore, occurs in the first postnatal week.
                                                                                                                                                                                                                                               In the neonate, therefore, receptive fields are not only dominated by low-threshold inputs but are also larger and will therefore
                                                                                                                                                                                                                                         overlap more than in the adult, increasing the chance of activation by peripheral skin stimulation.
                                                                                                                                                                                                                                               Activity-Dependent Changes in Postnatal Dorsal Horn.
                                                                                                                                                                                                                                               (i) Repetitive stimulation of receptive fields.
                                                                                                                                                                                                                                               C-fiber-evoked activity is not observed in dorsal horn cells in the first postnatal week, and repetitive peripheral stimulation at C-fiber
                                                                                                                                                                                                                                         strength also has no observable effects on dorsal horn cell spike responses. From P10, repetitive C-fiber stimulation at three times the C-
                                                                                                                                                                                                                                         fiber threshold produces a classical wind-up, as reported in the adult dorsal horn ( 31  37 ) in 18% of cells. This percentage has
                                                                                                                                                                                                                                         increased to 40% of cells by P21.
                                                                                                                                                                                                                                               In contrast to the lack of C-fiber influence, stimulation of cells at twice the A-fiber threshold at a frequency of 0.5 Hz through pin
                                                                                                                                                                                                                                         electrodes placed in the center of the peripheral receptive field on the hindlimb can produce considerable sensitization of the dorsal horn
                                                                                                                                                                                                                                         cells in the neonate ( 28 ). This sensitization takes the form of a buildup of background activity in the cells during repetitive stimulation
                                                                                                                                                                                                                                         that outlasts the stimulation period, thereby producing a prolonged after-discharge of up to 138 s. It is particularly apparent in younger
                                                                                                                                                                                                                                         animals, and at P6, 19 of 57 cells (33%) display background firing during, and a prolonged after-discharge of, 70.6  18 s after repetitive
                                                                                                                                                                                                                                         A-fiber stimulation. At P10, 3 of 48 cells showed this type of sensitization (6%) with an after-discharge of 63 s, whereas at P21, it was not
                                                                                                                                                                                                                                         seen in any cells (n = 31) ( 28 ).
                                                                                                                                                                                                                                         Table 1. Convergence of afferent input to dorsal horn cells at different postnatal ages
                                                                                                                                                                                                                                                                             Cells with different input
                                                                                                                                                                                                                                         Age                                 Brush                          Pinch                         Brush and Pinch
                                                                                                                                                                                                                                         P3 (n = 22)                         20 (91%)                       1 (4.5%)                      1 (4.5%)
                                                                                                                                                                                                                                         P6 (n = 65)                         54 (83%)                       7 (11%)                       4 (16%)
                                                                                                                                                                                                                                         P10 (n = 53)                        22 (42%)                       12 (22%)                      19 (36%)
                                                                                                                                                                                                                                         P21 (n = 35)                        10 (29%)                       5 (14%)                       20 (57%)
                                                                                                                                                                                                                                              A-fiber-induced sensitization is not accompanied by an increase in the direct A-fiber-evoked spike discharge, but during the
                                                                                                                                                                                                                                         stimulation period, the sensitized units show a significant increase in activity outside of this short-latency evoked burst ( 28 ). The mean
                                                                                                                                                                                                                                         activity during the stimulation period, measured in the 40- to 2,000-ms period between stimuli, is 2.6  0.16 spikes in P6 sensitizing cells,
                                                                                                                                                                                                                                         significantly greater (p < 0.0001) than the 0.4  0.04 spikes in nonsensitizing cells. At P10, there is a similar pattern; the mean background
                                                                                                                                                                                                                                         activity for sensitizing cells was 15.7  0.84 spikes, whereas that of nonsensitized cells was 1.3  0.13 spikes, another significant
                                                                                                                                                                                                                                         difference (p < 0.0001).
                                                                                                                                                                                                                                              (ii) Experimental inflammation in rat pups.
                                                                                                                                                                                                                                              Carrageenan is reported to be a reliable agent in modeling inflammation in adults ( 38  44 ). After subcutaneous injection, edema
                                                                                                                                                                                                                                         develops rapidly, followed by hyperalgesia, which peaks at 34 hr and decreases to baseline by 2472 hr ( 40  42 ). In some cases, the
                                                                                                                                                                                                                                         period of hyperalgesia can last 1014 days ( 40 ). In view of the differences in sensory processing in the newborn compared with the adult
                                                                                                                                                                                                                                         dorsal horn, we examined the responses of newborn dorsal horn cells to a carrageenan inflammatory stimulus.
                                                                                                                                                                                                                                              The allodynia or drop in mechanical threshold that follows carrageenan injection ( 11 , 40 ) and hyperalgesia after mustard oil
                                                                                                                                                                                                                                         application ( 15 ) is clear, but smaller in amplitude, in neonates. Carrageenan-induced inflammation produces a parallel fall in von Frey
                                                                                                                                                                                                                                         thresholds at P3 and P10, whereas P21 animals show a significantly greater effect. At all ages, the effect increases with time, reaching a
                                                                                                                                                                                                                                         maximum at 4 hr postinjection, but is still maintained at 5 hr postinjection ( 11 ).
                                                                                                                                                                                                                                              This finding agrees with the responses of dorsal horn cells at this time. After carrageenan injection, dorsal horn cell receptive fields
                                                                                                                                                                                                                                         recorded in anesthetized rat pups in vivo were measured and expressed as a percentage of the plantar foot area ( Fig. 1 ). There was a
                                                                                                                                                                                                                                         significant increase in the size of peripheral receptive fields in animals in both the P10 and P21 age groups (P < 0.0001 in both cases). At
                                                                                                                                                                                                                                         P10, the size of the peripheral receptive fields increased 2.5-fold, and at P21 the increase was 3.4-fold. Mean size SEM (as a percentage
                                                                                                                                                                                                                                         of the plantar hindpaw area) of the peripheral receptive field in the inflamed group at P10 was 47.2  6.4%, and that of the control was
                                                                                                                                                                                                                                         19.1  2.0%. At P21, the mean size of the receptive field in the inflamed group was 51.8  12.2% and that of the control group was 14.9 
the authoritative version for attribution.
                                                                                                                                                                                                                                         1.6%.
                                                                                                                                                                                                                                              The receptive fields of adult dorsal horn neurons observed between 4 and 8.5 hr after injection of complete Freunds adjuvant
                                                                                                                                                                                                                                         expanded to 2.4 times their original size ( 45 ). This increase of the receptive field may be responsible for hyperalgesia. Because the
                                                                                                                                                                                                                                         receptive fields are larger, there is a greater degree of overlap, and so a single stimulus would activate many more neurons than in the
                                                                                                                                                                                                                                         control state, a summative effect ( 46 ).
                                                                                                                                                                                                                                              Effects of Primary Afferent Stimulation. The magnitude of the evoked response (number of spikes) after electrical stimulation at
                                                                                                                                                                                                                                         twice A-fiber threshold directly to the nerve significantly increased after inflammation in P10 cells. Mean  SEM evoked response for the
                                                                                                                                                                                                                                         control animals was 3.2  0.25 spikes, and for the inflamed animals it was 7.6  0.21 spikes. The Students t test gives a p < 0.0001 when
                                                                                                                                                                                                                                         comparing these
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                           FIG. 1. Peripheral receptive fields: change with age and after inflammation. Representations of the peripheral receptive fields. Those
                                                                                                                                                                                                                                           in black represent cells receiving afferents from inflamed skin, whilst those in grey are the control. The mean receptive field size
                                                                                                                                                                                                                                           SEM (as a percentage of the total plantar foot area) is 50  5.6% at P3, 36  2.9% at P6, 20  1.97% at P10, and 15  1.62% at P21.
                                                                                                                                                                                                                                           For those with inflamed feet, the receptive field sizes are 47  6.4% at P10 and 52  12.2% at P21.
                                                                                                                                                                                                                                              In addition, the number of cells with C-fiber-evoked activity increased from 0/6 in the control group to 4/7 in the inflamed group.
                                                                                                                                                                                                                                         Even such small numbers suggest an important effect of the carrageenan-induced central excitation on unmasking C-fiber-evoked spike
                                                                                                                                                                                                                                         activity.
                                                                                                                                                                                                                                              At P21, the mean magnitude of the evoked response for cells receiving an A afferent input was 6.8  0.32 spikes in the control group
                                                                                                                                                                                                                                         and 6.6  0.21 spikes in the inflamed group. The difference between these two groups is not significant (P = 0.59). For those cells
                                                                                                                                                                                                                                         responding to C afferent input, the magnitude of response was 4.9  0.5 spikes for the control group and 10.1  0.67 spikes for the
                                                                                                                                                                                                                                         inflamed group. These two results were significantly different, with a P < 0.0001.
                                                                                                                                                                                                                                              A Possible Role for N-methyl-D-aspartate (NMDA) Receptors. The results demonstrate important differences in the synaptic
                                                                                                                                                                                                                                         connectivity underlying sensory processing in the newborn spinal cord. The slow maturation of C-fiber afferent input appears to result in a
                                                                                                                                                                                                                                         predominance of A-fiber-evoked activity, such that processes that are exclusively activated by small-diameter nociceptive inputs in adults
                                                                                                                                                                                                                                         can be triggered by low-threshold large-diameter inputs in the first postnatal weeks.
                                                                                                                                                                                                                                              There are likely to be several mechanisms underlying this transient state of A-fiber-induced excitation, but we would like to propose
                                                                                                                                                                                                                                         that one important one could be in the developmental regulation of NMDA receptors.
                                                                                                                                                                                                                                              The neonatal spinal cord has a higher concentration of NMDA receptors in the grey matter than that observed in older animals ( 47 ).
                                                                                                                                                                                                                                         All laminae in the dorsal horn are uniformly labeled with NMDA-sensitive [3H]glutamate until day 1012, when higher densities
                                                                                                                                                                                                                                         gradually appear in substantia gelatinosa so that by P30, binding is similar to that in the adult. Furthermore, the affinity of the receptors for
                                                                                                                                                                                                                                         NMDA and the NMDA-evoked calcium efflux in rat substantia gelatinosa is high in the first postnatal week and then declines ( 48 ). This
                                                                                                                                                                                                                                         maturation is delayed by neonatal capsaicin treatment, suggesting that C-fiber afferent activity regulates the postnatal maturation of
                                                                                                                                                                                                                                         NMDA receptors ( 48 ). There is also considerable rearrangement of the subunit composition of the NMDA channel complex during spinal
                                                                                                                                                                                                                                         cord development ( 49 ).
                                                                                                                                                                                                                                              This study was supported by a European Community Biomedicine and Health grant (BMH4-CT95-0172). E.A.J. is a graduate student
                                                                                                                                                                                                                                         supported by the Medical Research Council of Great Britain and the Department of Anatomy, University College London.
                                                                                                                                                                                                                                         1. Ekholm, J. ( 1967 ) Acta Physiol Scand. Suppl. 297 , 1130 .
                                                                                                                                                                                                                                         2. Issler, H. & Stephens, J. A. ( 1983 ) J. Physiol. (London) 335 , 643654 .
                                                                                                                                                                                                                                         3. Fitzgerald, M. & Gibson, S. ( 1984 ) Neuroscience 13 , 933944 .
                                                                                                                                                                                                                                         4. Andrews, K. & Fitzgerald, M. ( 1994 ) Pain 56 , 95101 .
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         5. Fitzgerald, M. , Shaw, A. & MacIntosh, N. ( 1988 ) Dev. Med. Child Neurol. 30 , 520526 .
                                                                                                                                                                                                                                         6. Marsh, D. , Dickenson, A. H. , Hatch, D. & Fitzgerald, M. ( 1999 ) Pain , in press.
                                                                                                                                                                                                                                         7. Holmberg, H. & Schouenborg, J. ( 1996 ) J. Physiol. (London) 493 , 239252 .
                                                                                                                                                                                                                                         8. Lewin, G. R. , Ritter, A. M. & Mendell, L. M. ( 1993 ) J. Neurosci. 13 , 21362148 .
                                                                                                                                                                                                                                         9. Falcon, M. , Guendellman, D. , Stolberg, A. , Frenk, H. & Urca, G. ( 1996 ) Pain 67 , 203208 .
                                                                                                                                                                                                                                         10. Hu, D. , Hu, R. & Berde, C. B. ( 1997 ) Anesthesiology 86 , 957965 .
                                                                                                                                                                                                                                         11. Marsh, D. , Dickenson, A. H. , Hatch, D. & Fitzgerald, M. ( 1999 ) Pain , in press.
                                                                                                                                                                                                                                         12. Guy, E. R. & Abbott, F. V. ( 1992 ) Pain 51 , 8190 .
                                                                                                                                                                                                                                         13. Teng, C. J. & Abbott, F. V. ( 1998 ) Pain 76 , 337347 .
                                                                                                                                                                                                                                         14. Fitzgerald, M. & Gibson, S. ( 1984 ) Neuroscience 13 , 933944 .
                                                                                                                                                                                                                                         15. Jiang, M. C. & Gebhart, G. F. ( 1998 ) Pain 77 , 305313 .
                                                                                                                                                                                                                                         16. Fitzgerald, M. ( 1987 ) J. Physiol. (London) 383 , 7992 .
                                                                                                                                                                                                                                         17. Smith, C. L. ( 1983 ) J. Comp. Neurol. 220 , 2943 .
                                                                                                                                                                                                                                         18. Fitzgerald, M. , Reynolds, M. L. & Benowitz, L. I. ( 1991 ) Neuroscience 41 , 187199 .
                                                                                                                                                                                                                                         19. Fitzgerald, M. ( 1987 ) J. Comp. Neurol. 261 , 98104 .
                                                                                                                                                                                                                                         20. Pignatelli, D. , Ribeiro da Silva, A. & Coimbra, A. ( 1989 ) Brain Res. 491 , 3344 .
                                                                                                                                                                                                                                         21. Fitzgerald, M. & Swett, J. ( 1983 ) Neurosci. Lett. 43 , 149154 .
                                                                                                                                                                                                                                         22. Ozaki, S. & Snider, W. D. ( 1997 ) J. Comp. Neurol. 380 , 215229 .
                                                                                                                                                                                                                                         23. Fitzgerald, M. , Butcher, T. & Shortland, P. ( 1994 ) J. Comp. Neurol. 348 , 225233 .
                                                                                                                                                                                                                                         24. Mirnics, K. & Koerber, H. R. ( 1995 ) J. Comp. Neurol. 355 , 601614 .
                                                                                                                                                                                                                                         25. Coggeshall, R. E. , Jennings, E. A. & Fitzgerald, M. ( 1996 ) Brain Res. Dev. Brain Res. 92 , 8190 .
                                                                                                                                                                                                                                         26. Jennings, E. & Fitzgerald, M. ( 1996 ) Pain 68 , 301306 .
                                                                                                                                                                                                                                         27. Ma, Q. P. & Woolf, C. J. ( 1996 ) Pain 67 , 307316 .
                                                                                                                                                                                                                                         28. Jennings, E. & Fitzgerald, M. ( 1998 ) J. Physiol. (London) 509 , 859868 .
                                                                                                                                                                                                                                         29. Fitzgerald, M. ( 1988 ) Neurosci. Lett. 86 , 161166 .
                                                                                                                                                                                                                                         30. Fitzgerald, M. ( 1985 ) J. Physiol. (London) 364 , 118 .
                                                                                                                                                                                                                                         31. Mendell, L. M. & Wall, P. D. ( 1965 ) Nature (London) 206 , 9799 .
                                                                                                                                                                                                                                         32. Mendell, L. M. ( 1966 ) Exp. Neurol. 16 , 316332 .
                                                                                                                                                                                                                                         33. Woolf, C. J. , Thompson, S. W. & King, A. E. ( 1988 ) J. Physiol. (Paris) 83 , 255266 .
                                                                                                                                                                                                                                         34. Davies, S. N. , Martin, D. , Millar, J. D. , Aram, J. A. , Church, J. & Lodge, D. ( 1988 ) Eur. J. Pharmacol. 145 , 141151 .
                                                                                                                                                                                                                                         35. Dickenson, A. H. & Sullivan, A. F. ( 1990 ) Brain Res. 506 , 3139 .
                                                                                                                                                                                                                                         36. Thompson, S. W. , Woolf, C. J. & Sivilotti, L. G. ( 1993 ) J. Neurophysiol. 69 , 21162128 .
                                                                                                                                                                                                                                         37. Woolf, C. J. & Thompson, S. W. ( 1991 ) Pain 44 , 293299 .
                                                                                                                                                                                                                                         38. Xu, X. J. , Elfvin, A. & Wiesenfeld Hallin, Z. ( 1995 ) Neurosci. Lett. 196 , 116118 .
                                                                                                                                                                                                                                         39. Winter, C. A. , Risley, E. A. & Nuss, G. W. ( 1962 ) Proc. Soc. Exp. Biol. Med. 111 , 544547 .
                                                                                                                                                                                                                                         40. Kayser, V. & Guilbaud, G. ( 1987 ) Pain 28 , 99107 .
                                                                                                                                                                                                                                         41. Hargreaves, K. , Dubner, R. , Brown, F. , Flores, C. & Joris, J. ( 1988 ) Pain 32 , 7788 .
                                                                                                                                                                                                                                         42. Hylden, J. L. , Thomas, D. A. , Iadarola, M. J. , Nahin, R. L. & Dubner, R. ( 1991 ) Eur. J. Pharmacol. 194 , 135143 .
                                                                                                                                                                                                                                         43. Stanfa, L. C. , Sullivan, A. F. & Dickenson, A. H. ( 1992 ) Pain 50 , 345354 .
                                                                                                                                                                                                                                         44. Stanfa, L. C. , Misra, C. & Dickenson, A. H. ( 1996 ) Brain Res. 737 , 9298 .
                                                                                                                                                                                                                                         45. Hylden, J. L. , Nahin, R. L. , Traub, R. J. & Dubner, R. ( 1989 ) Pain 37 , 229243 .
                                                                                                                                                                                                                                         46. Dubner, R. & Ruda, M. A. ( 1992 ) Trends Neurosci. 15 , 96103 .
                                                                                                                                                                                                                                         47. Gonzalez, D. L. , Fuchs, J. L. & Droge, M. H. ( 1993 ) Neurosci. Lett. 151 , 134137 .
                                                                                                                                                                                                                                         48. Hori, Y. & Kanda, K. ( 1994 ) Dev. Brain Res. 80 , 141148 .
                                                                                                                                                                                                                                         49. Watanabe, M. , Mishina, M. & Inoue, Y. ( 1994 ) J. Comp. Neurol. 345 , 314319 .
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
TRANSCRIPTIONAL AND POSTTRANSLATIONAL PLASTICITY AND THE GENERATION OF INFLAMMATORY PAIN 7723
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                         Harvard Medical School, 149 13th Street, Room 4310, Charlestown, MA 02129. e-mail: [email protected] .
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
TRANSCRIPTIONAL AND POSTTRANSLATIONAL PLASTICITY AND THE GENERATION OF INFLAMMATORY PAIN 7724
                                                                                                                                                                                                                                         dorsal root ganglion (DRG) and dorsal horn that elicit these changes, from the mediators to the receptors through the intracellular signal-
                                                                                                                                                                                                                                         transduction cascades and finally culminating on the posttranslational and transcriptional changes that result.
                                                                                                                                                                                                                                           FIG. 1. State-dependent sensory processing in the somatosensory system. (A) The basal sensitivity of the system is normally such that
                                                                                                                                                                                                                                           only a high-intensity C or A stimulus produces pain. (B) C fiber inputs induce immediate posttranslational changes in the dorsal
                                                                                                                                                                                                                                           horn, resulting in central sensitization and altering basal sensitivity such that low intensity stimuli result in pain. (C) Conditioning C
                                                                                                                                                                                                                                           fiber strength inputs induce activity-dependent transcriptional changes in the DRG and dorsal horn, resulting in a potentiated system
                                                                                                                                                                                                                                           augmenting responsiveness to subsequent C fiber inputs. (D) Inflammation results in both a potentiated system and phenotypic
                                                                                                                                                                                                                                           switches such that both C fiber and low-intensity A fiber inputs can evoke central sensitization.
                                                                                                                                                                                                                                               State One: Nociception (Normal Transmission). The activation of high-threshold nociceptive primary afferent neurons, mainly
                                                                                                                                                                                                                                         unmyelinated C fibers but A6 fibers as well, by intense but nondamaging stimuli, results within seconds in a transient, well localized pain
                                                                                                                                                                                                                                         that does not require long-term nontranscriptional or transcriptional changes. Primary sensory neurons have three clear functions with
                                                                                                                                                                                                                                         respect to their role in nociception: transduction of noxious but not low-intensity peripheral stimuli; conduction of action potentials to the
                                                                                                                                                                                                                                         central nervous system; and transmission to central neurons. Distinct transducers, receptors, ion channels, and transmitters mediate these
                                                                                                                                                                                                                                         functions. Tables 1 and 2summarize the key ion channels and receptors expressed in dorsal root ganglion cells, the functions they mediate,
                                                                                                                                                                                                                                         and their posttranslational modulation or transcriptional regulation.
                                                                                                                                                                                                                                               Nociceptive transduction. Peripheral nociceptive transduction involves the detection of hot and cold (but not warm or cool) thermal
                                                                                                                                                                                                                                         stimuli and intense (but not innocuous) mechanical stimuli, as well as a sensitivity to chemical irritants such as the pungent ingredient in
                                                                                                                                                                                                                                         chili peppers, capsaicin. This sensitivity is mediated by multiple specialized receptors, including heat-sensitive ion channels like the
                                                                                                                                                                                                                                         vanniloid receptor VR1, which is gated by protons and activated by capsaicin ( 4 , 5 ), channels sensitive to protons and possibly
                                                                                                                                                                                                                                         mechanical stimuli, the Na+/degenerin family including ASIC (acid-sensing ionic channel) and DRASIC (dorsal root acid-sensing ionic
                                                                                                                                                                                                                                         channel) ( 6 ), and receptors sensitive to chemical stimuli alone (the histamine, bradykinin, purine, and serotonin receptors) ( 7  10 ).
                                                                                                                                                                                                                                         Chemical stimuli are a very prominent component of inflammatory pain, where they are generated as part of the inflammatory response
                                                                                                                                                                                                                                         (see below). A chemical component of nociception occurs only on exposure to nondamaging external chemical irritants, such as plant or
                                                                                                                                                                                                                                         insect stings. The relative expression of these transduction elements in specific subsets of nociceptor sensory neurons is beginning to
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
TRANSCRIPTIONAL AND POSTTRANSLATIONAL PLASTICITY AND THE GENERATION OF INFLAMMATORY PAIN 7725
                                                                                                                                                                                                                                         reveal specialization of modality sensitivity. The purine receptor P2X3, for example, is expressed only on glial cell linederived
                                                                                                                                                                                                                                         neurotrophic factor-responsive c-Ret-expressing C fibers ( 11 ), whereas VR1 is expressed both in these cells and in the TrkA-expressing
                                                                                                                                                                                                                                         nerve growth factor (NGF)-responsive neurons ( 5 ).
                                                                                                                                                                                                                                              Nociceptive conduction. Voltage-gated sodium channels, which are responsible for the rising phase of the action potential and play a
                                                                                                                                                                                                                                         key role, with potassium channels, in determining the excitability of the sensory neurons, mediate conduction in nociceptors by
                                                                                                                                                                                                                                         transferring input from the peripheral nerve terminals to the spinal cord. Neuronal voltage-gated sodium channels can be classified into
                                                                                                                                                                                                                                         two types: those sensitive to nanomolar concentrations of the puffer fish toxin (tetrodotoxin-sensitive, TTXs) and those resistant to all but
                                                                                                                                                                                                                                         micromolar concentrations of tetrodotoxin (tetrodotoxinresistant, TTXr) ( 12 ). DRG neurons express several distinct kinetic types of
                                                                                                                                                                                                                                         sodium current. Small-diameter, highthreshold nociceptor neurons coexpress a rapidly inactivating, fast TTXs current and a slowly
                                                                                                                                                                                                                                         activating and inactivating TTXr sodium current ( 13 , 14 ). Large diameter cells only express a TTXs sodium current ( 13 ).
                                                                                                                                                                                                                                              Within the DRG, several voltage-gated sodium channels are known to be responsible for the TTXs current (PN1, rSCP6/ PN4, rBI,
                                                                                                                                                                                                                                         rBII, and rBIII), and these show a wide distribution across large and small neuronal cells ( 15  17 ). Two sensory neuron-specific TTXr
                                                                                                                                                                                                                                         sodium channels have been cloned [SNS/PN3 ( 18 , 19 ) and SNS2/NaN ( 20 , 21 )]. SNS2/NaN is found only in small sensory neurons,
                                                                                                                                                                                                                                         where it is colocalized with SNS/PN3 ( 20 ), with which it may form functional heteromultimers, generating the native TTXr current. SNS/
                                                                                                                                                                                                                                         PN3 is also found without SNS2 in a subpopulation of larger neurons ( 20 ). The expression of TTXr sodium channels in nociceptive
                                                                                                                                                                                                                                         neurons points to a specific role for these channels in contrib
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
TRANSCRIPTIONAL AND POSTTRANSLATIONAL PLASTICITY AND THE GENERATION OF INFLAMMATORY PAIN 7726
                                                                                                                                                                                                                                         uting to the excitability of nociceptors, although the TTXs current may be sufficient for conduction, and the TTX current plays more of a
                                                                                                                                                                                                                                         modulatory role in the peripheral terminals and in generating ectopic inputs.
                                                                                                                                                                                                                                              Nociceptive transmission. Transfer of synaptic input from nociceptors to specific laminae in the dorsal horn is highly topographically
                                                                                                                                                                                                                                         organized and activates particular subsets of second-order projection neurons ultimately leading, following activation of specific brain
                                                                                                                                                                                                                                         centers, to the sensation of acute pain as well emotional, cognitive, and autonomic responses. Primary afferent neurons, by virtue of their
                                                                                                                                                                                                                                         peripheral transduction specialization, central termination site, or temporal characteristics, encode stimulus modality, intensity, location,
                                                                                                                                                                                                                                         and duration ( 22 ). This is relayed with great fidelity to second-order neurons. Synaptic transmission for C fibers is mediated by glutamate
                                                                                                                                                                                                                                         acting on -amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors on dorsal horn neurons, generating fast excitatory
                                                                                                                                                                                                                                         postsynaptic potentials with a slow trailing edge contributed to by the N-methyl-D-aspartate (NMDA) receptor ( 23 , 24 ). If the stimulus
                                                                                                                                                                                                                                         intensity is high enough, release of neuropeptides from dense core vesiclesparticularly substance Pwill occur, which will, via an
                                                                                                                                                                                                                                         activation of the NK1 receptor, generate a greater postsynaptic response, reflecting the greater input and representing, therefore, a form of
                                                                                                                                                                                                                                         intensity coding ( 25 ). Antagonism or deletion of the NMDA, NK1, or mGluR receptor does not normally interfere, though, with the
                                                                                                                                                                                                                                         nociceptive response to normal non-tissue-damaging noxious stimuli ( 26 ); this is largely mediated by glutamate acting on its AMPA
                                                                                                                                                                                                                                         receptor and reflects the necessity for a high-safety, fast early warning protective pain system. The NMDA, NK1, or mGluR receptors play
                                                                                                                                                                                                                                         a key role in the initiation or maintenance of changes in synaptic transmission that constitute central sensitization ( 27  32 ), where the
                                                                                                                                                                                                                                         specific link between a noxious stimulus and pain is lost and pain can be produced by normally innocuous inputs ( 2 , 33  35 ).
                                                                                                                                                                                                                                              Neural plasticity and pain. Sensory neurons can undergo functional, chemical, and structural changes in response to changes in their
                                                                                                                                                                                                                                         environment that modify their transduction, conduction, and transmission and move them from their specific role in mediating normal
                                                                                                                                                                                                                                         nociceptive transmission to a new modified condition contributing to an altered state of sensibility. The key issues, then, are what produces
                                                                                                                                                                                                                                         these changes, how and where they manifest, and what the sensory consequences are. We will analyze this in terms of two forms of
                                                                                                                                                                                                                                         plasticity, activity- and signal molecule-dependent, and two kinds of change, that induced immediately by posttranslational modifications
                                                                                                                                                                                                                                         in the neurons and that caused by an alteration in expression of key effector molecules, which takes some time to manifest. The functional
                                                                                                                                                                                                                                         significance of this plasticity, both for basal sensibility and stimulus-evoked hypersensitivity, will be discussed.
                                                                                                                                                                                                                                              State Two: Immediate Changes (Posttranslational Modulation of the Dorsal Horn). The first example of stimulusevoked
                                                                                                                                                                                                                                         hypersensitivity is that of C fiber-mediated central sensitization ( 2 ). Nociceptive pain is characterized by a clear distinction in the sensory
                                                                                                                                                                                                                                         responses to a noxious or low-intensity stimulus, pain, and an innocuous sensation, respectively. Activation of C fibers, can, however,
                                                                                                                                                                                                                                         result in a situation where basal sensitivity changes such that normal low-threshold A inputs begin to evoke pain and tactile allodynia and
                                                                                                                                                                                                                                         noxious inputs provoke a greater pain response, hyperalgesia (refs. 2 and 33  35 ; Fig. 1 ). Central sensitization, which in normal
                                                                                                                                                                                                                                         noninflamed situations, can be produced only by C fiber input ( 36 ), is the direct manifestation of a complex set of activity-dependent
                                                                                                                                                                                                                                         posttranslational changes in dorsal horn neurons. Relatively brief C fiber inputs (lasting only tens of seconds) can initiate very rapid
                                                                                                                                                                                                                                         changes in membrane excitability, which manifest both as a progressive increase in excitability during the course of the stimulus, windup,
                                                                                                                                                                                                                                         and poststimulus changes that can last for several hours (central sensitization) ( 2 , 36 ). Windup is a manifestation of the removal by
                                                                                                                                                                                                                                         successive synaptic depolarization of the voltage-dependent Mg2+ block of the NMDA receptor amplifying the response to each
                                                                                                                                                                                                                                         subsequent input. Central sensitization is quite different. Fig. 2 summarizes some of the salient features. Presynaptic transmitter/
                                                                                                                                                                                                                                         neuromodulator release [glutamate, substance P, and brain-derived neurotrophic factor (BDNF)] results in changes in signal-transduction
                                                                                                                                                                                                                                         pathways in dorsal horn neurons as a result of the activation of ligand-gated ion channels (NMDA-Rglutamate), metabotropic receptors
                                                                                                                                                                                                                                         (mGluR glutamate, NK1substance P) and tyrosine kinase receptors (TrkBBDNF). Not shown in Fig. 2 but likely to have major effects
                                                                                                                                                                                                                                         on posttranslational changes in the dorsal horn are the prostanoids. These act through prostaglandin E and prostacyclin and IP receptors
                                                                                                                                                                                                                                         and may be released pre- and postsynaptically (reviewed in ref. 37 ).
                                                                                                                                                                                                                                              Activation of these multiple receptors results in an increase in intracellular calcium, both via calcium inflow and release from
                                                                                                                                                                                                                                         intracellular stores ( 38 ) and a consequent activation of calcium-dependent enzymes (protein kinase C and calcium calmodulin kinase),
                                                                                                                                                                                                                                         protein kinase A (via G protein coupled-receptors), and tyrosine kinases (via the TrkB receptor) (see Fig. 2 ). These pathways converge
                                                                                                                                                                                                                                         and diverge in a complex fashion and are linked such that TrkB, which itself is a tyrosine kinase, also activates other tyrosine kinases src
                                                                                                                                                                                                                                         and protein kinase C ( 39 , 40 ). The targets for these different kinases are membrane-bound receptors/ion channels, of which the NMDA
                                                                                                                                                                                                                                         and AMPA are best characterized, although others are certainly involved, including activation of neuronal nitric-oxide synthase and
                                                                                                                                                                                                                                         prostaglandin production, with generation of retrograde signals to the presynaptic terminal. Phosphorylation of the NMDA receptor at
                                                                                                                                                                                                                                         either serine/threonine residues on its NR1 subunit ( 41 ) or on tyrosine residues on its NR2 subunit ( 42 , 43 ) is a major factor. This
                                                                                                                                                                                                                                         posttranslational modification results in dramatic changes in NMDA-receptor channel kinetics and a reduction in its voltage-dependent
                                                                                                                                                                                                                                         Mg2+ block ( 42 , 44 , 45 ). Both of these changes augment subsequent responsiveness to synaptically released glutamate, increasing
                                                                                                                                                                                                                                         synaptic strength and enabling previously subthreshold inputs to drive the output of the cell ( 46 ). This
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 2. Posttranslational changes within dorsal horn neurons after release of transmitters from C fiber central terminals. These
                                                                                                                                                                                                                                           transmitters/neuromodulators act on receptors and ion channels in the dorsal horn to activate protein kinases that phosphorylate
                                                                                                                                                                                                                                           membrane-bound NMDA and AMPA receptors and alter their functional properties, increasing membrane excitability and thereby
                                                                                                                                                                                                                                           eliciting central sensitization.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
TRANSCRIPTIONAL AND POSTTRANSLATIONAL PLASTICITY AND THE GENERATION OF INFLAMMATORY PAIN 7727
                                                                                                                                                                                                                                         effective increase in gain alters receptive field properties ( 46  48 ) and pain sensitivity, causing tactile allodynia and pin prick
                                                                                                                                                                                                                                         hyperalgesia, far beyond the site of the C fiber-activating stimulus that initiated the central sensitization ( 49 ).
                                                                                                                                                                                                                                              State Three: Early Changes in Transcription. C fiber inputs, in addition to the generation of central sensitization, which occurs
                                                                                                                                                                                                                                         within seconds of the appropriate activation of dorsal horn neurons, also generate an activity-dependent change in transcription in dorsal
                                                                                                                                                                                                                                         root ganglion and dorsal horn neurons, which has functional consequences that take hours to manifest. BDNF mRNA levels, for example,
                                                                                                                                                                                                                                         increase in the DRG 2 hours after C fiber stimuli, whereas such inputs also increase TrkB message in the dorsal horn (unpublished
                                                                                                                                                                                                                                         observations). These changes are very likely to result from an increase in calcium influx through voltage-gated calcium channels in the
                                                                                                                                                                                                                                         DRG, secondary to increased electrical activity. Such changes in intracellular free calcium will cause changes in the phosphorylation and
                                                                                                                                                                                                                                         therefore activation of a whole host of transcription factors, a well characterized example of which in neuronal systems is the cAMP
                                                                                                                                                                                                                                         responsive element-binding protein (CREB) ( Fig. 3 ). Seconds to minutes after calcium influx, neuronal CREB is phosphorylated at
                                                                                                                                                                                                                                         Ser-133 ( 50 ). Activity-dependent CREB Ser-133 phosphorylation has also been demonstrated in mouse embryonic DRG cells in culture
                                                                                                                                                                                                                                         as has activity-dependent regulation of mitogen-activated protein (MAP) kinase ( 51 ). Multiple signal-transduction cascades converge on
                                                                                                                                                                                                                                         CREB Ser-133 phosphorylation (reviewed in ref. 52 ). In hippocampal neurons, phosphorylation of CREB is mediated by the enzyme
                                                                                                                                                                                                                                         CaMKIV (calciumcalmodulin-dependent protein kinase IV). Activation of this enzyme in turn is mediated by CaMK kinase (CamKK).
                                                                                                                                                                                                                                         Interestingly, Ji et al. ( 53 ) have shown that CamKIV immunostaining is present mostly in small neurons of the DRG. Calcium-dependent
                                                                                                                                                                                                                                         BDNF transcription in cortical neurons is mediated through CREB ( 54 , 55 ). It is likely, then, that at least some of the activity-dependent
                                                                                                                                                                                                                                         up-regulation of BDNF within the DRG is also CREB-dependent. Calcium influx into PC12 cells, mediated by P2X2 receptors as opposed
                                                                                                                                                                                                                                         to voltage-gated calcium channels, can activate the MAP kinase cascade via Pyk2 ( 56 ). Pyk2 provides a link between free cellular
                                                                                                                                                                                                                                         calcium and the Map kinase cascade ( 57 ).
                                                                                                                                                                                                                                           FIG. 3. Putative DRG activity- and signal molecule-dependent second-messenger cascades leading to transcription.
                                                                                                                                                                                                                                           Activitydependent calcium flux through voltage-gated calcium channels may increase BDNF transcription via CREB
                                                                                                                                                                                                                                           phosphorylation on Ser-133. NGF acts via TrkA to activate the MAP kinase cascade through ras. Myc phosphorylation through this
                                                                                                                                                                                                                                           pathway can result in E box transactivation via its association with P Max. The preprotachykinin promoter contains multiple E box
                                                                                                                                                                                                                                           units and thus maybe regulated by these transcription factors. RSK2 links the MAP kinase cascade to CREB phosphorylation. ATP
                                                                                                                                                                                                                                           can stimulate the MAP kinase cascade via Pyk2.
                                                                                                                                                                                                                                               BDNF produced in TrkA-expressing small neurons in the DRG is transported to their central terminals, where BDNF is located in
                                                                                                                                                                                                                                         dense core vesicles ( 58 ) and contributes not to basal sensitivity, which appears to be the result primarily of glutamate acting on AMPA
                                                                                                                                                                                                                                         receptors, but to C fiber-induced central sensitization (unpublished observations) ( Fig. 2 ). An increase in the amount of this presynaptic
                                                                                                                                                                                                                                         neuromodulator in C fibers as a result of prior activity in these fibers, combined with an increase in its high-affinity receptor in the dorsal
                                                                                                                                                                                                                                         horn, will result in what we have termed a potentiated system. A potentiated system is one where the same stimulus applied a second time,
                                                                                                                                                                                                                                         some considerable time after the first, will produce a greater response, because the first stimulus has changed the system after a slow onset,
                                                                                                                                                                                                                                         but for a relatively long period ( Fig. 1 ). There are features of this change into a potentiated sensory system that are reminiscent of the
                                                                                                                                                                                                                                         activity-dependent transcriptional changes that contribute to the persistence of long-term potentiation in cortical cells and imply that this
                                                                                                                                                                                                                                         state constitutes a kind of pain memory. The potentiation of the sensory system needs, though, to be differentiated from windup, where a
                                                                                                                                                                                                                                         progressive increase in responsiveness occurs rapidly (within 1 or 2 seconds), during the course of a train of inputs. Potentiation refers to
                                                                                                                                                                                                                                         an effect that only manifests several hours after the input and is due to a change in transcription. Although the system is potentiated, this is
                                                                                                                                                                                                                                         still only to C fiber inputs, which evoke an augmented hypersensitivity because of changes intrinsic only to these fibers and their receptive
                                                                                                                                                                                                                                         dorsal horn neurons. This situation differs from inflammation where A fibers acquire the capacity to induce central hypersensitivity (see
                                                                                                                                                                                                                                         below).
                                                                                                                                                                                                                                               State Four: Inflammatory Changes in DRG and Dorsal Horn Neurons. Inflammation is associated with tissue damage, which
                                                                                                                                                                                                                                         results in the leak of intracellular contents into the extracellular fluid, the recruitment of inflammatory cells, and the production and release
                                                                                                                                                                                                                                         of a broad range of neuroactive agents by inflammatory and noninflammatory cells, including ions (K+ and H+) ( 6 ), amines (5-
                                                                                                                                                                                                                                         hydroxytryptamine, histamine) ( 9 ), kinins (bradykinin) ( 8 ), prostanoids (PGE2) ( 37 ), purines (ATP) ( 7 ), NO, cytokines (IL-1, TNF,
                                                                                                                                                                                                                                         Il-6) ( 9 , 10 ) and growth factors (leukemia inhibitory factor, NGF) ( 10 ). Some of these inflammatory agents may be sufficient by
the authoritative version for attribution.
                                                                                                                                                                                                                                         themselves to activate the peripheral nerve endings of those nociceptors that express the appropriate receptors (Tables 1 and 2 ), generating
                                                                                                                                                                                                                                         inward currents and sensory inflow. Most of these agents, however, act by changing the sensory neuron rather than directly activating it.
                                                                                                                                                                                                                                         These changes include early posttranslational changes both in the peripheral terminals of nociceptors, altering transduction sensitivity
                                                                                                                                                                                                                                         (peripheral sensitization), and in dorsal horn neurons secondary to activity in C fibers (central sensitization). Both peripheral sensitization
                                                                                                                                                                                                                                         and central sensitization alter basal sensitivity to noxious and normally innocuous stimuli. There are, in addition, later and longer-lasting
                                                                                                                                                                                                                                         transcription-dependent changes in the DRG and in the dorsal horn that are due to a complex combination of activity and retrograde
                                                                                                                                                                                                                                         transport of specific signal molecules produced as a result of the inflammation. These changes result both in a potentiated nociceptive
                                                                                                                                                                                                                                         system, as described above, and one in which phenotypic switches alter the central responses elicited by low-threshold A fiber inputs, the
                                                                                                                                                                                                                                         phenomenon of progressive tactile hypersensitivity. Both the potentiation of the system and the phenotypic changes manifest as a change
                                                                                                                                                                                                                                         in stimulus-evoked rather than basal hypersensitivity.
                                                                                                                                                                                                                                               Posttranslational changes. A defining feature of nociceptors is their normal high threshold for activation. After inflamma
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
TRANSCRIPTIONAL AND POSTTRANSLATIONAL PLASTICITY AND THE GENERATION OF INFLAMMATORY PAIN 7728
                                                                                                                                                                                                                                         tion, or even after repeated noxious stimuli, however, the peripheral terminal threshold drops such that lower intensity stimuli can now
                                                                                                                                                                                                                                         initiate activity in the nociceptors. This peripheral sensitization, which can be detected within a very short period, is the result of changes
                                                                                                                                                                                                                                         either in the transducing receptor molecules themselves or in sodium channels in the terminal ( Fig. 4 ). A change in the transducer
                                                                                                                                                                                                                                         molecule is best exemplified by VR1, where repeated heat stimuli by themselves result in a progressively augmenting inward current
                                                                                                                                                                                                                                         through the ion channel, and exposure to protons does the same thing, by mechanisms currently not known ( 4 , 5 ). The other contributor
                                                                                                                                                                                                                                         to peripheral sensitization is phosphorylation of membrane-bound receptors/ion channels. Many inflammatory mediators activate either
                                                                                                                                                                                                                                         protein kinase A or C (see Fig. 4 ), both of which can phosphorylate the TTXr sodium channel SNS/PN3 and contribute to a greater
                                                                                                                                                                                                                                         sodium current in the terminal ( 59  61 ). The recently discovered TTXr channel SNS2 has different protein kinase C and protein kinase
                                                                                                                                                                                                                                         A consensus regions on its intracellular loops ( 20 ), and its contribution to the change in sodium current is undetermined. These
                                                                                                                                                                                                                                         sensitizing changes occur locally in the peripheral terminal, independent of transcription in the DRG. However, transcription of the very
                                                                                                                                                                                                                                         elements that show posttranslational changes in these terminals is highly regulated in the cell soma. Inflammation up-regulates both VR1
                                                                                                                                                                                                                                         and SNS/SNS2 ( 5 , 20 , 62 ). Inflammation is associated with an increase in peripheral NGF levels ( 63 ), and this may be the key signal
                                                                                                                                                                                                                                         molecule for many of the transcriptional changes (see below). Although peripheral sensitization does not itself require transcription, an
                                                                                                                                                                                                                                         increase in the substrate for such sensitization is very likely to amplify the phenomenon. Because of the delay inherent in the initiation of
                                                                                                                                                                                                                                         changes in expression and transport of proteins,
                                                                                                                                                                                                                                           FIG. 4. Posttranslational changes in transduction mechanisms/ion channels at nociceptor peripheral terminals induced by
                                                                                                                                                                                                                                           inflammatory mediators increases sensitivity and reduces threshold (peripheral sensitization), which occurs as a result of changes in
                                                                                                                                                                                                                                           the transducer proteins themselves (e.g., VR1) and as a result of a PKC- and PKA-mediated phosphorylation of TTXr sodium
                                                                                                                                                                                                                                           channels. Both activity and retrograde transport of signal molecules can also induce transcriptional changes in the DRG, which
                                                                                                                                                                                                                                           increase transducer molecules (VR1), ion channels (SNS/SNS2), and synaptic neuromodulators (BDNF/substance P) both altering
                                                                                                                                                                                                                                           phenotype and potentiating the system.
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
TRANSCRIPTIONAL AND POSTTRANSLATIONAL PLASTICITY AND THE GENERATION OF INFLAMMATORY PAIN 7729
                                                                                                                                                                                                                                         such transcription-dependent augmentation will only manifest many hours after the onset of inflammation ( Fig. 4 ).
                                                                                                                                                                                                                                               Apart from the increased sensitivity of the peripheral terminals of nociceptors on exposure to inflammatory mediators, which will
                                                                                                                                                                                                                                         result in an area of increased sensitivity to thermal and mechanical stimuli localized to the site of inflammation, any C fiber input activated
                                                                                                                                                                                                                                         at the time of or during inflammation will also initiate central sensitization. This C fiber input will result in an NMDA receptor-sensitive
                                                                                                                                                                                                                                         increase in responsiveness to low- and high-intensity stimuli, both when applied to the site of the inflammation and in the contiguous
                                                                                                                                                                                                                                         noninflamed area. Tactile allodynia and pin prick hyperalgesia in the zone of secondary hyperalgesia ( 64 ), for example, are characteristic
                                                                                                                                                                                                                                         NMDA-receptor-mediated ( 65 ) features of central sensitization.
                                                                                                                                                                                                                                               Transcriptional changes. In addition to these posttranslational changes, an alteration in the expression of effector molecules in the
                                                                                                                                                                                                                                         DRG ( 66 ) and dorsal horn ( 67  69 ) is a prominent feature of inflammation and can be initiated in two ways. The first is as a result of an
                                                                                                                                                                                                                                         activity-dependent activation of the CREB transcription factor both in DRG and dorsal horn neurons (ref. 70 ; Fig. 3 ). As discussed
                                                                                                                                                                                                                                         above, this will result, after a delay of several hours for transcription and translation in the DRG and protein transport to central terminals,
                                                                                                                                                                                                                                         in a potentiated system in which the C fibers are primed to exert a greater effect on dorsal horn neurons as a result of an increased
                                                                                                                                                                                                                                         expression of neuromodulators like BDNF. In addition, the dorsal horn is simultaneously made hyperresponsive to such neuromodulators
                                                                                                                                                                                                                                         as a result of an activity-dependent increased expression of the TrkB receptor. The second way that transcriptional changes occur after
                                                                                                                                                                                                                                         inflammation is via the production in the inflamed tissue of specific signal molecules that bind to receptors on nociceptor sensory
                                                                                                                                                                                                                                         terminals. The ligandreceptor complex is then internalized and retrogradely transported to the cell body, where it activates specific signal-
                                                                                                                                                                                                                                         transduction cascades ( Fig. 3 ). NGF is the prototypical example of such a signal molecule whose level increases substantially in inflamed
                                                                                                                                                                                                                                         tissue and neutralization of which massively reduces inflammatory hypersensitivity (reviewed in ref. 71 ).
                                                                                                                                                                                                                                               Signals from extracellular growth factors such as NGF are transduced into intracellular responses through the small G protein Ras and
                                                                                                                                                                                                                                         MAP kinase (reviewed in refs. 72  74 ). The Ras cascade (see Fig. 3 ) involves the sequential activation of Ras, Raf, MAP kinase kinase,
                                                                                                                                                                                                                                         and MAP kinase itself [p44/p42 MAP kinase is also known as ERK 1/2 (extracellular signal regulated kinase)]. Subsequent to
                                                                                                                                                                                                                                         phosphorylation, a fragment of activated MAP kinase translocates to the nucleus, where it then regulates gene expression through the
                                                                                                                                                                                                                                         phosphorylation, and therefore activation, of various transcription factors including c-Myc, Elk-1, c-Fos, and c-Jun ( 72 , 73 ).
                                                                                                                                                                                                                                         Phosphorylation of these transcription factors results in their association into complexes. These complexes activate transcription of many
                                                                                                                                                                                                                                         downstream genes via their association with response elements present in the response gene promoter regions ( Fig.3 ). The MAP kinase
                                                                                                                                                                                                                                         cascade can also phosphorylate and thus activate CREB within neurons via the activation of Rsk2 by MAP kinase itself ( 75 , 76 ).
                                                                                                                                                                                                                                               One consequence of transcriptional changes in DRG neurons after inflammation is that some low-threshold A neurons acquire the
                                                                                                                                                                                                                                         chemical phenotype typical of C fibers. For example, the neuropeptide substance P is normally found almost exclusively in a subset of the
                                                                                                                                                                                                                                         TrkA-expressing C fibers, with only a very small number in TrkA expressing small myelinated A fibers ( 77 , 78 ). After inflammation,
                                                                                                                                                                                                                                         there is a NGF-dependent increase in substance P expression in C fibers ( 63 ), but also a novel expression of this neuropeptide in some
                                                                                                                                                                                                                                         large A fibers as well ( 3 ). This new expression, together with the inflammation-induced increase in NK1 receptors in the dorsal horn (
                                                                                                                                                                                                                                         79 ), will result not only in a potentiated system, but one in which the specific type of stimulus that can evoke central sensitization has
                                                                                                                                                                                                                                         changed. Stimulus-induced hypersensitivity can thus be mediated by low-intensity A inputs as well as high-intensity C fiber inputs (
                                                                                                                                                                                                                                         Fig. 1 ). This heightened sensibility manifests as progressive tactile hypersensitivity, where low-intensity mechanical stimulation of
                                                                                                                                                                                                                                         inflamed skin (light touch) produces a progressively incrementing increase in the excitability of spinal neurons, something they never can
                                                                                                                                                                                                                                         elicit in the normal situation, and this can be mimicked by A fiber stimulation ( 3 , 80  83 ).
                                                                                                                                                                                                                                                                                                        CONCLUSION
                                                                                                                                                                                                                                               A clear distinction needs to be made between nociception, the detection only of intense noxious stimuli, and inflammatory pain,
                                                                                                                                                                                                                                         which is evoked by normally innocuous stimuli and incorporates an exaggerated response to noxious stimuli. The transition from one state
                                                                                                                                                                                                                                         to the other involves multiple changes, some early and others late, some mediated by posttranslational changes and others reliant on
                                                                                                                                                                                                                                         altered gene expression, some evoked by activity and others in response to specific inflammatory mediators/signal molecules. Enormous
                                                                                                                                                                                                                                         progress has been made in unraveling what changes occur, when they occur, and the molecular mechanisms involved. Although much still
                                                                                                                                                                                                                                         remains to be determined, this new insight is translating into novel and more targeted approaches to treating inflammatory pain.
                                                                                                                                                                                                                                               The reviewed work was supported by the Medical Research Council G9431792, the Human Frontier Science Program RG73/96, the
                                                                                                                                                                                                                                         Wellcome Trust 039614, the European Union BMH4-CT 95 0172, and the National Institute of Neurological Disorders and Stroke R01
                                                                                                                                                                                                                                         NS3825301.
                                                                                                                                                                                                                                         1. Bessou, P. & Perl, E. R. ( 1969 ) J. Neurophysiol. 32 , 10251043 .
                                                                                                                                                                                                                                         2. Woolf, C. J. ( 1983 ) Nature (London) 306 , 686688 .
                                                                                                                                                                                                                                         3. Neumann, S. , Doubell, T. P. , Leslie, T. A. & Woolf, C. J. ( 1996 ) Nature (London) 384 , 360364 .
                                                                                                                                                                                                                                         4. Caterina, M. J. , Schumacher, M. A. , Tominaga, M. , Rosen, T. A. , Levine, J. D. & Julius, D. ( 1997 ) Nature (London) 389 , 816824 .
                                                                                                                                                                                                                                         5. Tominaga, M. , Caterina, M. J. , Malmberg, A. B. , Rosen, T. A. , Gilbert, H. , Skinner, K. , Raumann, B. E. , Basbaum, A. I. & Julius, D. ( 1998 )
                                                                                                                                                                                                                                              Neuron 21 , 531543 .
                                                                                                                                                                                                                                         6. Waldmann, R. & Lazdunski, M. ( 1998 ) Curr. Opin. Neurobiol. 8 , 418424 .
                                                                                                                                                                                                                                         7. Burnstock, G. & Wood, J. N. ( 1996 ) Curr. Opin. Neurobiol. 6 , 526532 .
                                                                                                                                                                                                                                         8. Walker, K. , Perkins, M. & Dray, A. ( 1995 ) Neurochem. Int. 26 , 117 .
                                                                                                                                                                                                                                         9. Dray, A. ( 1995 ) Br. J. Anaesth. 75 , 125131 .
                                                                                                                                                                                                                                         10. Levine, J. D. & Taiwo, Y. O. ( 1994 ) in Textbook of Pain , eds. Wall, P. D. & Melzack, R. ( Churchill Livingstone , New York ), pp. 4546 .
                                                                                                                                                                                                                                         11. Bradbury, E. J. , Burnstock, G. & McMahon, S. B. ( 1998 ) Mol. Brain Res. 12 , 256268 .
                                                                                                                                                                                                                                         12. Catterall, W. A. ( 1992 ) Physiol. Rev. 72 , S1548 .
                                                                                                                                                                                                                                         13. Caffrey, J. M. , Eng, D. L. , Black, J. A. , Waxman, S. G. & Kocsis, J. D. ( 1992 ) Brain Res. 592 , 283297 .
                                                                                                                                                                                                                                         14. Roy, M. L. & Narahashi, T. ( 1992 ) J. Neurosci. 12 , 21042111 .
                                                                                                                                                                                                                                         15. Waxman, S. G. , Kocsis, J. D. & Black, J. A. ( 1994 ) J. Neurophysiol. 72 , 466470 .
                                                                                                                                                                                                                                         16. Black, J. A. & Waxman, S. G. ( 1996 ) Dev. Neurosci. 18 , 139152 .
                                                                                                                                                                                                                                         17. Black, J. A. , Dib-Hajj, S. , McNabola, K. , Jeste, S. , Rizzo, M. A. , Kocsis, J. D. & Waxman, S. G. ( 1996 ) Brain Res. Mol. Brain Res. 43 , 117
                                                                                                                                                                                                                                              131 .
                                                                                                                                                                                                                                         18. Akopian, A. N. , Sivilotti, L. & Wood, J. N. ( 1996 ) Nature (London) 379 , 257262 .
the authoritative version for attribution.
                                                                                                                                                                                                                                         19. Sangameswaran, L. , Delgado, S. G. , Fish, L. M. , Koch, B. D. , Jakeman, L. B. , Stewart, G. R. , Sze, P. , Hunter, J. C. , Eglen, R. M. & Herman, R.
                                                                                                                                                                                                                                              C. ( 1996 ) J. Biol. Chem. 271 , 59535956 .
                                                                                                                                                                                                                                         20. Tate, S. , Benn, S. , Hick, C. , Trezise, D. , John, V. , Mannion, R. J. , Costigan, M. , Plumpton, C. , Grose, D. , Gladwell, Z. , Kendall, G. , Dale, K. ,
                                                                                                                                                                                                                                              Bountra, C. & Woolf, C. J. ( 1998 ) Nat. Neurosci. 1 , 653655 .
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
TRANSCRIPTIONAL AND POSTTRANSLATIONAL PLASTICITY AND THE GENERATION OF INFLAMMATORY PAIN 7730
                                                                                                                                                                                                                                         21. Dib-Hajj, S. , Tyrrell, L. , Black, J. A. & Waxman, S. G. ( 1998 ) Proc. Natl. Acad. Sci. USA 95 , 89638968 .
                                                                                                                                                                                                                                         22. Willis, W. D. & Coggeshall, R. E. ( 1991 ) Sensory Mechanisms of the Spinal Cord ( Plenum , New York ).
                                                                                                                                                                                                                                         23. Yoshimura, M. & Jessell, T. M. ( 1990 ) J. Physiol. (London) 430 , 315335 .
                                                                                                                                                                                                                                         24. King, A. E. , Thompson, S. W. N. , Urban, L. & Woolf, C. J. ( 1988 ) Neurosci. Lett. 89 , 286292 .
                                                                                                                                                                                                                                         25. Cao, Y. Q. , Mantyh, P. W. , Carlson, E. J. , Gillespie, A. M. , Epstein, C. J. & Bausbaum, A. I. ( 1998 ) Nature (London) 392 , 390394 .
                                                                                                                                                                                                                                         26. Woolf, C. J , Mannion, R. J. & Neumann, S. ( 1998 ) Neuron 20 , 10631066 .
                                                                                                                                                                                                                                         27. Woolf, C. J. & Thompson, S. W. N. ( 1991 ) Pain 44 , 293299 .
                                                                                                                                                                                                                                         28. Ma, Q.-P. & Woolf, C. J. ( 1995 ) J. Physiol. (London) 486.3 , 769777 .
                                                                                                                                                                                                                                         29. Dubner, R. ( 1991 ) in Neuronal Plasticity and Pain Following Peripheral Tissue Inflammation or Nerve Injury , eds. Bond, M. R. , Charlton, J. E. &
                                                                                                                                                                                                                                              Woolf, C. J. ( Elsevier , Amsterdam ), pp. 263276 .
                                                                                                                                                                                                                                         30. McMahon, S. B. , Lewin, G. R. & Wall, P. D. ( 1993 ) Curr. Opin. Neurobiol. 3 , 602610 .
                                                                                                                                                                                                                                         31. Coderre, T. J. & Melzack, R. ( 1992 ) J. Neurosci. 12 , 36653670 .
                                                                                                                                                                                                                                         32. Ren, K. & Dubner, R. ( 1993 ) Neurosci. Lett. 163 , 2226 .
                                                                                                                                                                                                                                         33. Woolf, C. J. , Shortland, P. & Sivilotti, L. G. ( 1994 ) Pain 58 , 141155 .
                                                                                                                                                                                                                                         34. Koltzenburg, M. , Torebjork, H. E. & Wahren, L. K. ( 1994 ) Brain 117 , 579591 .
                                                                                                                                                                                                                                         35. Kilo, S. , Schmelz, M. , Koltzenburg, M. & Handwerker, H. O. ( 1994 ) Brain 117 , 385396 .
                                                                                                                                                                                                                                         36. Woolf, C. J. & Wall, P. D. ( 1986 ) J. Neurosci. 6 , 14331443 .
                                                                                                                                                                                                                                         37. Bley, K. R. , Hunter, J. C. , Eglen, R. M. & Smith, J. A. ( 1998 ) Trends Pharmacol. Sci. 19 , 141147 .
                                                                                                                                                                                                                                         38. Heath, M. J. , Womack, M. D. & MacDermott, A. B. ( 1994 ) J. Neurophysiol. 72 , 11921198 .
                                                                                                                                                                                                                                         39. Zirrgiebel, U. , Ohga, Y. , Carter, B. , Berninger, B. , Inagaki, N. , Thoenen, H. & Lindholm, D. ( 1995 ) J. Neurochem. 65 , 2241 2250 .
                                                                                                                                                                                                                                         40. Iwasaki, Y. , Gay, B. , Wada, K. & Koizumi, S. ( 1998 ) J. Neurochem. 71 , 106111 .
                                                                                                                                                                                                                                         41. Suen, P.-C , Wu, K. , Xu, J.-L. , Lin, S. Y. , Levine, E. S. & Black, I. B. ( 1998 ) Brain Res. Mol. Brain Res. 59 , 215228 .
                                                                                                                                                                                                                                         42. Yu, X. M. , Askalan, R. , Keil, G. J.2. & Salter, M. W. ( 1997 ) Science 275 , 674678 .
                                                                                                                                                                                                                                         43. Lin, S. Y. , Wu, K. , Levine, E. S. , Mount, H. T. , Suen, P.-C. & Black, I. B. ( 1998 ) Brain Res. Mol. Brain Res. 55 , 2027 .
                                                                                                                                                                                                                                         44. Wang, T. & Salter, M. W. ( 1994 ) Nature (London) 369 , 233235 .
                                                                                                                                                                                                                                         45. Chen, L. & Huang, L.-Y. M. ( 1992 ) Nature (London) 356 , 521523 .
                                                                                                                                                                                                                                         46. Woolf, C. J. & King, A. E. ( 1990 ) J. Neurosci. 10 , 27172726 .
                                                                                                                                                                                                                                         47. Cook, A. J. , Woolf, C. J. , Wall, P. D. & McMahon, S. B. ( 1987 ) Nature (London) 325 , 151153 .
                                                                                                                                                                                                                                         48. Simone, D. A. , Baumann, T. K. , Collins, J. G. & LaMotte, R. H. ( 1989 ) Brain Res. 486 , 185189 .
                                                                                                                                                                                                                                         49. Torebjork, H. E. , Lundberg, L. E. R. & LaMotte, R. H. ( 1992 ) J. Physiol. (London) 448 , 765780 .
                                                                                                                                                                                                                                         50. Ginty, D. D. , Kornhauser, J. M. , Thompson, M. A. , Bading, H. , Mayo, K. E. , Takahashi, J. S. & Greenberg, M. E. ( 1993 ) Science 260 , 238241 .
                                                                                                                                                                                                                                         51. Fields, R. D. , Eshete, F. , Stevens, B. & Itoh, K. ( 1997 ) J. Neurosci. 17 , 72527266 .
                                                                                                                                                                                                                                         52. Bito, H. , Deisseroth, K. & Tsien, R. W. ( 1997 ) Curr. Opin. Neurobiol. 7 , 419429 .
                                                                                                                                                                                                                                         53. Ji, R. R. , Shi, T.-J. , Xu, Z.-Q. , Zhang, Q. , Sakagami, H. , Tsubochi, H. , Kondo, H. & Hokfelt, T. ( 1996 ) Brain Res. 721 , 167173 .
                                                                                                                                                                                                                                         54. Tao, X. , Finkbeiner, S. , Arnold, D. B. , Shaywitz, A. J. & Greenberg, M. E. ( 1998 ) Neuron 20 , 709726 .
                                                                                                                                                                                                                                         55. Shieh, P. B. , Hu, S. C. , Bobb, K. , Timmusk, T. & Ghosh, A. ( 1998 ) Neuron 20 , 727740 .
                                                                                                                                                                                                                                         56. Swanson, K. D. , Reigh, C. & Landreth, G. E. ( 1998 ) J. Biol. Chem. 273 , 1996519971 .
                                                                                                                                                                                                                                         57. Lev, S. , Moreno, H. , Martinez, R. , Canoll, P. , Peles, E. , Musacchio, J. M. , Plowman, G. D. , Rudy, B. & Schlessinger, J. ( 1995 ) Nature
                                                                                                                                                                                                                                              (London) 376 , 737745 .
                                                                                                                                                                                                                                         58. Michael, G. J. , Averill, S. , Nitkunan, A. , Rattray, M. , Bennett, D. L. H. , Yan, Q. & Priestley, J. V. ( 1997 ) J. Neurosci. 17 , 84768490 .
                                                                                                                                                                                                                                         59. Gold, M. S. , Levine, J. D. & Correa, A. M. ( 1998 ) J. Neurosci. 18 , 1034510355 .
                                                                                                                                                                                                                                         60. England, S. , Bevan, S. & Docherty, R. J. ( 1996 ) J. Physiol. (London) 495 , 429440 .
                                                                                                                                                                                                                                         61. Gold, M. S. , Reichling, D. B. , Schuster, M. J. & Levine, J. D. ( 1996 ) Proc. Natl. Acad. Sci. USA 93 , 11081112 .
                                                                                                                                                                                                                                         62. Okuse, K. , Chaplan, S. R. , McMahon, S. B. , Luo, Z. D. , Calcutt, N. A. , Scott, B. P. , Akopian, A. N. & Wood, J. N. ( 1997 ) Mol. Cell. Biol. 10 ,
                                                                                                                                                                                                                                              196207 .
                                                                                                                                                                                                                                         63. Woolf, C. J. , Safieh-Garabedian, B. , Ma, Q.-P. , Crilly, P. & Winter, J. ( 1994 ) Neuroscience 62 , 327331 .
                                                                                                                                                                                                                                         64. Koltzenburg, M. , Lundberg, L. E. R. & Torebjork, H. E. ( 1992 ) Pain 51 , 207220 .
                                                                                                                                                                                                                                         65. Stubhaug, A. , Breivik, H. , Eide, P. K. , Kreunen, M. & Foss, A. ( 1997 ) Acta Anaesthesiol. Scand. 41 , 11241132 .
                                                                                                                                                                                                                                         66. Leslie, T. A. , Emson, P. C. , Dowd, P. M. & Woolf, C. J. ( 1995 ) Neuroscience 67 , 753761 .
                                                                                                                                                                                                                                         67. Noguchi, K. , Dubner, R. & Ruda, M. A. ( 1992 ) Neuroscience 46 , 561570 .
                                                                                                                                                                                                                                         68. Noguchi, K. , Kowalski, K. , Traub, R. , Solodkin, A. , Iadarola, M. J. & Ruda, M. A. ( 1991 ) Mol. Brain Res. 10 , 227233 .
                                                                                                                                                                                                                                         69. Ruda, M. A. , Iadarola, M. J. , Cohen, L. V. & Young, W. S., III. ( 1988 ) Proc. Natl. Acad. Sci. USA 85 , 622626 .
                                                                                                                                                                                                                                         70. Ji, R. R. & Rupp, F. ( 1997 ) J. Neurosci. 17 , 17761785 .
                                                                                                                                                                                                                                         71. Woolf, C. J. ( 1996 ) Philos. Trans. R. Soc. London B 351 , 441448 .
                                                                                                                                                                                                                                         72. Davis, R. J. ( 1993 ) J. Biol. Chem. 268 , 1455314556 .
                                                                                                                                                                                                                                         73. Seger, R. & Krebs, E. G. ( 1995 ) FASEB J. 9 , 726735 .
                                                                                                                                                                                                                                         74. Fukunaga, K. & Miyamoto, E. ( 1998 ) Mol. Neurobiol. 16 , 7995 .
                                                                                                                                                                                                                                         75. Xing, J. , Ginty, D. D. & Greenberg, M. E. ( 1996 ) Science 273 , 959963 .
                                                                                                                                                                                                                                         76. Impey, S. , Obrietan, K. , Wong, S. T. , Poser, S. , Yano, S. , Wayman, G. , Deloulme, J. C. , Chan, G. & Storm, D. R. ( 1998 ) Neuron 21 , 869883 .
                                                                                                                                                                                                                                         77. Averill, S. & McMahon, S. B. ( 1998 ) Eur. J. Neurosci. 7 , 1484 1494 .
                                                                                                                                                                                                                                         78. McMahon, S. B. , Armanini, M. P. , Ling, L. H. & Phillips, H. S. ( 1994 ) Neuron 12 , 11611171 .
                                                                                                                                                                                                                                         79. Krause, J. E. , DiMaggio, D. A. & McCarson, K. E. ( 1995 ) Can. J. Physiol. Pharmacol. 73 , 854859 .
                                                                                                                                                                                                                                         80. Ma, Q.-P. & Woolf, C. J. ( 1996 ) Pain 67 , 97106 .
                                                                                                                                                                                                                                         81. Ma, Q.-P. & Woolf, C. J. ( 1997 ) NeuroReport 8 , 807810 .
                                                                                                                                                                                                                                         82. Ma, Q.-P. , Allchorne, A. J. & Woolf, C. J. ( 1998 ) Pain 77 , 4957 .
                                                                                                                                                                                                                                         83. Ma, Q.-P. & Woolf, C. J. ( 1997 ) Eur. J. Pharmacol. 322 , 165171 .
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
CELLULAR MECHANISMS OF NEUROPATHIC PAIN, MORPHINE TOLERANCE, AND THEIR INTERACTIONS 7731
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
CELLULAR MECHANISMS OF NEUROPATHIC PAIN, MORPHINE TOLERANCE, AND THEIR INTERACTIONS 7732
                                                                                                                                                                                                                                         nociceptive neurons. These experiments are complemented by studies that demonstrate elevated dorsal horn levels of translocated PKC or
                                                                                                                                                                                                                                         increased biosynthesis of PKC in neuropathic rats ( 20 , 21 ).
                                                                                                                                                                                                                                                Contributions of Central Excitation to Hyperalgesia. It has been proposed that central sensitization may be manifested as
                                                                                                                                                                                                                                         increases in spontaneous and stimulus-evoked neuronal activity within the spinal cord, which, in turn, contribute to the development and
                                                                                                                                                                                                                                         maintenance of neurogenic and inflammatory pain syndromes. There may be an increase in spontaneous neural activity of pain
                                                                                                                                                                                                                                         transmission pathways and hence spontaneous pain. This increased spontaneous action potential activity includes that of spinal cord dorsal
                                                                                                                                                                                                                                         horn neurons ( 22 , 23 ), and that of pain-related thalamic neurons ( 24 ) of CCI rats with neuropathic hyperalgesia ( 25 ). These extensive
                                                                                                                                                                                                                                         elevations in neural activity, which have also been mapped by using the 2-deoxyglucose metabolic technique ( 26 , 27 ), occur within the
                                                                                                                                                                                                                                         spinal cord and a variety of pain-related brain regions even in the absence of overt somatic stimulation. Furthermore, responses of central
                                                                                                                                                                                                                                         pain-related neurons of CCI rats show exaggerated responses to innocuous and noxious peripheral stimulation. This is indicated by
                                                                                                                                                                                                                                         increased responses of spinothalamic neurons to mechanical or thermal stimulation ( 23 ). An exaggerated response to innocuous
                                                                                                                                                                                                                                         stimulation may contribute to allodynia, whereas the enhanced response to noxious stimulation is likely related to hyperalgesia. Peripheral
                                                                                                                                                                                                                                         receptive fields also expand after tissue inflammation ( 28 ). Given the role of receptive field size in neuronal recruitment, expanded
                                                                                                                                                                                                                                         receptive fields would mean that a nociceptive stimulus would activate more central neurons than would normally occur, leading to
                                                                                                                                                                                                                                         exaggerated pain and exaggerated spatial radiation of the painful sensation. Taken together, elevated spontaneous discharges, increased
                                                                                                                                                                                                                                         stimulus-evoked impulse frequencies, and expanded receptive fields are likely to operate in concert to cause persistent hyperalgesia,
                                                                                                                                                                                                                                         spontaneous pain, allodynia, and radiation of pain.
                                                                                                                                                                                                                                                Contributions of Central Disinhibition to Hyperalgesia. Excitatory amino acid-induced PKC translocation/activation and NO
                                                                                                                                                                                                                                         production may also result in disinhibitory processes associated with excitotoxic consequences including neuronal death within the CNS.
                                                                                                                                                                                                                                         The involvement of such excitotoxic processes in mechanisms of injury-induced pain syndromes has been suggested by several
                                                                                                                                                                                                                                         experimental observations. There is histological evidence showing that peripheral nerve injury induces excitotoxic transsynaptic
                                                                                                                                                                                                                                         morphological changes of superficial dorsal horn (laminae III) neurons (dark neurons), which have been proposed to be inhibitory
                                                                                                                                                                                                                                         interneurons ( 29 ). Importantly, a PARS has been shown to be activated in CCI rats, which is likely to contribute to the development of
                                                                                                                                                                                                                                         dark neurons ( 30 ). The activation of PARS occurs in the presence of DNA fragmentation, a process that may be initiated by the
                                                                                                                                                                                                                                         production of NO and that may eventually lead to programmed cell death. These morphological changes may reflect a pathological
                                                                                                                                                                                                                                         process in which injury-induced central responses result in a persistent imbalance of the excitatoryinhibitory circuitry within the spinal
                                                                                                                                                                                                                                         cord dorsal horn. This possibility is supported by the demonstration of A-mediated mechanical allodynia in neuropathic pain patients (
                                                                                                                                                                                                                                         31 ) and the demonstration that pharmacological blockade of the spinal cord -aminobutyric acid and glycine inhibitory system augments
                                                                                                                                                                                                                                         thermal hyperalgesia in rats with sciatic nerve injury ( 32 ). Thus, in addition to direct central sensitzation described above, the possible
                                                                                                                                                                                                                                         disinhibition resulting from the loss of function of spinal cord inhibitory interneurons may also contribute to central hyperexcitability after
                                                                                                                                                                                                                                         peripheral nerve injury and inflammation.
                                                                                                                                                                                                                                                Central Mechanisms Subserving Morphine Tolerance. Similar mechanisms, including the activation of NMDA receptors,
                                                                                                                                                                                                                                         translocation and activation of PKC, and the production of NO have been implicated in the development and maintenance of morphine
                                                                                                                                                                                                                                         tolerance. Detailed discussion of these mechanisms of morphine tolerance is presented elsewhere ( 9 ). The following sections will
                                                                                                                                                                                                                                         emphasize new evidence indicating the involvement of PARS in mechanisms of morphine tolerance.
                                                                                                                                                                                                                                                As discussed above for the development of hyperalgesia, a consequence of excessive NMDA receptor activation is the initiation of a
                                                                                                                                                                                                                                         cascade of intracellular events leading to PARS activation ( 33 ). In view of the evidence implicating NMDA receptor activation and the
                                                                                                                                                                                                                                         subsequent intracellular events resulting from this, we have conducted a series of experiments to examine the possible role of PARS in the
                                                                                                                                                                                                                                         development of morphine tolerance. In addition, because a consequence of PARS activation may include alterations in cell morphology,
                                                                                                                                                                                                                                         we examined the development of dark neurons as a marker of altered cellular morphology.
                                                                                                                                                                                                                                                For these experiments, adult male SpragueDawley rats were prepared for intrathecal (i.t.) injection by implanting a polyethylene
                                                                                                                                                                                                                                         (PE-10) tube into the lumbar spinal cord. The vehicle or drugs were delivered slowly (13 minutes) through the i.t. catheter followed by 10
                                                                                                                                                                                                                                         l of saline, which flushed the catheter. To examine the antinociceptive effects of morphine, the tail-flick test was used. The light bulb
                                                                                                                                                                                                                                         intensity of the tail-flick device was set to produce baseline tail-flick latencies between 3.5 and 4.5 sec. To ensure that no tissue damage
                                                                                                                                                                                                                                         occurred, the light bulb was automatically turned off after 8 sec, even if no tail-flick occurred. An average of three tail-flick trials each
                                                                                                                                                                                                                                         separated by a 1-min intertrial interval constituted the mean baseline latency (BL). The antinociceptive effects of morphine and the
                                                                                                                                                                                                                                         development of tolerance were then determined by measuring test (tail-flick) latencies (TL) after drug administration. Thus, data were
                                                                                                                                                                                                                                         expressed as percent maximal possible antinociceptive effect (%MPAE) using the equation %MPAE = [(TL  BL)/(8  BL)]  100.
                                                                                                                                                                                                                                                The procedure for the histological examination of dark neurons was similar to that described previously ( 29 , 30 ). It has been
                                                                                                                                                                                                                                         reported by Sugimoto et al. ( 29 ) that there are three principal characteristics of dark neurons: (i) irregular cellular outlines; (ii) increased
                                                                                                                                                                                                                                         amounts of chromatin throughout the nucleoplasm and cytoplasm, which is why they are called dark neurons; and (iii) intensely and
                                                                                                                                                                                                                                         homogeneously stained nucleoplasm with almost indiscernible heterochromatin. Dorsal horn neurons that are normal sometimes show
                                                                                                                                                                                                                                         increased cytoplasmic staining also, but they do not exhibit enhanced nucleoplasmic staining. Some glial cells, especially
                                                                                                                                                                                                                                         oligodendrocytes, also exhibit chromophilia that is as dark as that seen in dark neurons. Although this is true, oligodendrocytes are
                                                                                                                                                                                                                                         distinguishable from dark neurons, because glial cells have aggregates of heterochromatin that are seen under high-power magnification,
                                                                                                                                                                                                                                         whereas dark neurons do not exhibit this characteristic. Neurons that exhibited all three of the above-mentioned characteristics were
                                                                                                                                                                                                                                         counted as dark neurons, whereas other cells were excluded.
                                                                                                                                                                                                                                                The spinal cord sections were divided into three zones for examination by microscope. They were divided, by using Rexeds laminar
                                                                                                                                                                                                                                         system, into laminae III, IIIIV, and VVI. Sections were examined with the experimenter blind to the treatment regimen. Dark neurons
                                                                                                                                                                                                                                         were counted in each zone mentioned above under medium-power magnification. If there was a dark neuron that was in question, it was
                                                                                                                                                                                                                                         then viewed under high-power magnification to discern if it was indeed a dark neuron. At least three 1-m spinal cord sections were
                                                                                                                                                                                                                                         viewed for each rat. This yielded an average number of dark neurons for each subdivision.
                                                                                                                                                                                                                                                The tail-flick data were analyzed by using two-way ANOVA to discern differences among treatment groups. When main effects were
                                                                                                                                                                                                                                         seen, a WallerDuncan D ratio t test was performed to determine the source of variations between the groups. Dark neurons were counted
                                                                                                                                                                                                                                         for the left and right sides of the dorsal horn in laminae IVI. The numbers were then averaged and analyzed by using a two-way
the authoritative version for attribution.
                                                                                                                                                                                                                                         ANOVA. The total number of dark neurons from a sampled region was analyzed to determine (i) differences in the number of dark
                                                                                                                                                                                                                                         neurons between the left and right side of the dorsal horn; (ii) differences in numbers of dark neurons among sampled dorsal horn regions
                                                                                                                                                                                                                                         (i.e., laminae III);
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
CELLULAR MECHANISMS OF NEUROPATHIC PAIN, MORPHINE TOLERANCE, AND THEIR INTERACTIONS 7733
                                                                                                                                                                                                                                         and (iii) differences in the number of dark neurons among treatment groups.
                                                                                                                                                                                                                                               An initial experiment was performed to determine the effect of the chronic administration of morphine on the development of dark
                                                                                                                                                                                                                                         neurons. Three groups of rats (n = 59 per group) were used: (i) rats treated with saline i.t. once a day; (ii) rats treated with 10 g of
                                                                                                                                                                                                                                         morphine i.t. once daily; and (iii) rats treated with 20 g of morphine i.t. once daily. The doses of morphine given have previously been
                                                                                                                                                                                                                                         shown to induce the development of tolerance to the antinociceptive effects of morphine ( 34 ). All of the agents were given once daily for
                                                                                                                                                                                                                                         8 days.
                                                                                                                                                                                                                                               On day 8, those rats receiving morphine and saline showed the development of tolerance to the analgesic effects of morphine. Rats
                                                                                                                                                                                                                                         that were made tolerant to morphine exhibited a reliable increase in the number of dark neurons in the dorsal horn of the lumbar spinal
                                                                                                                                                                                                                                         cord (P < 0.01). Several features characterized this increase in dark neurons. Dark neurons were primarily located in laminae III and to a
                                                                                                                                                                                                                                         much lesser degree to laminae IIIIV. Also, there was no statistical difference in the number of dark neurons observed on the left and right
                                                                                                                                                                                                                                         sides of the spinal cord (P > 0.05). Because chronic administration of morphine induced tolerance and the development of dark neurons,
                                                                                                                                                                                                                                         we examined the effect of the selective PARS inhibitor benzamide, which has been shown to prevent the development of hyperalgesia in
                                                                                                                                                                                                                                         the CCI model ( 30 ), on the development of morphine tolerance and dark neurons resulting from chronic morphine administration. For
                                                                                                                                                                                                                                         this experiment, seven groups of rats were used. They included rats receiving 100, 200, or 400 nmol benzamide and 20 g of morphine on
                                                                                                                                                                                                                                         days 18, rats receiving 400 nmol benzamide and saline on days 18, rats receiving 20 g morphine and saline on days 18, rats receiving
                                                                                                                                                                                                                                         only saline on days 18, and rats receiving saline on days 17 and 20 g of morphine on day 8.
                                                                                                                                                                                                                                               As shown in Fig. 1 , coadministration of 20 g of morphine with 200 or 400 nmol (not 100 nmol) benzamide for 7 days reliably
                                                                                                                                                                                                                                         attenuated the development of tolerance (P < 0.01). Neither baseline tail-flick latency nor the response to a single injection of 20 g of
                                                                                                                                                                                                                                         morphine changed after repeated saline treatment for 7 days. Coadministration of 20 g of morphine with benzamide (100400 nmol) for
                                                                                                                                                                                                                                         7 days also reliably prevented the increase in dark neurons (P < 0.01; Fig. 2 ). Neither repeated benzamide (400 nmol) treatment alone nor
                                                                                                                                                                                                                                         a single injection of 20 g of morphine on day 8 (the 20*/0 group) affected the occurrence of dark neurons as compared with the saline
                                                                                                                                                                                                                                         group.
                                                                                                                                                                                                                                           FIG. 1. Effect of benzamide on morphine tolerance. Tolerance to the antinociceptive effect of morphine developed in rats treated with
                                                                                                                                                                                                                                           20 g of morphine for 7 days. Coadministration of 20 g of morphine with 200 or 400 nmol (not 100 nmol) benzamide for 7 days
                                                                                                                                                                                                                                           reliably attenuated the development of tolerance. Neither baseline tail-flick latency nor the response to a single injection of 20 g of
                                                                                                                                                                                                                                           morphine changed after repeated saline treatment for seven days. **, P < 0.01, as compared with that of day 1 in each corresponding
                                                                                                                                                                                                                                           group. Neither repeated benzamide (400 nmol) treatment alone nor a single injection of 20 of g morphine on day 8 (the 20*/0 group)
                                                                                                                                                                                                                                           affected the degree of tolerance as compared with the saline group. MPAE%, percent of maximal possible antinociceptive effect.
                                                                                                                                                                                                                                              The data from the previous experiment showed that benzamide was effective in inhibiting the development of morphine tolerance and
                                                                                                                                                                                                                                         dark neurons. The specificity of this effect to PARS inhibition was examined by utilizing other PARS inhibitors. For this experiment, four
                                                                                                                                                                                                                                         groups of rats were used; they included rats receiving 400 nmol benzamide and 20 g of morphine on days 18, rats receiving 200 nmol 3-
                                                                                                                                                                                                                                         aminobenzamide and 20 g of morphine on days 18, rats receiving 1 mol niacinamide (nicotinamide) and 20 g of morphine on days 1
                                                                                                                                                                                                                                         8, and rats receiving 20 g of morphine and saline on days 18. Coadministration of 20 g of morphine with either 200 nmol 3-
                                                                                                                                                                                                                                         aminobenzamide or 1 mol niacinamide (nicotinamide) for 7 days reliably (P < 0.01) attenuated the development of tolerance as
                                                                                                                                                                                                                                         compared with that of day 1 in the same group ( Fig. 3 ). As shown in Fig. 4 , coadministration of 20 g of morphine with either 200 nmol
                                                                                                                                                                                                                                         3-aminobenzamide or 1 mol niacinamide for 7 days also reliably (P < 0.05 and P < 0.01 for the drugs respectively) prevented the
                                                                                                                                                                                                                                         increase in dark neurons as compared with the morphine + saline group.
                                                                                                                                                                                                                                              To confirm that the development of tolerance and dark neurons in morphine-treated rats is associated with the activation of opioid
                                                                                                                                                                                                                                         receptors, we examined the effect of the opioid receptor antagonist, naltrexone, on the ability of morphine to produce tolerance and dark
                                                                                                                                                                                                                                         neurons. For this experiment, two groups of rats were used; they included rats receiving 10 mg/kg naltrexone intraperitoneally 5 min
                                                                                                                                                                                                                                         before 20 g of morphine i.t. on days 18 and rats receiving 20 g of morphine and saline on days 18. Coadministration of 20 g of
                                                                                                                                                                                                                                         morphine with 10 mg/kg naltrexone for 7 days reliably prevented both the development of the antinociceptive tolerance (P < 0.01) and the
                                                                                                                                                                                                                                         increase in dark neurons (P < 0.01) as compared with the morphine + saline group.
                                                                                                                                                                                                                                              The major findings of this series of studies are (i) the incidence of dark neurons increased significantly within the spinal cord dorsal
                                                                                                                                                                                                                                         horn, particularly the superficial laminae III, of rats injected daily for 8 days with i.t. morphine; (ii) benzamide and other PARS inhibitors
                                                                                                                                                                                                                                         reduced or prevented the development of
the authoritative version for attribution.
                                                                                                                                                                                                                                           FIG. 2. Effect of benzamide on incidence of dark neurons. Coadministration of 20 g of morphine with benzamide (100400 nmol)
                                                                                                                                                                                                                                           for 7 days reliably prevented the increase in dark neurons. Neither repeated benzamide (400 nmol) treatment alone nor a single
                                                                                                                                                                                                                                           injection of 20 g of morphine on day 8 (the 20*/0 group) affected the occurrence of dark neuron as compared with the saline group.
                                                                                                                                                                                                                                           **, P < 0.01, as compared with each of the rest groups.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
CELLULAR MECHANISMS OF NEUROPATHIC PAIN, MORPHINE TOLERANCE, AND THEIR INTERACTIONS 7734
                                                                                                                                                                                                                                         analgesic tolerance and dark neurons; and (iii) the development of dark neurons after repeated morphine injection is opioid receptor-
                                                                                                                                                                                                                                         mediated, because concurrent administration of naltrexone with morphine prevents the development of dark neurons. These studies
                                                                                                                                                                                                                                         demonstrate that in vivo administration of morphine for 8 days produces dark neurons similar in morphology and location to transsynaptic
                                                                                                                                                                                                                                         alterations resulting from chronic constrictive injury of the sciatic nerve. The morphological characteristics of these cells are consistent
                                                                                                                                                                                                                                         with those of cells undergoing programmed cell death.
                                                                                                                                                                                                                                           FIG. 3. Effect of nicotinamide and 3-aminobenzamide on morphine tolerance. Coadministration of 20 g of morphine with either 200
                                                                                                                                                                                                                                           nmol 3-aminobenzamide or 1 mol niacinamide (nicotinamide) for 7 days reliably attenuated the development of tolerance. **, P <
                                                                                                                                                                                                                                           0.01, as compared with that of day 1 in the same group.
                                                                                                                                                                                                                                              These results, along with others, indicate that the cascade of events leading to the formation of dark neurons in the spinal cord after
                                                                                                                                                                                                                                         morphine administration involves activation of the NMDA receptor, PKC, NO, and PARS. Thus, morphine tolerance may be reduced by
                                                                                                                                                                                                                                         intervening at any one of these steps. These findings suggest that neurotoxicity contributes to neural mechanisms underlying opioid
                                                                                                                                                                                                                                         tolerance and that PARS inhibitors are protective from such neurotoxicity. The results from these studies, combined with our results in
                                                                                                                                                                                                                                         similar studies using the CCI model ( 30 ), suggest that opioids may exacerbate the excitotoxicity underlying at least certain types of
                                                                                                                                                                                                                                         neuropathic pain. A corollary of this is that the excitotoxicity from neuropathic pain may, under some circumstances, reduce the response
                                                                                                                                                                                                                                         to opioids.
                                                                                                                                                                                                                                           FIG. 4. Effect of nicotinamide and 3-aminobenzamide on incidence of dark neurons. Coadministration of 20 g of morphine with
                                                                                                                                                                                                                                           either 200 nmol 3-aminobenzamide or 1 mol niacinamide (nicotinamide) for 7 days also reliably prevented the increase in dark
                                                                                                                                                                                                                                           neurons. a, P < 0.05; b, P < 0.01, as compared with the MSO4 + saline group.
                                                                                                                                                                                                                                               Several perplexing observations about neuropathic pain, opioid tolerance, and the interactions between them led us to propose an
                                                                                                                                                                                                                                         early model of the events involved ( 34 ) and now lead us to propose a revised version of this model here ( Fig. 5 ). These observations
                                                                                                                                                                                                                                         include the following. (i) Neuropathic pain syndromes often present with symptoms indicative of both hyperexitability (e.g., hyperalgesia)
                                                                                                                                                                                                                                         and disinhibition (e.g., spontaneous pain, allodynia). Some of these symptoms, such as hyperalgesia, can be at least partially reversed in
                                                                                                                                                                                                                                         animal models by NMDA receptor antagonists ( 35 ), whereas others, such as allodynia, cannot ( 36 ). (ii) NMDA receptor antagonists
                                                                                                                                                                                                                                         prevent, but do not acutely reverse, tolerance to opioids ( 37 ). (iii) Our model assumes, for numerous reasons ( 34 ), that postsynaptic
                                                                                                                                                                                                                                         opioid and NMDA receptors are on the same neurons, at least in the spinal cord. In fact, we have reported direct immunohistochemical
                                                                                                                                                                                                                                         evidence of this ( 38 ). Opioids are known to produce hyperpolarization via an inwardly rectifying K+ channel. Despite the hyperpolarized
                                                                                                                                                                                                                                         state in these cells, voltage/ligand-gated NMDA receptors must be activated during the process of tolerance development, because NMDA
                                                                                                                                                                                                                                         receptor antagonists block the development of tolerance. (iv) At the spinal cord level, it is likely, except in the presence of nociceptive
                                                                                                                                                                                                                                         input, that presynaptic elements release only small amounts of glutamate onto postsynaptic elements in the superficial laminae of the
                                                                                                                                                                                                                                         dorsal horn that contain NMDA receptors related to opioid tolerance, yet tolerance to opioids occurs in the absence of nociceptive input.
                                                                                                                                                                                                                                         (v) PKC translocation to the cell membrane is greatly increased by chronic as compared with acute morphine treatment ( 34 ). (vi) Chronic
                                                                                                                                                                                                                                         opioid administration produces hyperalgesia, which can be reversed by NMDA receptor antagonists ( 27 ). (vii) CCI, which produces
                                                                                                                                                                                                                                         hyperalgesia causes a rightward shift in the dose response curve to morphine ( 39 ). (viii) NO is involved in opioid tolerance ( 40 ). (ix)
                                                                                                                                                                                                                                         CCI-induced hyperalgesia ( 30 ) and opioid tolerance ( 39 ) are associated with the development of dark neurons ( 30 ). As reviewed in this
                                                                                                                                                                                                                                         article, the development of hyperalgesia, morphine tolerance, and dark neurons can be prevented by inhibiting the nuclear repair enzyme
                                                                                                                                                                                                                                         PARS.
                                                                                                                                                                                                                                               A model of our current working hypothesis concerning the development of neuropathic pain, opioid tolerance, and their interactions
                                                                                                                                                                                                                                         that is consistent with these observations is presented in Fig. 5 . In this model, excessive release of glutamate resulting from peripheral
                                                                                                                                                                                                                                         events, such as those occurring in the CCI model, initiates a series of intracellular events, which, via different messenger systems, leads to
                                                                                                                                                                                                                                         (i) at least partially NMDA antagonist reversible hyperexcitability that results from a PKC-mediated alteration of NMDA receptors
                                                                                                                                                                                                                                         (pathway 1, Fig. 5 ); (ii) events such as allodynia, which are not reversible with NMDA antagonists ( 36 ) but are prevented by inhibition
                                                                                                                                                                                                                                         of the NO/PARS pathway ( 30 ) and thus may be mediated by cellular dysfunction resulting from depletion of cellular energy stores
the authoritative version for attribution.
                                                                                                                                                                                                                                         (pathway 2, Fig.5 ). This cellular dysfunction may be morphologically manifested by the appearance of dark neurons, which can also be
                                                                                                                                                                                                                                         prevented by inhibition of the NO/PARS pathway ( 30 ); and (iii) the rightward shift of the morphine doseresponse curve resulting from
                                                                                                                                                                                                                                         CCI ( 39 ), which has been hypothesized to result from relatively long duration, PKC-mediated alterations in gene expression (pathway 3,
                                                                                                                                                                                                                                         Fig. 5 ), which may result in PKC-mediated opioid receptor/K+ channel uncoupling ( 41 ).
                                                                                                                                                                                                                                               With regard to opioid tolerance, in this model, activation of the -opioid receptor may initiate PKC translocation to the membrane (
                                                                                                                                                                                                                                         42 ). This PKC translocation allows the NMDA receptor to function as a ligand-gated channel by removal of the voltagedependent Mg2+
                                                                                                                                                                                                                                         blockade ( 15 ). The removal of the Mg2+ blockade from the NMDA receptor allows for an increased influx of Ca2+ despite membrane
                                                                                                                                                                                                                                         hyperpolarization by -opioids and low levels of presynaptic glutamate release. This influx of Ca2+ has two effects. It activates either a
                                                                                                                                                                                                                                         separate pool of PKC (PKC2) or much greater amounts of the original pool of PKC1. The other
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
CELLULAR MECHANISMS OF NEUROPATHIC PAIN, MORPHINE TOLERANCE, AND THEIR INTERACTIONS 7735
                                                                                                                                                                                                                                         pool of PKC may be translocated directly to the membrane, modifying various excitatory amino acid and/or other receptors (pathway 1),
                                                                                                                                                                                                                                         and/or it may modify nuclear transcription (pathway 3), the products of which result in delayed and persistent changes in cellular function
                                                                                                                                                                                                                                         such as opioid receptor/K+ channel uncoupling ( 41 ). This consequence cannot be reversed by acute administration of NMDA antagonists.
                                                                                                                                                                                                                                         A second effect of the influx of Ca2+ is that it activates NO synthase, which increases the production of NO and superoxide ( 43 ) (pathway
                                                                                                                                                                                                                                         2). The simultaneous generation of these two molecules favors the production of peroxynitrite (ONOO) ( 44 ). ONOO is a very potent
                                                                                                                                                                                                                                         initiator of DNA strand breakage ( 45 ). Thus, ONOO initiates the production of the nuclear repair enzyme, PARS. Pronounced activation
                                                                                                                                                                                                                                         of PARS can result in cell dysfunction and eventually cell death because of inhibition of mitochondrial respiration and depletion of
                                                                                                                                                                                                                                         cellular energy stores ( 33 ). This then leads to the formation of dark neurons perhaps by way of programmed cell death. Such an
                                                                                                                                                                                                                                         excitotoxic cascade may underlie at least some aspects of opioid tolerance. A third series of events initiated by influx of Ca2+ is the
                                                                                                                                                                                                                                         activation of either a separate pool of PKC (PKC2, Fig. 5 ) or much greater amounts of the original pool of PKC1. This other pool of PKC
                                                                                                                                                                                                                                         may function as a transcription factor resulting in the production of more PKC (PKC3, Fig. 5 ), which can modulate -opioid receptor
                                                                                                                                                                                                                                         responsiveness ( 46 ), resulting in desensitization of the -opioid receptor coupling through its associated G protein to an inwardly
                                                                                                                                                                                                                                         rectifying K+ channel, a mechanism know to exist in vitro ( 40 ).
                                                                                                                                                                                                                                           FIG. 5. A proposed model for the excitotoxic formation of dark neurons in the dorsal horn of the spinal cord from peripheral nerve
                                                                                                                                                                                                                                           injury or repeated morphine administration. Excessive excitation of the NMDA receptor and subsequent influx of Ca2+ occurs either
                                                                                                                                                                                                                                           directly by glutamate release from primary afferent input (CCI model) or indirectly by activation of -opioid receptors (repeated
                                                                                                                                                                                                                                           opiate administration). Activation of the -opioid receptor results in indirect NMDA receptor activation by initiating a second-
                                                                                                                                                                                                                                           messenger PKC translocation to the membrane( 16 ) This PKC translocation activates the NMDA receptor by removal of the Mg2+
                                                                                                                                                                                                                                           blockade( 17 ). The removal of the Mg2+ blockade from the NMDA receptor allows for an increased influx of Ca2+. The influx of Ca2
                                                                                                                                                                                                                                           +, via direct or indirect activation of the NMDA receptor, has several effects ( 5 ). It activates either a separate pool of PKC (PKC ) or
                                                                                                                                                                                                                                                                                                                                                                                2
                                                                                                                                                                                                                                           much greater amounts of the original pool of PKC1. This second pool of PKC may be translocated directly to the membrane,
                                                                                                                                                                                                                                           modifying various excitatory amino acid or other receptors. It also may function as a transcription factor, resulting in the production
                                                                                                                                                                                                                                           of more PKC (PKC3), which can result in uncoupling of the -opioid receptor from its associated G protein. Another effect of the
                                                                                                                                                                                                                                           influx of Ca2+ is that it activates NO synthase, which increases the production of NO. In addition, the influx of Ca2+ results in the
                                                                                                                                                                                                                                           production of superoxide from mitochondria. The simultaneous generation of these two molecules favors the production of
                                                                                                                                                                                                                                           peroxynitrite (ONOO), a very potent initiator of DNA strand breakage, which, in turn, initiates the production of the nuclear repair
                                                                                                                                                                                                                                           enzyme, PARS. Pronounced activation of PARS can result in cell dysfunction and eventually cell death because of inhibition of
                                                                                                                                                                                                                                           mitochondrial respiration and depletion of cellular energy stores, which in turn may lead to the formation of dark neurons, perhaps by
                                                                                                                                                                                                                                           way of programmed cell death. PKCx, various pools of protein kinase C; GPro, heterotrimeric guanine nucleotide binding protein;
                                                                                                                                                                                                                                           NOS, nitric oxide synthase.
                                                                                                                                                                                                                                               Although we have focused here on NMDA receptor activation as a primary initiator of neuropathic pain, opioid tolerance, and their
                                                                                                                                                                                                                                         interactions, these events, as we have pointed out previously (34), are likely to involve additional factors. It is likely that Ca2+/calmodulin,
                                                                                                                                                                                                                                         cAMP, and other second and third messengers, non-NMDA receptors, and cholecystokinin ( 47 ) and other nonglutamate receptors also
                                                                                                                                                                                                                                         participate. Nevertheless, it is clear from the data reviewed here that the complex events resulting from NMDA receptor activation are of
                                                                                                                                                                                                                                         critical importance in neuropathic pain, opioid tolerance, and their interactions.
                                                                                                                                                                                                                                               Clinical Implications. Recent progress in investigating neural mechanisms subserving neuropathic and inflammatory pain as well as
                                                                                                                                                                                                                                         opioid tolerance has significantly advanced our knowledge about pain and pain modulation. These studies represent two important
                                                                                                                                                                                                                                         frontiers in pain research. First, these studies have led to the concept that pathological pain may reflect a disease process with both
the authoritative version for attribution.
                                                                                                                                                                                                                                         dynamic and progressive changes during its course ( 1 , 2 , 9 ). A key feature of this process is that neuronal plastic changes occur within
                                                                                                                                                                                                                                         the CNS in association with the progress of pathological pain states. This concept provides, at least in part, a basis for explaining
                                                                                                                                                                                                                                         pathological pain that often persists long after the initial insults. Second, mechanisms of opioid tolerance may involve neuroplastic
                                                                                                                                                                                                                                         changes within the CNS as well ( 9 , 39 ). Neuroplastic changes in relation to opioid tolerance have much in common with those of
                                                                                                                                                                                                                                         pathological pain, both of which begin with the activation of NMDA receptors ( 9 ). Evidence also exists indicating that interactions do
                                                                                                                                                                                                                                         indeed occur between cellular and intracellular mechanisms of pathological pain and opioid tolerance, and such interactions are likely to
                                                                                                                                                                                                                                         be a contributing factor to a generally weak analgesic effect of opioids in pathological pain states ( 39 ).
                                                                                                                                                                                                                                               Thus far, little information exists with regard to interactions between hyperalgesia and analgesic tolerance in man. It is conceivable
                                                                                                                                                                                                                                         that reduced morphine analgesia after repeated administration to patients with chronic pain could result from the development of both
                                                                                                                                                                                                                                         pharmacological tolerance to morphine analgesia and tolerance-associated hyperalgesia. Because of the coincidental development of
                                                                                                                                                                                                                                         morphine tolerance and tolerance-associated hyperalgesia, progressively higher morphine doses may be needed to overcome both
                                                                                                                                                                                                                                         conditions. In turn, a vicious cycle may be initiated involving higher opiate doses, more tolerance, and greater hyperalgesia. Thus, the need
                                                                                                                                                                                                                                         for higher opiate doses in a clinical setting of opiate treatment could be partly due to the
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
CELLULAR MECHANISMS OF NEUROPATHIC PAIN, MORPHINE TOLERANCE, AND THEIR INTERACTIONS 7736
                                                                                                                                                                                                                                         development of hyperalgesia that may result from repeated opiate administration. Conceivably, the development of tolerance-associated
                                                                                                                                                                                                                                         hyperalgesic states may also contribute to withdrawal signs of opioid dependence. This possibility also suggests that an inappropriate
                                                                                                                                                                                                                                         opiate treatment schedule in pain management may precipitate unexpected hyperalgesic responses to preexisting pain conditions and could
                                                                                                                                                                                                                                         be a source of the clinical complexity with respect to the responsiveness to opiate treatment.
                                                                                                                                                                                                                                               On the other hand, evidence indicating hyperalgesia-associated reduction of morphine antinociception ( 27 ) has bearing on the
                                                                                                                                                                                                                                         controversy concerning opiate effects or lack of effects on neuropathic pain states in man. It is conceivable that the diversity of clinical
                                                                                                                                                                                                                                         response patterns to opiate treatment in neuropathic pain patients may result from varying degrees of CNS neuronal plastic changes
                                                                                                                                                                                                                                         initiated by nerve injury or injury to other tissues. Such neuronal plastic changes can underlie the development of neuropathic pain
                                                                                                                                                                                                                                         syndromes and result in reduced morphine analgesia even before opiate treatment starts ( 9 ). To complicate matters further, neuropathic
                                                                                                                                                                                                                                         pain syndromes as well as other chronic pain states (such as cancer pain) often present a dynamic and progressive course that demands
                                                                                                                                                                                                                                         increased opiate doses for adequate pain relief.
                                                                                                                                                                                                                                               The complexity of opioid tolerance, hyperalgesia, and their interactions calls for a new look into some clinical issues of pain
                                                                                                                                                                                                                                         management. Because the development of pathological pain states often involves the activation of NMDA receptors and because there
                                                                                                                                                                                                                                         exists an intimate relationship between the NMDA and opioid receptor systems that may lead to changes in responsiveness to opioid
                                                                                                                                                                                                                                         analgesics, early recognition of clinical conditions that may lead to the development of pathological pain states would be of utmost
                                                                                                                                                                                                                                         importance. Such clinical conditions may include, but may not be limited to, nerve injury, tissue inflammation, and prolonged and ongoing
                                                                                                                                                                                                                                         peripheral nociceptive input (such as those seen in persistent postoperative pain). Some of these conditions (e.g., postoperative pain) may
                                                                                                                                                                                                                                         be treated preemptively to produce a favorable outcome. However, nerve injury and inflammatory tissue diseases often occur in an
                                                                                                                                                                                                                                         unpredictable manner, thereby obviating the possibility of preemptive treatment. Thus, effective early interruption of ongoing nociceptive
                                                                                                                                                                                                                                         input from the injured site (e.g., using nerve block or field block), thereby reducing CNS activation of NMDA receptors, could be the key
                                                                                                                                                                                                                                         to preventing or minimizing changes in NMDA and opioid systems that may eventually lead to persistent pain and reduced opioid
                                                                                                                                                                                                                                         effectiveness for treating such pain states.
                                                                                                                                                                                                                                               As discussed above, interactions between NMDA and opioid receptors could occur in both directions ( 9 ). Thus, any condition that
                                                                                                                                                                                                                                         results in activation of NMDA receptors within the CNS could modulate opioid receptors, causing reduced efficacy of opioid analgesia;
                                                                                                                                                                                                                                         conversely, repeated treatment with opioids could set up a condition mimicking ongoing nociceptive input through interactions between
                                                                                                                                                                                                                                         opioid and NMDA receptors ( 9 , 39 ). This concept is the basis for recommending a combined use of opioids and clinically available
                                                                                                                                                                                                                                         NMDA receptor antagonists ( 9 ). Importantly, such a strategy should be integrated into the treatment regimen not only for chronic pain
                                                                                                                                                                                                                                         management (treating an existing pain condition) but also for preventing an evolving pain condition, such as that after nerve injury. By the
                                                                                                                                                                                                                                         same token, effective nerve block or field block should be part of an integrated therapeutic regimen for treating clinical conditions that
                                                                                                                                                                                                                                         may later lead to the development of intractable chronic pain states. It should be noted that, although early treatment of pain after tissue
                                                                                                                                                                                                                                         injury and inflammation with opioids often provides satisfactory clinical pain relief, opioids alone offer little help for stopping the process
                                                                                                                                                                                                                                         of an evolving pathological pain state, because evidence presented in this article and elsewhere suggests that opioids alone could actually
                                                                                                                                                                                                                                         contribute to the development of neuronal plastic changes via interactions with NMDA receptors. It can be anticipated that our further
                                                                                                                                                                                                                                         understanding of neural mechanisms subserving hyperalgesia, opioid tolerance, and their interactions would advance and improve clinical
                                                                                                                                                                                                                                         management of debilitating and intractable pain syndromes.
                                                                                                                                                                                                                                               Portions of this work were supported by Public Health Service Grants DA08835 and NS24009.
                                                                                                                                                                                                                                         1. Woolf, C. J. & Thompson, S. W. N. ( 1991 ) Pain 44 , 293299 .
                                                                                                                                                                                                                                         2. Dubner, R. ( 1991 ) in Proceedings of 5th World Congress on Pain , ed. Bond, M. , Charlton, E. & Woolf, C. J. ( Elsevier , Amsterdam ), Vol. 5 , pp.
                                                                                                                                                                                                                                              263276 .
                                                                                                                                                                                                                                         3. Seltzer, Z. , Cohn, S. , Ginzgurg, R. & Berlin, B. ( 1991 ) Pain 45 , 6975 .
                                                                                                                                                                                                                                         4. Davar, G. , Hama, A. , Deykin, A. , Vos, B. & Maciewicz, R. ( 1991 ) Brain Res. 553 , 327330 .
                                                                                                                                                                                                                                         5. Mao, J. , Mayer, D. J. , Hayes, R. L. , Lu, J. & Price, D. D. ( 1992 ) Brain Res. 598 , 271278 .
                                                                                                                                                                                                                                         6. Mao, J. , Price, D. D. , Mayer, D. J. , Lu, J. & Hayes, R. L. ( 1992 ) Brain Res. 576 , 254262 .
                                                                                                                                                                                                                                         7. Mao, J. , Price, D. D. , Hayes, R. L. , Lu, J. , Mayer, D. J. & Frenk, H. ( 1993 ) Brain Res. 605 , 164168 .
                                                                                                                                                                                                                                         8. Tal, M. & Bennett, G. J. ( 1993 ) Neurosci. Lett. 151 , 107110 .
                                                                                                                                                                                                                                         9. Mao, J. , Price, D. D. & Mayer, D. J. ( 1995 ) Pain 62 , 259274 .
                                                                                                                                                                                                                                         10. Kajander, K. C. , Wakisaka, S. & Bennett, G. J. ( 1992 ) Neurosci. Lett. 138 , 225228 .
                                                                                                                                                                                                                                         11. Sato, J. & Perl, E. R. ( 1991 ) Science 251 , 16081610 .
                                                                                                                                                                                                                                         12. Kaczmarek, L. K. ( 1987 ) Trends Neurosci. 10 , 3034 .
                                                                                                                                                                                                                                         13. Numann, R. , Catterall, W. A. & Scheuer, T. ( 1991 ) Science 254 , 115118 .
                                                                                                                                                                                                                                         14. West, J. W. , Numann, R. , Murphy, B. J. , Scheuer, T. & Catterall, W. A. ( 1991 ) Science 254 , 866868 .
                                                                                                                                                                                                                                         15. Chen, L. & Huang, L. Y. M. ( 1992 ) Nature (London) 356 , 521523 .
                                                                                                                                                                                                                                         16. Liu, H. , Wang, H. , Sheng, L. Y. , Jan, Y. N. & Basbaum, A. I. ( 1994 ) Soc. Neurosci. Abstr. 20 , 482 .
                                                                                                                                                                                                                                         17. Kitto, K. F , Haley, J. E. & Wilcox, G. L. ( 1992 ) Neurosci. Lett. 148 , 15 .
                                                                                                                                                                                                                                         18. Palecek, J. , Paleckova, V. , Dougherty, P. M. & Willis, W. D. ( 1994 ) J. Neurophysiol. 71 , 529537 .
                                                                                                                                                                                                                                         19. Gerber, G. , Kangraga, I. , Ryu, P. D. , Larew, J. S. A. & Randic, M. ( 1989 ) J. Neurosci. 9 , 36063617 .
                                                                                                                                                                                                                                         20. Mao, J. , Mayer, D. J. , Hayes, R. L. & Price, D. D. ( 1993 ) J. Neurophysiol. 70 , 470481 .
                                                                                                                                                                                                                                         21. Mao, J. , Price, D. D. , Phillips, L. L. , Lu, J. & Mayer, D. J. ( 1995 ) Brain Res. 677 , 257267 .
                                                                                                                                                                                                                                         22. Laird, J. M. A. & Bennett, G. J. ( 1993 ) J. Neurophysiol. 69 , 20712085 .
                                                                                                                                                                                                                                         23. Palecek, J. , Paleckova, V. , Dougherty, P. M. , Carlton, S. M. & Willis, W. D. ( 1992 ) J. Neurophysiol. 67 , 15621573 .
                                                                                                                                                                                                                                         24. Guilbaud, G. , Benoist, J. M. , Jazat, F. & Gautron, M. ( 1990 ) J. Neurophysiol. 64 , 15371554 .
                                                                                                                                                                                                                                         25. Bennett, G. J. & Xie, Y. K. ( 1988 ) Pain 33 , 87107 .
                                                                                                                                                                                                                                         26. Mao. J. , Coghill, R. C. , Mayer, D. J. & Hayes, R. L. ( 1992 ) Pain 50 , 89100 .
                                                                                                                                                                                                                                         27. Mao, J. , Mayer, D. J. & Price, D. D. ( 1993 ) J. Neurosci. 13 , 26892702 .
                                                                                                                                                                                                                                         28. Hylden, J. L. K. , Nahin, R. L. , Traub, R. J. & Dubner, R. ( 1989 ) Pain 37 , 329243 .
                                                                                                                                                                                                                                         29. Sugimoto, T. , Bennett, G. J. & Kajander, K. C. ( 1990 ) Pain 42 , 205213 .
                                                                                                                                                                                                                                         30. Mao, J. , Price, D. D. , Zhu, J. , Lu, J. & Mayer, D. J. ( 1997 ) Pain 72 , 355366 .
                                                                                                                                                                                                                                         31. Price, D. D. , Long, S. & Huitt, C. ( 1992 ) Pain 49 , 163174 .
the authoritative version for attribution.
DOES A NEUROIMMUNE INTERACTION CONTRIBUTE TO THE GENESIS OF PAINFUL PERIPHERAL NEUROPATHIES? 7737
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                               GARY J. BENNETT *
                                                                                                                                                                                                                                               Department of Neurology, MCP Hahnemann University, Philadelphia, PA 19102
                                                                                                                                                                                                                                               ABSTRACT Painful peripheral neuropathies are precipitated by nerve injury from disease or trauma. All such injuries will
                                                                                                                                                                                                                                         be accompanied by an inflammatory reaction, a neuritis, that will mobilize the immune system. The role of the inflammation itself
                                                                                                                                                                                                                                         is difficult to determine in the presence of structural damage to the nerve. A method has been devised to produce a focal neuritis in
                                                                                                                                                                                                                                         the rat sciatic nerve that involves no more than trivial structural damage to the nerve. This experimental focal neuritis produces
                                                                                                                                                                                                                                         neuropathic pain sensations (heat- and mechano-hyperalgesia, and cold- and mechano-allodynia) in the ipsilateral hind paw. The
                                                                                                                                                                                                                                         abnormal pain sensations begin in 12 days and last for 46 days, with a subsequent return to normal. These results suggest that
                                                                                                                                                                                                                                         there is a neuroimmune interaction that occurs at the outset of nerve injury (and perhaps episodically over time in slow developing
                                                                                                                                                                                                                                         conditions like diabetic neuropathy) that produces neuropathic pain. The short duration of the phenomena suggest that they may
                                                                                                                                                                                                                                         prime the system for more slowly developing mechanisms of abnormal pain (e.g., ectopic discharge in axotomized primary
                                                                                                                                                                                                                                         afferent neurons) that underlie the chronic phase of painful neuropathy.
                                                                                                                                                                                                                                               Painful peripheral neuropathies begin with nerve injury caused by disease or trauma. This injury will result in an inflammatory
                                                                                                                                                                                                                                         reaction, a neuritis, that will mobilize the immune system. It is important to note that this will occur not only in cases of injury caused by
                                                                                                                                                                                                                                         infection and autoimmune disorder (e.g., herpes zoster and Guillain-Barr syndrome) but also in cases of sterile injury because cellular
                                                                                                                                                                                                                                         debris is an inflammatory and immune stimulus. It is difficult to study the role of the inflammation and the immune system response when
                                                                                                                                                                                                                                         it presents together with structural damage to axons because the structural damage itself gives rise to pathogenic mechanisms that lead to
                                                                                                                                                                                                                                         pain: for example, ectopic discharge in injured nociceptors. We have devised a method for producing a focal neuritis in the rat that is
                                                                                                                                                                                                                                         accompanied by little or no structural injury to the nerve ( 1 ). We find that this neuritis produces neuropathic pain sensations in the
                                                                                                                                                                                                                                         ipsilateral hind paw, even though the inflammation is at mid-thigh level.
                                                                                                                                                                                                                                               We have used adult male SpragueDawley rats. The neuritis is produced by loosely wrapping the nerve at mid-thigh level with
                                                                                                                                                                                                                                         hemostatic oxidized cellulose (Oxycell, Parke-Davis) that then is saturated with an inflammatory stimulus. The Oxycell does not constrict
                                                                                                                                                                                                                                         the nerve; it serves merely as a sponge for the inflammatory stimulus. As the stimulus, we have used both  carrageenan and complete
                                                                                                                                                                                                                                         Freunds adjuvant with about equal effects; the results described below were obtained with complete Freunds adjuvant. As a control, we
                                                                                                                                                                                                                                         (i) have treated the opposite nerve with Oxycell saturated with saline (ii) and have examined animals with unilateral Oxycell/saline
                                                                                                                                                                                                                                         treatment. These control procedures do not evoke abnormal pain sensations in the hind paw. As a control for the general effects of a
                                                                                                                                                                                                                                         painful thigh, and for the possibility of a systemic response to the inflammatory stimulus, we created an experimental unilateral myositis
                                                                                                                                                                                                                                         by implanting Oxycell/complete Freunds adjuvant in a pocket made in biceps femoris at the same level as the nerve treatment. Animals
                                                                                                                                                                                                                                         with the myositis did not have abnormal pain responses in the hind paw.
                                                                                                                                                                                                                                               Rats with the focal neuritis have heat- and mechano-hyperalgesia and cold- and mechano-allodynia on the ipsilateral hind paw.
                                                                                                                                                                                                                                         Responses from the contralateral hind paw are normal regardless of whether the contralateral side is untreated or treated with Oxycell/
                                                                                                                                                                                                                                         saline.
                                                                                                                                                                                                                                               Heat-hyperalgesia was measured with the paw-flick method of Hargreaves et al. ( 2 ). Abnormal sensitivity was noted within 12
                                                                                                                                                                                                                                         days of treatment and reached peak severity 34 days after treatment. Responsiveness returned to normal within 56 days. The maximum
                                                                                                                                                                                                                                         severity of the heat-hyperalgesia was slightly less than what we have seen with an experimental traumatic nerve injury [the chronic
                                                                                                                                                                                                                                         constriction injury (CCI) model of Bennett and Xie ( 3 )]. All animals with the neuritis developed obvious heat-hyperalgesia.
                                                                                                                                                                                                                                               Mechano-hyperalgesia was measured with the pin-prick method, and mechano-allodynia was measured with the von Frey hair
                                                                                                                                                                                                                                         method as described by Tal and Bennett ( 4 ). Mechano-hyperalgesia and mechano-allodynia were noted within 12 days of treatment,
                                                                                                                                                                                                                                         reached peak severity after 34 days, and resolved to normal within 56 days. The maximum severity of both was comparable to that seen
                                                                                                                                                                                                                                         in CCI rats ( 3 ). All animals with the neuritis developed obvious mechano-hyperalgesia and mechano-allodynia.
                                                                                                                                                                                                                                               Cold-allodynia was assayed with a slight modification of the method described by Choi et al. ( 5 ): 0.15 ml of acetone is sprayed onto
                                                                                                                                                                                                                                         the plantar hind paw while the animal stands on a floor made of screening. On our own forearm skin, this stimulus produces a strong but
                                                                                                                                                                                                                                         non-painful cooling sensation (as the acetone evaporates). Normal rats either ignore the stimulus, or it produces a very brief and small
                                                                                                                                                                                                                                         withdrawal reflex. Neuropathic animals react with a large and prolonged withdrawal response (painful peripheral neuropathy patients with
                                                                                                                                                                                                                                         cold-allodynia complain that this stimulus produces a severe burning pain sensation). Approximately one-half of the neuritis rats displayed
                                                                                                                                                                                                                                         neuropathic responses to cold. This is in contrast to the CCI model, in which nearly every rat has an abnormal response. When present,
                                                                                                                                                                                                                                         cold-allodynia was detected within 23 days of treatment, peaked within 34 days, and resolved within 45 days. Light- and electron-
                                                                                                                                                                                                                                         microscopic analyses of the treated region of the nerve harvested at the time of peak symptom severity (34 days after treatment)
the authoritative version for attribution.
DOES A NEUROIMMUNE INTERACTION CONTRIBUTE TO THE GENESIS OF PAINFUL PERIPHERAL NEUROPATHIES? 7738
                                                                                                                                                                                                                                         showed that some cases had no detectable axonal injury. In one case only (of three that were examined), we detected 20 degenerating
                                                                                                                                                                                                                                         axons confined to a small patch just below the epineurium. The presence of a few tens of degenerating axons is trivial and is very highly
                                                                                                                                                                                                                                         unlikely to produce the marked signs of neuropathic pain that were present in every animal. In all cases, there were clear signs of an
                                                                                                                                                                                                                                         endoneurial inflammatory reaction (even though the inflammatory stimulus was applied to the outside of the nerve). The spaces between
                                                                                                                                                                                                                                         axons was greater than normal, indicating an edematous reaction, and immune cells (macrophages, polymorphonuclear leukocytes, and
                                                                                                                                                                                                                                         monocytes) were present throughout the endoneurial compartment. Immunocytochemical staining identified CD4 and CD8 T-lymphocytes
                                                                                                                                                                                                                                         amongst this infiltrate. The T-cells surrounded the nerve on the outside surface of the epineurium (to be expected because this was where
                                                                                                                                                                                                                                         the inflammatory stimulus was applied), but they were also within the nerve. The endoneurial cells were most abundant toward the center.
                                                                                                                                                                                                                                         This suggests that the cells within the nerve arrived via the endoneurial vasculature because, if they had migrated from the outside, they
                                                                                                                                                                                                                                         would have been concentrated just beneath the epineurium.
                                                                                                                                                                                                                                               The neuropathic pain produced by the neuritis lasted for only a few days. It therefore cannot be the sole mechanism for chronic
                                                                                                                                                                                                                                         painful peripheral neuropathies. It is possible, however, that it is of clinical importance. First, there may be conditions under which nerves
                                                                                                                                                                                                                                         are chronically inflamed or suffer from repeated episodes of inflammation. Of importance, the inflammation may be in structures near
                                                                                                                                                                                                                                         nerves that expose the nerve as an innocent bystander to an inflammatory milieu. For example, in diabetes, nerves may experience
                                                                                                                                                                                                                                         repeated episodes of inflammation as the underlying microvascular and metabolic disease processes wax and wane. Nerves that are near
                                                                                                                                                                                                                                         tumors may be bathed in an inflammatory soup of tumor-products and inflammatory cytokines directed against the malignancy. It is
                                                                                                                                                                                                                                         noteworthy that the nucleus pulposus is a very potent inflammatory stimulus ( 6 ), so that a dorsal root lying near a leaky intervertebral
                                                                                                                                                                                                                                         disc may be exposed to an inflammatory environment. Second, in those conditions under which the neuritis is likely to be acute (e.g., post-
                                                                                                                                                                                                                                         herpetic neuralgia), the neuritis-evoked neuropathic pain may prime the nervous system such that pathogenic mechanisms that develop
                                                                                                                                                                                                                                         later (e.g., spontaneous ectopic discharge in injured nociceptors, sprouting sympathetic efferent axons in the dorsal root ganglia, A- low-
                                                                                                                                                                                                                                         threshold mechanoreceptors invading laminae I and II) are able to produce chronic neuropathic pain.
                                                                                                                                                                                                                                               Current work with this model is attempting to determine the relative roles of the inflammatory mediators derived from the
                                                                                                                                                                                                                                         cycloxygenase and lipoxygenase cascades and the proinflammatory cytokines. We have not found any effect in the neuritis model with
                                                                                                                                                                                                                                         indomethacin, suggesting that the arachidenic acid pathways are not involved (J.-E. Baos, S. Shiiba, and G.J.B., unpublished results). It
                                                                                                                                                                                                                                         has already been shown that tumor necrosis factor  is found in CCI nerves and that tumor necrosis factor  injected into the nerve
                                                                                                                                                                                                                                         produces neuropathic pain symptoms ( 7 ). Sommers et al. ( 8 ) have shown that inhibition of tumor necrosis factor  release with
                                                                                                                                                                                                                                         thalidomide reduces neuropathic pain in the CCI model. We have replicated the results of Sommers et al. in CCI rats but have not found
                                                                                                                                                                                                                                         any effect of thalidomide in the neuritis model (J.-E. Baos, S. Shiiba, and G.J.B., unpublished results). The difference may be attributable
                                                                                                                                                                                                                                         to differing immune system responsesprimarily to cellular debris in the case of the CCI model but to bacterial epitopes in the neuritis
                                                                                                                                                                                                                                         model. We have found that other immune suppressants are effective in the neuritis model; for example, cyclosporin A works well (S.
                                                                                                                                                                                                                                         Shiiba, J.-E. Baos, and G.J.B., unpublished results).
                                                                                                                                                                                                                                         1. Eliav, E. , Herzberg, U. , Ruda, M. A. & Bennett, G. J. ( 1999 ) Pain , in press.
                                                                                                                                                                                                                                         2. Hargreaves, K. , Dubner, R. , Brown, F. , Flores, C. & Joris, J. ( 1988 ) Pain 32 , 7788 .
                                                                                                                                                                                                                                         3. Bennett, G. J. & Xie, Y.-K. ( 1988 ) Pain 33 , 87107 .
                                                                                                                                                                                                                                         4. Tal, M. & Bennett, G. J. ( 1994 ) Pain 57 , 375382 .
                                                                                                                                                                                                                                         5. Choi, Y. , Yoon, Y. W. , Na, H. S. , Kim, S. H. & Chung, J. M. ( 1994 ) Pain 59 , 369376 .
                                                                                                                                                                                                                                         6. Olmarker, K. , Blomquist, J. , Strmberg, J. , Nannmark, U. , Thomsen, P. & Rydevik, B. ( 1995 ) Spine 20 , 665669 .
                                                                                                                                                                                                                                         7. Wagner, R. & Myers, R. R. ( 1996 ) NeuroReport 7 , 28972901 .
                                                                                                                                                                                                                                         8. Sommer, C. , Marziniak, M. & Myers, R. R. ( 1998 ) Pain 74 , 8391 .
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                               ALLAN I. BASBAUM *
                                                                                                                                                                                                                                               Departments of Anatomy and Physiology and W. M. Keck Foundation Center for Integrative Neuroscience, University of California,
                                                                                                                                                                                                                                         San Francisco, CA 94143
                                                                                                                                                                                                                                               ABSTRACT To address the neurochemistry of the mechanisms that underlie the development of acute and persistent pain,
                                                                                                                                                                                                                                         our laboratory has been studying mice with deletions of gene products that have been implicated in nociceptive processing. We
                                                                                                                                                                                                                                         have recently raised mice with a deletion of the preprotachykinin-A gene, which encodes the peptides substance P (SP) and
                                                                                                                                                                                                                                         neurokinin A (NKA). These studies have identified a specific behavioral phenotype in which the animals do not detect a window of
                                                                                                                                                                                                                                         pain intensities; this window cuts across thermal, mechanical, and chemical modalities. The lowered thermal and mechanical
                                                                                                                                                                                                                                         withdrawal thresholds that are produced by tissue or nerve injury, however, were still present in the mutant mice. Thus, the
                                                                                                                                                                                                                                         behavioral manifestations of threshold changes in nociceptive processing in the setting of injury do not appear to require SP or
                                                                                                                                                                                                                                         NKA. To identify relevant neurochemical factors downstream of the primary afferent, we are also studying the dorsal horn second
                                                                                                                                                                                                                                         messenger systems that underlie the development of tissue and nerve injury-induced persistent pain states. We have recently
                                                                                                                                                                                                                                         implicated the  isoform of protein kinase C (PKC) in the development of nerve injury-induced neuropathic pain. Acute pain
                                                                                                                                                                                                                                         processing, by contrast, is intact in the PKC-null mice. Taken together, these studies emphasize that there is a distinct
                                                                                                                                                                                                                                         neurochemistry of acute and persistent pain. Persistent pain should be considered a disease state of the nervous system, not merely
                                                                                                                                                                                                                                         a prolonged acute pain symptom of some other disease conditions.
                                                                                                                                                                                                                                               Recent studies have focused attention on the organization of two major small-diameter primary afferent systems that target dorsal
                                                                                                                                                                                                                                         horn neurons. Broadly speaking, they can be divided into a peptide-containing class of C fibers and one that is generally not associated
                                                                                                                                                                                                                                         with peptides ( 1 ). The peptide-containing neurons can be demonstrated by immunostaining dorsal root ganglia with antisera directed
                                                                                                                                                                                                                                         against the preprotachykinin-A product substance P (SP). These neurons also coexpress calcitonin gene-related peptide (CGRP) and the
                                                                                                                                                                                                                                         neurotrophin receptor TrkA. The second group of afferents can be distinguished because they express a cell surface -galactosyl epitope,
                                                                                                                                                                                                                                         which can be stained with the lectin IB4. Most of the latter neurons also express a fluoride-resistant acid phosphatase (FRAP) ( 2 ) and the
                                                                                                                                                                                                                                         P2X3 subtype of purinergic receptor ( 3 ). It has been suggested, however, that the failure of the peptide-containing group to immunostain
                                                                                                                                                                                                                                         for the P2X3 receptor reflects the inability of antisera to recognize the epitope in that class of neurons, because of coassembly with another
                                                                                                                                                                                                                                         subtype of purinergic receptor (E. W. McCleskey, personal communication). Perhaps of greater relevance to the functional differences of
                                                                                                                                                                                                                                         these two broad classes of afferent is that their patterns of axon termination in the spinal cord dorsal horn differ greatly. The terminals of
                                                                                                                                                                                                                                         the peptide-containing afferents are concentrated in laminae I and outer II of the superficial dorsal horn. The IB4-labeled afferents target
                                                                                                                                                                                                                                         the inner part of lamina II. The same region can be stained for the P2X3 receptor and for FRAP.
                                                                                                                                                                                                                                               Because these afferents are of small diameter, it has generally been assumed that they are nociceptors, even though some
                                                                                                                                                                                                                                         nonnociceptive, low-threshold mechanoreceptive, small-diameter afferents have been identified ( 4 ). Interestingly, the latter group did not
                                                                                                                                                                                                                                         express SP. Given that many neurons in the inner part of lamina II respond to nonnoxious stimuli ( 5 , 6 ), it has been suggested that this
                                                                                                                                                                                                                                         region does not receive input from nociceptors. Despite these observations, we recently provided strong evidence in support of the
                                                                                                                                                                                                                                         hypothesis that the majority of the small-diameter afferents are nociceptors. Specifically, we showed that neurons in both populations
                                                                                                                                                                                                                                         express the vanilloid-1 receptor (VR1) to which capsaicin, the pungent ingredient in hot pepper, binds with high affinity ( 7 ). The fact that
                                                                                                                                                                                                                                         these neurons express VR1 suggests that, at the minimum, these afferents are responsive to a noxious chemical stimulus. Furthermore,
                                                                                                                                                                                                                                         because the capsaicin receptor can be activated by noxious thermal stimuli ( 8 ), it is also likely that the predominantly nonpeptide
                                                                                                                                                                                                                                         population of C fiber includes heat nociceptors. Importantly, not all of the neurons were double-labeled, and we identified a population of
                                                                                                                                                                                                                                         dorsal root ganglion (DRG) cells that was VR1 positive, but stained for neither SP nor IB4. Thus additional classes of primary afferent
                                                                                                                                                                                                                                         nociceptor remain to be characterized. The complexity of the DRG population is highlighted even further by the recent observation that
                                                                                                                                                                                                                                         there are at least two classes of heat nociceptors. Because only one of these responds to capsaicin ( 9 ) it will be of interest to determine the
                                                                                                                                                                                                                                         neurochemical phenotype of the non-VR1 heat nociceptor.
                                                                                                                                                                                                                                               These results establish that the two major classes of small-diameter primary afferent include nociceptors, but that is just the first step
                                                                                                                                                                                                                                         in establishing their contribution to the generation of pain. For example, it is not at all clear to what extent they differ in the types of pain
                                                                                                                                                                                                                                         provoked by their activation. We also do not know whether coincident activity in the different classes of afferent affects the type/quality of
                                                                                                                                                                                                                                         pain that is provoked. Given that the afferents target very different populations of neurons in the superficial dorsal horn, it is also of
                                                                                                                                                                                                                                         interest to address the contribution of these downstream neuronal populations. Importantly, many of the neurons targeted by the peptide
                                                                                                                                                                                                                                         population are projection neurons, which transmit the nociceptive message to brainstem and/or thalamus. These are clustered in lamina I of
                                                                                                                                                                                                                                         the dorsal horn. Indeed, the large majority of neurons that express the neurokinin-1 (NK-1) receptor, which is targeted by SP-containing
                                                                                                                                                                                                                                         afferents, are projection neurons ( 10 ). By contrast, the nonpeptide group of small-diameter afferents targets a region of the superficial
                                                                                                                                                                                                                                         dorsal horn that exclusively contains interneurons, namely the inner part of lamina II.
the authoritative version for attribution.
                                                                                                                                                                                                                                               The problem is not a simple one to address, as it is presently not possible to selectively remove one or the other class of afferent or
                                                                                                                                                                                                                                         postsynaptic neuron. The only exception to this are the very recent studies of Mantyh and colleagues ( 11 ), who used a SP-saporin
                                                                                                                                                                                                                                         conjugate to selectively destroy superficial dorsal horn neurons that express the NK-1 receptor. Our approach to this problem has been to
                                                                                                                                                                                                                                         study the phenotype of mice in which the genes for critical neurotransmitters, neurotransmitter receptors, or second messenger systems
                                                                                                                                                                                                                                         have been deleted. In the following discussion, I will highlight some of our progress in attributing different properties of the pain response
                                                                                                                                                                                                                                         to distinct gene products in afferents and second-order neurons.
                                                                                                                                                                                                                                                                                PREPROTACHYKININ-A PRODUCTS AND ACUTE PAIN
                                                                                                                                                                                                                                               Our first studies evaluated the phenotype of mice in which we deleted the preprotachykinin-A (PPT-A) gene, which encodes the two
                                                                                                                                                                                                                                         tachykinin peptides, SP and neurokinin A (NKA) ( 12 ). These animals appeared normal in simple motor tests, including running on a
                                                                                                                                                                                                                                         rotating rod and when examined in an open field. As expected, we also found that plasma extravasation (PE) in response to injection of
                                                                                                                                                                                                                                         capsaicin was almost abolished in the mutant mice. That result was expected, as peripheral release of SP/NKA is generally required to
                                                                                                                                                                                                                                         evoke PE ( 13 ), a hallmark of neurogenic inflammation. In tests of acute pain, using mild to moderate noxious stimuli, the mutant mice
                                                                                                                                                                                                                                         also responded as the wild-type mice did. By contrast, when the stimulus intensity was increased we observed increased pain responses
                                                                                                                                                                                                                                         only in the wild type. For example, when the heat stimulus was increased from 52.5C to 55C, only the wild-type mice showed a
                                                                                                                                                                                                                                         decreased latency to respond. Decreased responsiveness in the setting of very intense stimuli was true for noxious chemical, thermal, and
                                                                                                                                                                                                                                         mechanical stimuli. These results indicate that SP/NKA contribute to the intensity coding of stimuli across modalities, but that the coding
                                                                                                                                                                                                                                         range is limited to one in which the stimulus is very intense. The fact that very intense stimuli are required to reveal the phenotype is
                                                                                                                                                                                                                                         consistent with our previous studies, which found that internalization of the NK-1 receptor (which provides a measure of tachykinin
                                                                                                                                                                                                                                         release) occurs only when the stimulus is very intense ( 14 ). This result is also consistent with neurochemical studies that found that
                                                                                                                                                                                                                                         higher intensities and higher frequencies of stimulation are required to evoked primary afferent release of SP, compared with glutamate (
                                                                                                                                                                                                                                         15 , 16 ). The latter is a major neurotransmitter of small-diameter primary afferents, and is found in the same terminals, albeit in a different
                                                                                                                                                                                                                                         population of synaptic vesicles ( 17 ). We presume that as the threshold for activating C-fiber nociceptors is crossed, glutamate is released
                                                                                                                                                                                                                                         at central synapses. Via an action at an -amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor, the glutamate
                                                                                                                                                                                                                                         depolarizes postsynaptic projection neurons and interneurons of the dorsal horn, resulting in an acute pain message.
                                                                                                                                                                                                                                               What about the contribution of SP/NKA to persistent pain conditions? Allodynia refers to the production of pain (including reflex
                                                                                                                                                                                                                                         withdrawal) by nonnoxious stimuli. Hyperalgesia refers to an exaggerated pain response produced by noxious stimuli. Although most
                                                                                                                                                                                                                                         studies have reported that the development of allodynia and hyperalgesia results from injury-induced long-term changes/central
                                                                                                                                                                                                                                         sensitization of dorsal horn neurons secondary to activity at the N-methyl-D-aspartate (NMDA) receptor ( 18 , 19 ), other studies have
                                                                                                                                                                                                                                         provided evidence for a contribution of tachykinins, notably SP ( 20  22 ). It was thus surprising that we found no change in the
                                                                                                                                                                                                                                         development of mechanical or thermal allodynia after injury. Importantly, the tests of allodynia involve stimuli that are at or near
                                                                                                                                                                                                                                         threshold. For example, within 24 hr of injecting complete Freunds adjuvant into the hindpaw, which produces a profound hindlimb
                                                                                                                                                                                                                                         inflammation, there is a dramatic drop in the thermal and mechanical withdrawal threshold (allodynia) of the affected limb. We found that
                                                                                                                                                                                                                                         comparable allodynia was present in both wild-type and PPT-A-null mice. The thermal and mechanical allodynia produced after partial
                                                                                                                                                                                                                                         sciatic nerve section ( 23 ) was also intact, despite the loss of SP/NKA. Importantly, we did not specifically test for hyperalgesia, as this
                                                                                                                                                                                                                                         would have required using a noxious stimulus in the setting of injury.
                                                                                                                                                                                                                                               To determine whether the loss of the PPT-A gene products differentially affects the development of allodynia and hyperalgesia, we
                                                                                                                                                                                                                                         are using an electrophysiological approach to study the response of dorsal horn neurons under conditions of inflammation, and in response
                                                                                                                                                                                                                                         to noxious stimuli. In this setting the mice are anesthetized and thus it is possible to administer noxious stimuli, even to an inflamed
                                                                                                                                                                                                                                         hindpaw. Consistent with the behavioral analysis, our electrophysiological results to date suggest that SP/NKA are predominantly
                                                                                                                                                                                                                                         involved under acute conditions, when the stimulus is very intense. We do not have an explanation for the differences between our results
                                                                                                                                                                                                                                         and those of others that implicate SP and NKA in the development of allodynic states. A particularly interesting discrepancy concerns the
                                                                                                                                                                                                                                         observations of Neumann et al. ( 24 ), who reported that in the setting of persistent inflammation, SP is synthesized de novo in large-
                                                                                                                                                                                                                                         diameter afferents and that it contributes to the A  (i.e., large fiber-mediated) mechanical allodynia.
                                                                                                                                                                                                                                               Our results with the PPT-A-null mice also bear on the receptors that are targeted by SP and NKA. It is generally assumed that SP
                                                                                                                                                                                                                                         exerts its effects via the NK-1 receptor ( 25 ). It is, thus, of great interest that the phenotypes of the PPT-A mice and of mice with a
                                                                                                                                                                                                                                         deletion of the NK-1 receptor ( 26 ) differ. Although we found that high-intensity stimuli are not processed normally in the PPT-A mutant
                                                                                                                                                                                                                                         mice, the response to acute noxious stimuli appears to be intact in the NK-1 receptor mutant animals. Furthermore, although the NK-1
                                                                                                                                                                                                                                         receptor mutants showed reduced second-phase formalin test responses, the PPT-A mutant mice had only reduced first-phase behavior.
                                                                                                                                                                                                                                         Although there is disagreement concerning the extent to which central sensitization contributes to second-phase pain in the formalin test (
                                                                                                                                                                                                                                         27 , 28 ), the possibility that the time course and magnitude of central sensitization differ in the wild-type and mutant mice needs to be
                                                                                                                                                                                                                                         tested.
                                                                                                                                                                                                                                               We hypothesize that the differences between the PPT-A mutant and NK-1 receptor mutant mice are indicative of an action of SP and/
                                                                                                                                                                                                                                         or NKA at an additional tachykinin receptor. Although SP predominantly targets the NK-1 receptor, NKA has a higher affinity for the
                                                                                                                                                                                                                                         NK-2 receptor and has reasonable affinity for the NK-3 site ( 25 ). Importantly, there is evidence for an independent contribution of the
                                                                                                                                                                                                                                         NK-2 receptor (and thus of NKA) to nociceptive processing ( 29  31 ). Of particular interest, iontophoresis of NK-2 antagonists reduces
                                                                                                                                                                                                                                         the hyperexcitability of dorsal horn neurons in the setting of inflammation and blocks the central effects of NKA, but not of SP ( 32 ).
                                                                                                                                                                                                                                         Fleetwood-Walker and colleagues ( 33 ) found that NK-2 antagonists were far more effective than NK-1 antagonists at reducing
                                                                                                                                                                                                                                         inflammation-induced up-regulation of dynorphin message in superficial dorsal horn. In preliminary studies, we found significant
                                                                                                                                                                                                                                         induction of Fos expression in laminae I and II after intrathecal injection of NKA in mice with a deletion of the NK-1 receptor. Finally,
                                                                                                                                                                                                                                         Duggan et al. ( 34 ) provided evidence that the central effects of SP and NKA differ, in part because of their differential susceptibility to
                                                                                                                                                                                                                                         the endopeptidases that degrade them. Thus, if NKA targets a receptor to which SP has much lower affinity, its loss in the PPT-A-null
                                                                                                                                                                                                                                         mouse could have profound consequences independent of the loss of SP.
                                                                                                                                                                                                                                               Paradoxically, despite the pharmacological evidence for an independent NK-2 receptor contribution, neither in situ hybridization nor
                                                                                                                                                                                                                                         immunocytochemistry, with one recent exception ( 35 ), has provided convincing evidence that the NK-2
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         receptor is present in the dorsal horn. One possibility is that there is another receptor to which NKA binds, different from the cloned NK-2
                                                                                                                                                                                                                                         receptor, but sensitive to NK-2 antagonists. A somewhat comparable situation exists for  opioid receptors ( 36 ), which are best
                                                                                                                                                                                                                                         distinguished pharmacologically. If we are to understand the mechanisms through which SP/NKA influence short- and long-term
                                                                                                                                                                                                                                         nociceptive processing in the dorsal horn, it is essential that this paradox be addressed. To this end we are now generating mice that
                                                                                                                                                                                                                                         encode either SP or NKA, but not both. We will study these mice in both behavioral and electrophysiological tests of nociception and
                                                                                                                                                                                                                                         compare their phenotypes with those of the PPT-A-null and NK-1 receptor-null mice. Hopefully, this approach will address the differential
                                                                                                                                                                                                                                         contribution of SP and NKA to nociceptive processing.
                                                                                                                                                                                                                                                                                     PROTEIN KINASE C  AND PERSISTENT PAIN
                                                                                                                                                                                                                                               The results described above provide important information on the contribution of the peptide-containing class of primary afferents to
                                                                                                                                                                                                                                         nociceptive processing. Much more difficult is the analysis of the contribution of the nonpeptide class of small-diameter afferents.
                                                                                                                                                                                                                                         Although these neurons express the P2X3 receptor, it is difficult to selectively activate this receptor in vivo so as to address its contribution
                                                                                                                                                                                                                                         to the generation of pain. Because of the difficulty in directly evaluating the afferents, we turned to a likely central target of the afferents,
                                                                                                                                                                                                                                         namely neurons in the inner part of the lamina II.
                                                                                                                                                                                                                                               These studies involved mice with a deletion of the gene that encodes the  isoform of protein kinase C (PKC) ( 37 ). These mice
                                                                                                                                                                                                                                         were of interest for several reasons. Most importantly, there is considerable evidence that PKC contributes to the development of the long-
                                                                                                                                                                                                                                         term changes that underlie injury-associated allodynia and hyperalgesia ( 38  40 ). Unfortunately, there are no selective antagonists
                                                                                                                                                                                                                                         available for the many isoforms of PKC; thus it is impossible to determine their individual contribution. Furthermore, because the different
                                                                                                                                                                                                                                         isoforms are differentially distributed in the dorsal horn (see below), an understanding of the mechanisms through which they operate at
                                                                                                                                                                                                                                         the dorsal horn circuit level is very difficult to achieve. Thus, the opportunity to study mice with a deletion of one isoform is particularly
                                                                                                                                                                                                                                         useful. These mice were also of also interest because, unlike most PKC isoforms, PKC is not expressed until after birth; thus the
                                                                                                                                                                                                                                         likelihood that the deletion is associated with major developmental abnormalities, or for compensatory responses to its loss during
                                                                                                                                                                                                                                         development, is significantly reduced. Any compensatory responses to the deletion would have to have occurred postnatally. The PKC-
                                                                                                                                                                                                                                         null mice are also of interest because studies in hippocampus revealed that the mice show a reduction of long-term potentiation (LTP) (
                                                                                                                                                                                                                                         41 ). Although LTP and long-term changes in spinal cord nociceptive processing secondary to injury are not identical, it was reasonable to
                                                                                                                                                                                                                                         hypothesize that pain-related behaviors that have been associated with alterations in the excitability of dorsal horn nociresponsive neurons
                                                                                                                                                                                                                                         would be altered in these mice.
                                                                                                                                                                                                                                           FIG. 1. (Left) Micrograph from the lumbar spinal cord of the rat. It illustrates the restricted distribution of PKC immunoreactivity to
                                                                                                                                                                                                                                           interneurons in the inner part of lamina II of the superficial dorsal horn. (Right) Distribution of Fos-immunoreactive neurons in the
                                                                                                                                                                                                                                           L4 cord dorsal horn evoked by an injection of formalin into the ipsilateral hindpaw. The rat was killed 1 hr after the formalin
                                                                                                                                                                                                                                           injection. Note that there are many labeled neurons in the most superficial laminae (I and outer II), but that there is a distinct band
                                                                                                                                                                                                                                           (corresponding to the inner part of lamina II; arrows) in which there is very little Fos expression. Thus although the small-diameter
                                                                                                                                                                                                                                           afferents that innervate lamina II are chemonociceptors, their activation appears not to induce Fos in the neurons that they target.
                                                                                                                                                                                                                                           (60.)
                                                                                                                                                                                                                                              Finally, and perhaps, most importantly, we found that the dorsal horn distribution of PKC differs greatly from that of the other
                                                                                                                                                                                                                                         isoforms. First, to our knowledge, PKC is the only isoform that is not found in dorsal root ganglia. Furthermore, PKC immunostaining in
                                                                                                                                                                                                                                         the spinal cord is restricted to a subpopulation of interneurons in the inner part of lamina II ( Fig. 1 ). This distribution greatly differs from
                                                                                                                                                                                                                                         that of other isoforms (e.g., PKC, 1, and 2), which are found throughout the superficial dorsal horn as well as more ventrally. The
                                                                                                                                                                                                                                         presence of PKC in the inner part of lamina II raises the possibility that the phenotype of the deletion mutant is, at least in part, related to
                                                                                                                                                                                                                                         loss of the consequences of activation of the nonpeptide population of primary afferents that target this region. Importantly, the restriction
                                                                                                                                                                                                                                         of PKC to a single interneuron population significantly limits the circuits through which it can influence the development of persistent
                                                                                                                                                                                                                                         pain conditions. It also offers the hope that further neuroanatomical studies at the light and electron microscopic level will be able to
                                                                                                                                                                                                                                         identify the major afferent inputs to and the connections made by these neurons so that the circuits through which these neurons exert their
                                                                                                                                                                                                                                         effects can be understood.
                                                                                                                                                                                                                                              Our studies proved incredibly informative. In distinct contrast to the mice with a deletion of the PPT-A gene, we found that in the
                                                                                                                                                                                                                                         PKC-null mice there was no defect in the processing of acute pain messages. For example, the PKC-null and wild-type mice behaved
                                                                                                                                                                                                                                         identically in tests of thermal pain. By contrast, when we tested the mice in models that involved persistent injury, for example, after
                                                                                                                                                                                                                                         partial sciatic nerve injury, we found a significant decrease in the magnitude of the mechanical and thermal allodynia that developed. This
the authoritative version for attribution.
                                                                                                                                                                                                                                         difference persisted for the duration of the experiment. We concluded that in the absence of PKC, partial nerve injury
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         does not induce the hyperexcitability of dorsal horn neurons that is at the basis of the subsequent allodynia.
                                                                                                                                                                                                                                               Given that PKC is concentrated in interneurons of the inner part of lamina II, we presume that changes in the circuits that involve
                                                                                                                                                                                                                                         these neurons are critical to the failure of allodynia to develop after nerve injury. On the other hand, PKC is present throughout the
                                                                                                                                                                                                                                         neuroaxis. Thus, the defect in nerve injury-induced processing of nociceptive messages need not reflect the loss of enzyme function in the
                                                                                                                                                                                                                                         spinal cord. In preliminary studies using electrophysiological methods in the mouse spinal cord ( 42 ), however, we found that injury in the
                                                                                                                                                                                                                                         PKC-null mice does not lead to an enhancement of the response of nociceptive neurons to nonnoxious stimuli, as it does in the wild-type
                                                                                                                                                                                                                                         mice. This finding more directly points to the cord as the locus of the defect.
                                                                                                                                                                                                                                               Finally, we found that the spinal cord neuroanatomical consequences of nerve injury are also significantly reduced in the PKC-null
                                                                                                                                                                                                                                         mice. For example, numerous studies have demonstrated that there is a decrease in immunoreactivity for SP ( 43 ) and an increase in
                                                                                                                                                                                                                                         staining for the NK-1 receptor after sciatic nerve section ( 44 ). Although comparable changes were readily demonstrated in the wild-type
                                                                                                                                                                                                                                         mice, the changes were much reduced, and in some cases absent, in the mutant mice. Similarly, although we found that partial sciatic
                                                                                                                                                                                                                                         nerve section induces an up-regulation of neuropeptide Y immunoreactivity in the superficial dorsal horn, this was also significantly
                                                                                                                                                                                                                                         reduced in the PKC-null mice. Taken together, these results suggest either that the signal that is sent from the site of nerve injury to the
                                                                                                                                                                                                                                         dorsal root ganglia and spinal cord is not transmitted properly in the PKC-null mice, or that the response to the signal by dorsal horn
                                                                                                                                                                                                                                         neurons is altered. We favor the latter hypothesis and suggest that it is a signal in the nonpeptide population of small-diameter nociceptors
                                                                                                                                                                                                                                         that is not processed normally by the PKC interneurons of the inner part of lamina II. How this comes about, and the extent to which such
                                                                                                                                                                                                                                         changes can be prevented or reversed, remains to be determined.
                                                                                                                                                                                                                                               The fact that the PKC-null mice behaved normally in tests of acute pain suggests that the contribution of these neurons is manifest
                                                                                                                                                                                                                                         only when noxious inputs persist, i.e., in the setting of injury. In fact, it is remarkably difficult to demonstrate that neurons in the inner part
                                                                                                                                                                                                                                         of lamina II, are activated by acute noxious stimuli. As noted above, electrophysiological studies have emphasized the responsiveness of
                                                                                                                                                                                                                                         many of the neurons in inner II to nonnoxious mechanical stimuli ( 5 , 6 ). Furthermore, even a very intense noxious stimulus rarely
                                                                                                                                                                                                                                         induces Fos expression in the interneurons of inner lamina II ( 45 ). This is true even when formalin ( Fig. 1 ) or capsaicin is used as the
                                                                                                                                                                                                                                         stimulus. Because the bulk of the small-diameter afferents that target inner lamina II express the capsaicin receptor, it is likely that this
                                                                                                                                                                                                                                         stimulus activates the interneurons of this region. We have tried repeatedly to find a stimulus that will induce Fos in the PKC population
                                                                                                                                                                                                                                         of neurons, but have not succeeded. To our knowledge, the only consistent stimulus that induces Fos in neurons of inner II is kainate
                                                                                                                                                                                                                                         injection in the raphe magnus of the medulla ( 46 ). Studies in our laboratory, however, found that even this stimulus rarely induces Fos
                                                                                                                                                                                                                                         expression in the PKC population of interneurons. It is, of course, possible that the PKC neurons do not express Fos; that alone indicates
                                                                                                                                                                                                                                         that they are unusual, at least when compared with other dorsal horn neurons that receive inputs from primary afferent nociceptors.
                                                                                                                                                                                                                                               Given the nature of the afferents to the inner part of lamina II (i.e., that they express VR1) we find it very difficult to accept the
                                                                                                                                                                                                                                         notion that this region exclusively, or even predominantly, subserves a nonnociceptive function. Rather, it appears that the conditions
                                                                                                                                                                                                                                         under which these neurons are activated remains to be established. In part to address this question, we have established an in vitro spinal
                                                                                                                                                                                                                                         cord slice preparation in which translocation of PKC in these interneurons (which occurs when the enzyme is activated) can be
                                                                                                                                                                                                                                         monitored. We hope that this will permit a more extensive analysis of the neurochemistry of the inputs that activate these neurons. That
                                                                                                                                                                                                                                         information will then be used to study the neurons in the intact animal.
                                                                                                                                                                                                                                               In summary, our studies using mice with deletions of specific genes have demonstrated that particular features of the responses to
                                                                                                                                                                                                                                         acute and persistent injury conditions are differentially influenced by SP/NKA and PKC. Our present studies are directed at identifying
                                                                                                                                                                                                                                         the circuits through which these distinct phenotypes are generated. In one series of studies we are generating a very detailed analysis of the
                                                                                                                                                                                                                                         circuits in which PKC-containing interneurons participate and a comprehensive description of their neurochemical phenotype ( 47 ). That
                                                                                                                                                                                                                                         information will provide a better understanding of the mechanisms through which these neurons influence nociceptive processing in the
                                                                                                                                                                                                                                         setting of injury and will hopefully be useful in the development of approaches to treating these conditions.
                                                                                                                                                                                                                                               This work was supported by National Institutes of Health Grants NS 21445, DA 08377, DE 08973, and NS 14627.
                                                                                                                                                                                                                                         1. Snider, W. D. & McMahon, S. B. ( 1998 ) Neuron 20 , 629632 .
                                                                                                                                                                                                                                         2. Silverman, J. D. & Kruger, L. ( 1988 ) Somatosens. Res. 5 , 219246 .
                                                                                                                                                                                                                                         3. Vulchanova, L. , Riedl, M. S. , Shuster, S. J. , Buell, G. , Surprenant, A. , North, R. A. & Elde, R. ( 1997 ) Neuropharmacology 36 , 12291242 .
                                                                                                                                                                                                                                         4. Lawson, S. N. , Crepps, B. A. & Perl, E. R. ( 1997 ) J. Physiol (London) 505 , 177191 .
                                                                                                                                                                                                                                         5. Light, A. R. , Trevino, D. L. & Perl, E. R. ( 1979 ) J. Comp. Neurol. 186 , 325330 .
                                                                                                                                                                                                                                         6. Bennett, G. J. , Abdelmoumene, M. , Hayashi, H. & Dubner, R. ( 1980 ) J. Comp. Neurol. 194 , 809827 .
                                                                                                                                                                                                                                         7. Tominaga, M. , Caterina, M. J. , Malmberg, A. B. , Rosen, T. A , Gilbert, H. , Skinner, K. , Raumann, B. E. , Basbaum, A. I. & Julius, D. ( 1998 )
                                                                                                                                                                                                                                              Neuron 21 , 531543 .
                                                                                                                                                                                                                                         8. Caterina, M. J. , Schumacher, M. A , Tominaga, M. , Rosen, T. A. , Levine, J. D. & Julius, D. ( 1997 ) Nature (London) 389 , 816824 .
                                                                                                                                                                                                                                         9. Nagy, I. & Rang, H. ( 1998 ) Neuroscience 88 , 995998 .
                                                                                                                                                                                                                                         10. Marshall, G. E. , Shehab, S. A. S. , Spike, R. C. & Todd, A. J. ( 1996 ) Neuroscience 72 , 255263 .
                                                                                                                                                                                                                                         11. Mantyh, P. W. , Rogers, S. D. , Honor, P. , Allen, B. J. , Ghilardi, J. R. , Li, J. , Daughters, R. S. , Lappi, D. A , Wiley, R. G. & Simone, D. A. (
                                                                                                                                                                                                                                              1997 ) Science 278 , 275279 .
                                                                                                                                                                                                                                         12. Cao, Y. Q. , Mantyh, P. W. , Carlson, E. J. , Gillespie, A.-M. , Epstein, C. J. & Basbaum, A. I. ( 1998 ) Nature (London) 392 , 390394 .
                                                                                                                                                                                                                                         13. Lembeck, F. , Donnerer, J. & Bartho, L. ( 1982 ) Eur. J. Pharm. 85 , 171176 .
                                                                                                                                                                                                                                         14. Abbadie, C. , Trafton, J. , Liu, H. , Mantyh, P. W. & Basbaum, A. I. ( 1997 ) J. Neurosci. 17 , 80498060 .
                                                                                                                                                                                                                                         15. Marvizon, J. C. G. , Martinez, V. , Grady, E. F. , Bunnett, N. W. & Mayer, E. A. ( 1997 ) J. Neurosci. 17 , 81298136 .
                                                                                                                                                                                                                                         16. Duggan, A. W. , Riley, R. C. , Mark, M. A , MacMillan, S. J. A. & Schaible, H. G. ( 1995 ) Neuroscience 65 , 849858 .
                                                                                                                                                                                                                                         17. De Biasi, S. & Rustioni, A. ( 1988 ) Proc. Natl. Acad. Sci. USA 85 , 78207824 .
                                                                                                                                                                                                                                         18. Dickenson, A. H. , Chapman, V. & Green, G. M. ( 1997 ) Gen. Pharmacol. 28 , 633638 .
                                                                                                                                                                                                                                         19. Woolf, C. J. & Thompson, S. ( 1991 ) Pain 44 , 293299 .
                                                                                                                                                                                                                                         20. Dougherty, P. M. , Palecek, J. , Zorn, S. & Wiffis, W. D. ( 1993 ) Brain Res. Rev. 18 , 227246 .
                                                                                                                                                                                                                                         21. Xu, X. J. , Dalsgaard, C. J. & Wiesenfeld, H. Z. ( 1992 ) Neuroscience 51 , 641648 .
                                                                                                                                                                                                                                         22. Ma, Q. P. & Woolf, C. J. ( 1997 ) Eur. J. Pharmacol. 322 , 165171 .
                                                                                                                                                                                                                                         23. Malmberg, A B. & Basbaum, A. I. ( 1998 ) Pain 76 , 215222 .
                                                                                                                                                                                                                                         24. Neumann, S. , Doubell, T. P. , Leslie, T. & Woolf, C. J. ( 1996 ) Nature (London) 384 , 360364 .
                                                                                                                                                                                                                                         25. Maggi, C. A. ( 1995 ) Gen. Pharmacol. 26 , 911944 .
the authoritative version for attribution.
                                                                                                                                                                                                                                         26. De Felipe, C. , Herrero, J. F. , OBrien, J. A , Palmer, J. A , Doyle, C. A , Smith, A. J. , Laird, J. M. , Belmonte, C. , Cervero, F. & Hunt, S. P. (
                                                                                                                                                                                                                                              1998 ) Nature (London) 392 , 394397 .
                                                                                                                                                                                                                                         27. Malmberg, A. B. & Yaksh, T. L. ( 1992 ) Science 257 , 12761279 .
                                                                                                                                                                                                                                         28. Taylor, B. K. , Peterson, M. A. & Basbaum, A. I. ( 1997 ) J. Pharmacol. Exp. Ther. 280 , 876883 .
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
THE GENETIC MEDIATION OF INDIVIDUAL DIFFERENCES IN SENSITIVITY TO PAIN AND ITS INHIBITION 7744
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
THE GENETIC MEDIATION OF INDIVIDUAL DIFFERENCES IN SENSITIVITY TO PAIN AND ITS INHIBITION 7745
                                                                                                                                                                                                                                         icists!), the existing data regarding the nociceptive and analgesic sensitivity of laboratory rodent populations reveal great, and in some
                                                                                                                                                                                                                                         cases qualitative, variability.
                                                                                                                                                                                                                                               Rodent populations of use for genetic analysis can either be produced (e.g., inbred strains, recombinant inbred strains, artificially
                                                                                                                                                                                                                                         selected lines, transgenics) or identified (e.g., spontaneous mutants). The characteristics of these genetic models have been reviewed
                                                                                                                                                                                                                                         elsewhere (e.g., ref. 24 ). The most studied genetic rodent models of relevance to pain include the recombinant inbred (RI) CXBK mouse
                                                                                                                                                                                                                                         strain ( 25 , 26 ), the High Analgesia/Low Analgesia (HA/LA) mouse lines selectively bred for swim stress-induced analgesia (SIA) ( 27 ,
                                                                                                                                                                                                                                         28 ), the High Analgesic Response/Low Analgesic Response (HAR/LAR) mouse lines selectively bred for levorphanol analgesia ( 29 ),
                                                                                                                                                                                                                                         the High Autotomy/Low Autotomy (HA/LA) rat lines selectively bred for autotomy ( 30 ), and the normotensive Wistar Kyoto versus
                                                                                                                                                                                                                                         Spontaneously Hypertensive Rat (WKY/SHR). The former models have been thoroughly reviewed ( 24 ). The literature regarding the
                                                                                                                                                                                                                                         relationship between nociception and genetic or experimentally induced hypertension has also been recently reviewed ( 31 ).
                                                                                                                                                                                                                                               Strain differences. Although less well studied at present, the comparison of inbred strain responses is more relevant to the issue of the
                                                                                                                                                                                                                                         scope of individual differences than the aforementioned models. Inbred strains are derived by repeated (>20 generation) full-sibling (i.e.,
                                                                                                                                                                                                                                         brother  sister) mating ( 32 ). Mating of individuals with common ancestors increases the probability of offspring inheriting two copies of
                                                                                                                                                                                                                                         the same allele identical-by-descent. During inbreeding, therefore, genetic heterozygosity is progressively lost as alleles of initially
                                                                                                                                                                                                                                         segregating genes are fixed into a homozygous state.
                                                                                                                                                                                                                                         Table 1. Rat strain differences of relevance to pain
                                                                                                                                                                                                                                         Trait                Parameters                 Administration          Stimulus        Strain Difference *                            Ref.
                                                                                                                                                                                                                                         Nociceptive sensitivity
                                                                                                                                                                                                                                         Thermal                                                                 TW              WAG > F344                                     117
                                                                                                                                                                                                                                                                                                                 TF              LE = LEW = WIS > F344 = SD                     118
                                                                                                                                                                                                                                                                                                                 HP              LEW > F344                                     119
                                                                                                                                                                                                                                         Electrical                                                              FJ              F344 > SD                                      120
                                                                                                                                                                                                                                         Mechanical                                                              CD              LEW > F344                                     121
                                                                                                                                                                                                                                                                                                                 VF              F344 = LEW = WIS > SD                          42
                                                                                                                                                                                                                                         Chemical                                                                CFA             LEW > SD                                       122
                                                                                                                                                                                                                                                                                                                 CFA             LEW > AVN                                      123
                                                                                                                                                                                                                                         Neuropathic                                                             NT              SD > WKY                                       124
                                                                                                                                                                                                                                                                                                                 NT              LE = SAB = SD = WKY > LEW                      125
                                                                                                                                                                                                                                                                                                                 NT              BUF = SD > BN > WIS > LEW                      126
                                                                                                                                                                                                                                         Analgesia
                                                                                                                                                                                                                                         Morphine             015 mg/kg                 i.p.                    FJ              SD > F344                                     120
                                                                                                                                                                                                                                                              010 mg/kg                 i.p.                    TF              SD > WIS                                       127
                                                                                                                                                                                                                                                              010 mg/kg                 i.p.                    TW              F344 > WAG                                     117
                                                                                                                                                                                                                                                              020 mg/kg                 i.p.                    FT              F344 > LEW                                     41
                                                                                                                                                                                                                                                              010 mg/kg                 i.p.                    TF              LE = SD  LEW = WIS  F344                     118
                                                                                                                                                                                                                                                              010 mg/kg                 s.c.                    HP              SD > WKY                                       128
                                                                                                                                                                                                                                         Codeine              50400 mol/kg             s.c.                    TF              SD > DA                                        116
                                                                                                                                                                                                                                         Clonidine            060 g/kg                 i.p.                    FT              SD > WKY                                      129
                                                                                                                                                                                                                                                              10 g                      i.c.v.                  TW              WAG > F344                                     130
                                                                                                                                                                                                                                         TRH                  1 mg/kg                    i.p.                    TW              WAG > F344                                     131
                                                                                                                                                                                                                                         Serotonin            0300 g/kg                i.v.                    TF              SD > WKY                                       132
                                                                                                                                                                                                                                                              0.5 g                     i.c.v.                  TW              F344 > WAG                                     130
                                                                                                                                                                                                                                         Footshock            1.5mA                                              TW              F344 > SD                                     133
                                                                                                                                                                                                                                         Restraint            30 min in tubes                                    TF              LE = SD > F344 = LEW = WIS                     118
                                                                                                                                                                                                                                         Acupuncture          100 Hz, 13 mA                                     TF              P77PMC > WIS                                   134
                                                                                                                                                                                                                                         Analgesic tolerance
                                                                                                                                                                                                                                         Morphine            14  510 mg/kg             i.p.                    TF              SD > WIS                                       127
                                                                                                                                                                                                                                                             8  10 mg/kg                s.c.                    HP              WKY > SD                                       128
                                                                                                                                                                                                                                               Strain Abbreviations: BN, Brown Norway; BUF, Buffalo; DA, Dark Agouti; F344, Fischer 344; LE, LongEvans (outbred); LEW,
                                                                                                                                                                                                                                         Lewis; SAB, Sabra (outbred); SD, SpragueDawley (outbred); WAG, Wistar Albino Glaxo (WAG/GSto); WIS, Wistar (outbred); WKY,
                                                                                                                                                                                                                                         Wistar Kyoto. Genealogical origins of all inbred rat strains can be found at http://www.informatics.jax.org/bin/strains/search .
                                                                                                                                                                                                                                               Other Abbreviations: CD, colorectal distention; CFA, complete Freunds adjuvant; FJ, flinchjump test; FT, formalin test; HP, hot-
                                                                                                                                                                                                                                         plate test; i.c.v., intracerebroventricular, i.p., intraperitoneal; NT, sciatic and saphenous nerve transection; TF, radiant heat tail-flick test;
                                                                                                                                                                                                                                         TRH, thyrotropin-releasing hormone; TW, hot water tail-immersion/withdrawal test.
                                                                                                                                                                                                                                               * Only studies with significant strain differences are reported. Excluded are studies involving selected lines [including the
                                                                                                                                                                                                                                         spontaneously hypertensive rat (SHR)] and mutants.
                                                                                                                                                                                                                                                Morphine analgesia was significantly attenuated by pretreatment with p-chlorophenylalanine in SD, but not F344 rats.
                                                                                                                                                                                                                                                Clonidine analgesia was naloxone-reversible in SD rats but naloxone-insensitive in WKY rats.
                                                                                                                                                                                                                                                F344 rats also developed increased conditioned analgesia to footshock relative to SD rats.
                                                                                                                                                                                                                                               Tables 1 and 2 present some existing data regarding inbred (and outbred) strain differences of relevance to pain in rats and mice,
                                                                                                                                                                                                                                         respectively. The only obvious generalizations that can be made from Table 1 are the nociceptive sensitivity of the Lewis (LEW) inbred
                                                                                                                                                                                                                                         rat strain and the sensitivity to a wide variety of analgesic manipulations of the outbred Sprague Dawley (SD) strain. Multistrain
                                                                                                                                                                                                                                         comparisons (strain surveys) are far more common in the mouse because of the ready availability of over 30 major inbred strains.
the authoritative version for attribution.
                                                                                                                                                                                                                                         Obvious generalizations from mouse strain surveys are thus harder to make. One exception is the voluminous research demonstrating the
                                                                                                                                                                                                                                         relative sensitivity of the DBA/2 (D2) strain to opioid analgesia compared with the C57BL/6 (B6) strain (not shown in Table 2 , but
                                                                                                                                                                                                                                         reviewed in refs. 33 and 34 ). Although D2 mice display high, and B6 mice display low magnitudes of analgesia, the B6 strain is markedly
                                                                                                                                                                                                                                         more sensitive than the D2 strain to other opioid-mediated phenomena, including locomotor activation, learning/memory, and muscular
                                                                                                                                                                                                                                         rigidity (Straub tail).
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
THE GENETIC MEDIATION OF INDIVIDUAL DIFFERENCES IN SENSITIVITY TO PAIN AND ITS INHIBITION 7746
                                                                                                                                                                                                                                               Strain Abbreviations (substrain identifiers are omitted; in most cases, inbred strains were obtained from The Jackson Laboratory: B6,
                                                                                                                                                                                                                                         C57BL/6; BALB, BALB/c; C3H, C3H/He; CD-1, Hsd:ICR (outbred); CF-1, Hsd:NSA (outbred); CFW, HsdWin:CFW1 (outbred); D2,
                                                                                                                                                                                                                                         DBA/2; ICR, Institute for Cancer Research stock (many suppliers; outbred); SW, Swiss Webster (outbred). Genealogical origins of all
                                                                                                                                                                                                                                         inbred mouse strains can be found in Festing (37) or at http://www.informatics.jax.org/bin/strains/search .
                                                                                                                                                                                                                                               Other Abbreviations: AC, abdominal constriction (writhing) test; CAR, carrageenan; FT, formalin test; i.c.v., intracerebroventricular;
                                                                                                                                                                                                                                         i.p., intraperitoneal; i.v., intravenous; HP, hot-plate test; NalBzoH, naloxone benzoylhydrazone (a 3-opioid agonist); NT, sciatic and
                                                                                                                                                                                                                                         saphenous nerve transection; PNI, peripheral nerve injury (Chung model); TF, radiant heat tail-flick test; TW, hot water tail-immersion/
                                                                                                                                                                                                                                         withdrawal test; VF, von Frey fiber test.
                                                                                                                                                                                                                                               * Only studies with significant strain differences are reported. Excluded are studies involving selected lines, mutants, recombinant
                                                                                                                                                                                                                                         inbred (RI) strains, and non-extant populations. Also excluded are studies specifically comparing the B6 and D2 strains.
                                                                                                                                                                                                                                                Strain differences observed varied with sex.
                                                                                                                                                                                                                                                No analgesic tolerance whatsoever developed in the 129/SvEv substrain used.
                                                                                                                                                                                                                                               Qualitative strain differences. In addition to the quantitative strain differences compiled in Tables 1 and 2 , some very intriguing
                                                                                                                                                                                                                                         qualitative strain differences of relevance to pain have been noted. For instance, a number of investigations have suggested that certain
                                                                                                                                                                                                                                         strains activate opioid analgesic systems after exposure to stress, whereas other strains produce approximately equivalent amounts of SIA,
                                                                                                                                                                                                                                         but of a non-opioid (i.e., naloxone-insensitive) character ( 35  37 ). Vaccarino et al. ( 38 ) reported that naloxone injection produced
                                                                                                                                                                                                                                         paradoxical analgesia on the formalin test in BALB/c mice, but not B6 or outbred CD-1 mice. Fujimoto and colleagues ( 39 ) have
                                                                                                                                                                                                                                         demonstrated convincingly that heroin analgesia is mediated by -opioid receptor activation in outbred Institute for Cancer Research stock
the authoritative version for attribution.
                                                                                                                                                                                                                                         (ICR) mice, but by -receptor activation in outbred Swiss Webster (SW) mice. The same workers have recently ( 40 ) identified -, -, and
                                                                                                                                                                                                                                           -type heroin responders among inbred mouse strains. Vaccarino and Couret, Jr. ( 41 ) observed that the presence of formalin-induced pain
                                                                                                                                                                                                                                         during tolerance induction wholly prevented tolerance development in the Fischer 344 (F344) strain but not in the LEW strain. Lee et al. (
                                                                                                                                                                                                                                         42 ) observed a complete blockade of neuropathic mechanical allodynia after treatment with the -adrenergic receptor antagonist,
                                                                                                                                                                                                                                         phentolamine, in LEW rats; this treatment was wholly ineffective in F344 rats. The authors concluded that this neuropathy was
                                                                                                                                                                                                                                         sympathetically maintained in the former strain only. Finally, Proudfits laboratory has demonstrated ( 43 ) that electrical stimulation-
                                                                                                                                                                                                                                         produced analgesia is reversed by 2-adrenergic antagonists in SD rats from
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
THE GENETIC MEDIATION OF INDIVIDUAL DIFFERENCES IN SENSITIVITY TO PAIN AND ITS INHIBITION 7747
                                                                                                                                                                                                                                         the now defunct Sasco (Omaha, NE), but not in SD rats from HarlanSpragueDawley. This difference may be explained by the
                                                                                                                                                                                                                                         differential projection routes and dorsal horn termination fields (laminae VIIX versus laminae IIV, respectively) of pontospinal
                                                                                                                                                                                                                                         noradrenergic neurons in these substrains ( 44 ).
                                                                                                                                                                                                                                                                                      HERITABILITY OF PAIN-RELATED TRAITS
                                                                                                                                                                                                                                              That individual differences in pain-related traits exist, of course, does not imply that these differences are necessarily attributable to
                                                                                                                                                                                                                                         genetic factors. Familial aggregation of pain pathologies and extremes of pain sensitivity has been repeatedly noted in humans, but such
                                                                                                                                                                                                                                         findings have almost uniformly been attributed to shared environmental variance and/or familial modeling (e.g., refs. 45  49 ). To
                                                                                                                                                                                                                                         separate genetic and environmental factors, twin studies have been conducted (some even featuring adoption), comparing the concordance
                                                                                                                                                                                                                                         rates of pain-related traits in monozygotic (identical) versus dizygotic (fraternal) twins (see ref. 50 ). Heritability (i.e., the proportion of
                                                                                                                                                                                                                                         overall phenotypic variance accounted for by genetic factors) has been estimated as 3958% for migraine ( 51  53 ), 55% for menstrual
                                                                                                                                                                                                                                         pain ( 54 ), 50% for back pain ( 55 ), and 21% for sciatica ( 56 ).
                                                                                                                                                                                                                                              Only one report exists of a twin study of nonpathological, basal pain sensitivity. MacGregor et al. ( 57 ) assessed forehead pressure
                                                                                                                                                                                                                                         pain thresholds in 269 monozygotic pairs and 340 dizygotic pairs, and observed only a slight excess correlation in monozygotic versus
                                                                                                                                                                                                                                         dizygotic twins (r = 0.57 vs. 0.51, respectively). This excess corresponds to a heritability of this trait of only 10% and suggests that the
                                                                                                                                                                                                                                         twin correlations in pain thresholds are largely due to shared environmental factors.
                                                                                                                                                                                                                                              In contrast to this last study in humans, heritability estimates for nociceptive and analgesic sensitivity in mice are fairly high, ranging
                                                                                                                                                                                                                                         from 28% to 76% ( 29 , 58  60 )certainly well within the range considered for further genetic analysis. Because none of the murine
                                                                                                                                                                                                                                         nociceptive assays used are similar to the forehead pressure pain test used by MacGregor et al. ( 57 ), it is difficult at the present time to
                                                                                                                                                                                                                                         evaluate whether humans and mice truly differ in the contribution of genes to pain sensitivity. Even if genetic factors are ultimately
                                                                                                                                                                                                                                         demonstrated to play only a minor role in the determination of individual pain sensitivity in humans, genetic studies of pain may still prove
                                                                                                                                                                                                                                         highly valuable. Such studies, for example, may illuminate those components of pain processing circuitry in mice and humans that are
                                                                                                                                                                                                                                         especially amenable to alteration, knowledge likely to be useful for the development of novel analgesic strategies.
                                                                                                                                                                                                                                                                                 GENETIC CORRELATIONS AMONG PAIN PHENOTYPES
                                                                                                                                                                                                                                               The tools of classical genetics can be used, even in advance of the identification of the relevant genes, to determine whether traits
                                                                                                                                                                                                                                         share genetic mediation (see ref. 61 ). The fact that a given gene can influence more than one trait is known in genetic parlance as
                                                                                                                                                                                                                                         pleiotropy. Pleiotropic actions of genes result in the genetic correlation of traits, because allelic variation in a gene will influence all traits
                                                                                                                                                                                                                                         in which that gene participates. The determination of genetic correlation has proven to be very heuristic, leading to novel theories
                                                                                                                                                                                                                                         regarding the underlying physiological mediation of traits. A number of genetic correlations of pain-related phenotypes have been noted
                                                                                                                                                                                                                                         using selected lines and inbred strains (see ref. 24 ). I would like to focus on two intriguing findings, as follows.
                                                                                                                                                                                                                                               Genetic Correlation of Nociception and Opiate Analgesia. It has been demonstrated by several groups that a negative genetic
                                                                                                                                                                                                                                         correlation exists between initial nociceptive sensitivity and subsequent morphine analgesia ( 27 , 62 , 63 ). That is, mice that are initially
                                                                                                                                                                                                                                         sensitive to noxious stimuli tend to exhibit modest analgesic responses to morphine, whereas mice that are relatively resistant to basal
                                                                                                                                                                                                                                         nociception exhibit robust morphine analgesia. In two separate studies using multiple inbred strains, this correlation was estimated as r =
                                                                                                                                                                                                                                         0.63 to 0.85 ( 62 ) and r = 0.61 ( 58 ). Thus, mice are doubly advantaged or doubly disadvantaged with respect to nociception and
                                                                                                                                                                                                                                         its opiate inhibition. Interestingly, this negative correlation (albeit with n = 2 only) can also be observed when comparing the sexes.
                                                                                                                                                                                                                                               Genetic Correlations Among Nociceptive Assays. We recently tested 11 inbred strains on 12 separate measures of nociception in
                                                                                                                                                                                                                                         common use in the mouse ( 59 , 60 ). The assays used can be placed on a number of dimensions, including etiology (nociceptive,
                                                                                                                                                                                                                                         inflammatory, neuropathic), modality (thermal, chemical, mechanical), duration (acute, tonic, chronic), and location (cutaneous,
                                                                                                                                                                                                                                         subcutaneous, visceral). We reasoned that inbred strain variation could be exploited to identify clusters of genetically correlated
                                                                                                                                                                                                                                         nociceptive assays. Similar genetic mediation implies similar physiological mediation of assays, suggesting that they measure the same
                                                                                                                                                                                                                                         type of pain as defined mechanistically. Essentially, we were attempting to produce a natural rather than artificial taxonomy of
                                                                                                                                                                                                                                         nociception in the mouse, similar to that called for by Woolf et al. ( 64 ).
                                                                                                                                                                                                                                               The results of this effort were variously expected and surprising. By using multivariate analyses, we identified three obvious clusters
                                                                                                                                                                                                                                         of pain tests, in which within-cluster genetic correlations greatly exceeded between-cluster correlations: thermal (Hargreaves test,
                                                                                                                                                                                                                                         hotplate test, tail-immersion/ withdrawal test, and, surprisingly, autotomy), chemical (acetic acid abdominal constriction, magnesium
                                                                                                                                                                                                                                         sulfate abdominal constriction, acute- and tonic-phase formalin test), and mechanical + hypersensitivity (von Frey test, carrageenan
                                                                                                                                                                                                                                         thermal hypersensitivity, peripheral nerve injury thermal, and mechanical hypersensitivity) ( 59 , 60 ). Thus, the stimulus modality
                                                                                                                                                                                                                                         dimension accounted for the obtained genetic correlations to a far greater degree than any other factor. The presence or absence of
                                                                                                                                                                                                                                         neuropathy or inflammation was found to be essentially irrelevant as was the site or duration of the stimulus.
                                                                                                                                                                                                                                                                                     IDENTIFICATION OF PAIN-RELATED GENES
                                                                                                                                                                                                                                              The holy grail of pain genetics, of course, is the actual identification of pain-related genes and the polymorphisms within or near such
                                                                                                                                                                                                                                         genes that account for trait variability. Note that pain-related gene could be broadly defined as any gene encoding a protein of known
                                                                                                                                                                                                                                         pain relevance or of a gene whose null mutant exhibits a pain-related phenotype. Defined in this way, a large number of pain-related genes
                                                                                                                                                                                                                                         are known. However, if more properly defined as one in which allelic variation directly produces individual differences or pathology, only
                                                                                                                                                                                                                                         a handful of pain-related genes have been identified.
                                                                                                                                                                                                                                              Techniques. Essentially, there are two ways to identify genes associated with trait variability: (i) linkage analysis, including classical
                                                                                                                                                                                                                                         model-based linkage techniques and allele-sharing methods in humans and test-crosses in animals, and (ii) association studies (reviewed in
                                                                                                                                                                                                                                         ref. 65 ). Linkage analyses follow familial inheritance patterns, whereas association studies compare allele frequencies in defined
                                                                                                                                                                                                                                         populations. Given the increasingly large number of genes already cloned and mapped in Homo sapiens and Mus musculus, linkage studies
                                                                                                                                                                                                                                         may lead immediately to the identification of candidate genes, which can then be studied by using the latter approach once allelic variants
                                                                                                                                                                                                                                         are found. Candidate genes, of course, can also be evaluated by using nongenetic means, by investigating the physiology of the proteins
                                                                                                                                                                                                                                         they encode. Failing the identification of an already cloned candidate gene, positional cloning techniques (e.g., ref. 66 ) can be used to
the authoritative version for attribution.
                                                                                                                                                                                                                                         narrow the 20-centimorgan (cM)-wide chromosomal region identified by linkage down to the <1-cM-wide region required to realistically
                                                                                                                                                                                                                                         attempt DNA sequencing.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
THE GENETIC MEDIATION OF INDIVIDUAL DIFFERENCES IN SENSITIVITY TO PAIN AND ITS INHIBITION 7748
                                                                                                                                                                                                                                               Human Studies. A handful of single gene pain pathologies have recently been, or are on the verge of being, explained on the DNA
                                                                                                                                                                                                                                         sequence level.
                                                                                                                                                                                                                                               Congenital insensitivity to pain. Over 40 cases of congenital insensitivity to pain (CIP) with preservation of all other sensory
                                                                                                                                                                                                                                         modalities have been reported since the original description of a carnival performer known as The Human Pincushion (see ref. 67 ).
                                                                                                                                                                                                                                         Recently, the genetic basis of this neuropathy (CIP with anhidrosis; hereditary sensory and autonomic neuropathy type IV) was elucidated.
                                                                                                                                                                                                                                         Based in part on the striking similarities between CIP and the phenotype of null-mutant mice lacking the Ntrk1 gene encoding the high
                                                                                                                                                                                                                                         affinity, nerve growth factor-specific tyrosine kinase receptor, Indo et al. ( 68 ) considered the homologous human gene, NTRK1
                                                                                                                                                                                                                                         (previously known as TRKA), as a candidate for CIP. Direct sequencing of the coding region of TRKA in four unrelated CIP patients
                                                                                                                                                                                                                                         revealed three separate, exonic mutations: a single base (C) deletion, an A  C transversion, and a G  C transversion ( 68 ).
                                                                                                                                                                                                                                               Other sensory neuropathies. Nicholson et al. ( 69 ) performed a microsatellite marker genome screen on 102 members of four
                                                                                                                                                                                                                                         Australian kindreds with multiple individuals with hereditary sensory neuropathy type I, a disease featuring loss of all sensory modalities
                                                                                                                                                                                                                                         but especially pain and temperature. Linkage was established to a series of markers encompassing a 5-cM region of chromosome 9. An
                                                                                                                                                                                                                                         obvious candidate gene, NTRK2, encoding the tyrosine kinase receptor type 2 that is bound by brain-derived neurotrophic factor and
                                                                                                                                                                                                                                         neurotrophin-4, was excluded by informative recombination events. Positional cloning efforts facilitated by the development of a yeast
                                                                                                                                                                                                                                         artificial chromosome-based transcript map are ongoing ( 70 ).
                                                                                                                                                                                                                                               Another hereditary sensory neuropathy (type II), featuring loss of pain sensation and autoamputation, was recently subjected to
                                                                                                                                                                                                                                         genetic analysis in a consanguineous family with two affected sisters ( 71 ). These investigators used exclusion mapping, a mixed linkage/
                                                                                                                                                                                                                                         association strategy, to exclude a variety of known neurotrophin-related genes as candidates for this disorder.
                                                                                                                                                                                                                                               Migraine. An important recent finding of relevance to a more prevalent painful condition, migraine, has been the attribution of
                                                                                                                                                                                                                                         familial hemiplegic migraine and migraine-like episodic ataxia type 2 to mutations in the P/Q-type, calcium channel 1 subunit gene,
                                                                                                                                                                                                                                         CACNL1A4 ( 72 ). Previous linkage studies mapped the gene for these disorders to chromosome 19p13 (e.g., ref. 73 ). By using a
                                                                                                                                                                                                                                         technique called exon trapping, Ophoff et al. ( 72 ) cloned the 47-exon-long CACNL1A4 gene in this region, and identified four different
                                                                                                                                                                                                                                         missense point mutations in affected individuals that segregated with the disease in five families. Although familial hemiplegic migraine
                                                                                                                                                                                                                                         and episodic ataxia type 2 are rare, genetic factors are known to play a role in normal migraine as well, and common allelic variants of
                                                                                                                                                                                                                                         this or other ion channel genes may (or may not, ref. 74 ) contribute to its etiology ( 75 ). Of interest as well is a report of a familial
                                                                                                                                                                                                                                         migraine susceptibility locus on the X chromosome, which may explain the preponderance of this condition in females ( 76 ).
                                                                                                                                                                                                                                               Pathologies linked to human lymphocyte antigens. Other preliminary genetic investigations of painful pathologies with familial
                                                                                                                                                                                                                                         aggregationincluding reflex sympathetic dystrophy/ complex regional pain syndrome ( 77 ), rheumatoid arthritis (e.g., ref. 78 ), and
                                                                                                                                                                                                                                         fibromyalgia (e.g., ref. 79 )have demonstrated at least provisional linkage to or association with various human lymphocyte antigen
                                                                                                                                                                                                                                         regions or antigens. This system does not seem to be linked, however, to familial predisposition to discogenic low-back pain ( 80 ). It is
                                                                                                                                                                                                                                         expected that full-scale genetic investigations of these and other complex pain traits are ongoing or imminent. What are less likely to occur
                                                                                                                                                                                                                                         are investigations of nonpathological pain traits, owing to the added complexity introduced by a continuous phenotype. For genetic
                                                                                                                                                                                                                                         investigation of normal pain sensitivity and sensitivity to analgesia, animal studies provide much-needed statistical power.
                                                                                                                                                                                                                                               Animal Studies. At the present time, three published studies exist (although many more are ongoing in my laboratory and others)
                                                                                                                                                                                                                                         demonstrating linkage of a pain-related trait to chromosomal locations in the mouse. A two-step quantitative trait locus (QTL) mapping
                                                                                                                                                                                                                                         approach has been chosen in each case (see refs. 81 and 82 for a detailed description). First, RI strains of the 26-strain BxD set, developed
                                                                                                                                                                                                                                         by Taylor ( 83 ) from an F2 intercross between B6 and D2 mice, were phenotyped to identify provisional linkages. These linkages were
                                                                                                                                                                                                                                         then independently confirmed or disconfirmed by using new (B6  D2) F2 hybrids.
                                                                                                                                                                                                                                               Morphine analgesia. Belknap and Crabbe ( 84 ) tested BxD RI strains for a number of systemic morphine responses, including
                                                                                                                                                                                                                                         analgesia on the hot-plate test. Of eight broad chromosomal regions provisionally found to be linked with morphine analgesic magnitude,
                                                                                                                                                                                                                                         two have subsequently been confirmed beyond the level of suggestive linkage (P < .0016) as proposed by Lander and Kruglyak ( 85 ):
                                                                                                                                                                                                                                         the Mpmv5 region (020 cM) of mouse chromosome 10 ( 86 ) and the Myo5a region (3050 cM) of mouse chromosome 9 ( 87 ).
                                                                                                                                                                                                                                               The results to date of this ongoing QTL mapping study nicely illustrate the utility of the approach. In the Mpmv5 region lies the Oprm
                                                                                                                                                                                                                                         gene (8 cM) encoding the mouse -opioid receptor type. Oprm is an obvious candidate gene for morphine analgesic magnitude, implicated
                                                                                                                                                                                                                                         via pharmacological (see ref. 88 ) and transgenic ( 89 , 90 ) studies. Allelic variation at this QTL accounts for 2833% of the observed
                                                                                                                                                                                                                                         genetic variability, and F2 mice inheriting two copies of the D2 allele at this QTL exhibit 4-fold more analgesia from a 16 mg/kg dose of
                                                                                                                                                                                                                                         morphine than do F2 mice inheriting two copies of the B6 allele ( 86 ). This QTL has been statistically associated with other opioid traits
                                                                                                                                                                                                                                         as well, including morphine consumption ( 91 ) and whole-brain [3H]naloxone binding ( 86 ).
                                                                                                                                                                                                                                               One may have expected a priori that the gene encoding the -opioid receptor would be associated with morphine analgesic
                                                                                                                                                                                                                                         magnitude. The candidate gene on mouse chromosome 9 is perhaps more heuristic. Within this region lies the Htr1b gene (46 cM),
                                                                                                                                                                                                                                         encoding the serotonin-1B (5-HT1B) receptor subtype (the mouse analog of the human 5-HT1D receptor). Based on the hypothesis that
                                                                                                                                                                                                                                         Htr1b might represent the QTL for morphine analgesia on chromosome 9, we conducted a series of pharmacological experiments that
                                                                                                                                                                                                                                         provided substantial support for the involvement of spinal 5-HT1B receptors ( 87 ). Although data exist indicating a relationship between 5-
                                                                                                                                                                                                                                         HT1B receptors and opioid analgesia (e.g., ref. 92 ), there is still much confusion regarding the specific role of the 5-HT1A versus 5-HT1B
                                                                                                                                                                                                                                         subtypes, and until very recently subtype-specific ligands were decidedly lacking ( 93 ). Thus, it is unlikely that the studies we conducted
                                                                                                                                                                                                                                         would have been conceived of in the absence of the QTL mapping data.
                                                                                                                                                                                                                                               Basal nociceptive sensitivity. By using similar methodology to that described above, we recently mapped basal thermal nociceptive
                                                                                                                                                                                                                                         sensitivity by using the hot-plate test ( 94 ). The most promising of six putative linkages in BxD RI strainsthe D4Mit71 region (5070
                                                                                                                                                                                                                                         cM) of mouse chromosome 4was largely confirmed by using F2 mice. This QTL displayed evidence of sex specificity, with a combined
                                                                                                                                                                                                                                         BxD/F2 P value of 0.005 for males but only 0.085 for females. The identification of a candidate gene in this region, the Oprd1 gene (65
                                                                                                                                                                                                                                         cM) encoding the mouse -opioid receptor type, inspired a simple pharmacological experiment in which B6 and D2 mice of both sexes
                                                                                                                                                                                                                                         were administered ,-, -, and -specific antagonists prior to assessment of hot-plate sensitivity. As predicted by the hypothesis of Oprd1
                                                                                                                                                                                                                                         as a male-specific QTL for this trait, we observed a sex- and strain-dependent pattern of responses, with the -specific antagonist,
                                                                                                                                                                                                                                         naltrindole, lowering nociceptive latencies in the following order of efficacy: D2 male > B6 male > D2 female > B6 female ( 94 ).
the authoritative version for attribution.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
THE GENETIC MEDIATION OF INDIVIDUAL DIFFERENCES IN SENSITIVITY TO PAIN AND ITS INHIBITION 7749
                                                                                                                                                                                                                                              Nonopioid SIA. It has long been known that many forms of analgesia are resistant to antagonism by naloxone, representing the
                                                                                                                                                                                                                                         recruitment of non-opioid mechanisms ( 95 ). To shed light on the mediation of these powerful but little understood systems, we conducted
                                                                                                                                                                                                                                         a QTL mapping experiment of nonopioid SIA resulting from 3-min forced swims in 15C water ( 96 ). Six putative QTLs were identified
                                                                                                                                                                                                                                         in the BxD RI phase, of which four were subsequently disconfirmed by using F2 hybrids. Of the two remaining QTLs, one on
                                                                                                                                                                                                                                         chromosome 8 (5080 cM) was confirmed beyond Lander and Kruglyaks ( 85 ) threshold for significant linkage. This QTL (dubbed
                                                                                                                                                                                                                                         Siafq1) exhibited compelling evidence of sex specificity, reaching a combined BxD/F2 P value of 0.00000012 for females but only 0.038
                                                                                                                                                                                                                                         for males. Female F2 mice inheriting two copies of the D2 allele at this locus displayed 3-fold more SIA than those inheriting two copies
                                                                                                                                                                                                                                         of the B6 allele ( 96 ). This finding of a female-specific QTL for SIA is of special interest because we ( 36 , 97 ) and others (e.g., ref. 98 )
                                                                                                                                                                                                                                         had previously demonstrated the existence of qualitative sex differences in the neurochemical mediation of this trait.
                                                                                                                                                                                                                                                                         SEX-SPECIFIC GENETIC MEDIATION OF PAIN AND ANALGESIA
                                                                                                                                                                                                                                               The two findings of sex-specific QTLs described abovea male-specific QTL for baseline thermal nociceptive sensitivity and a
                                                                                                                                                                                                                                         female-specific QTL for non-opioid SIAexemplify a phenomenon we and others have found repeatedly, i.e., sex/ genotype interactions
                                                                                                                                                                                                                                         of relevance to nociception and its modulation. The discovery of sex-specific QTLs on autosomes, first reported by Melo et al. ( 99 ) for
                                                                                                                                                                                                                                         alcohol preference, was surprising to many, but now more and more examples are being uncovered. It should be emphasized that the
                                                                                                                                                                                                                                         existence of autosomal, sex-specific QTLs does not imply that the sexes possess or express different genes, but rather that different genes
                                                                                                                                                                                                                                         are associated with trait variability in each sex. The existence of sex-specific QTLs does, however, imply that males and females possess
                                                                                                                                                                                                                                         at least partially independent physiological mechanisms underlying the traits in question.
                                                                                                                                                                                                                                               Sex differences in nociception and analgesia are controversial, but when differences are found, males of a number of species
                                                                                                                                                                                                                                         consistently display higher thresholds, tolerance, and analgesic sensitivity (see refs. 100 and 101 for reviews). The inability of some to
                                                                                                                                                                                                                                         observe these sex differences has been attributed to estrus cycle variability, test specificity, and experimental parameters (e.g., ref. 102 ).
                                                                                                                                                                                                                                         Recent data from my laboratory suggest that an important factor contributing to variable results in this literature has been overlooked, i.e.,
                                                                                                                                                                                                                                         genotype of the test subjects. For example, a recent survey of supraspinal morphine analgesia in 11 inbred strains revealed no significant
                                                                                                                                                                                                                                         sex differences in morphine analgesic potency in seven of these strains ( 58 ). In three strains (AKR, B6, and SWR), males exhibited 3.5-
                                                                                                                                                                                                                                         to 7-fold higher sensitivity to intracerebroventricularly administered morphine than their female counterparts. Finally, one strain (CBA)
                                                                                                                                                                                                                                         was identified in which females were 5-fold more sensitive to morphine than males. In another, just completed study specifically
                                                                                                                                                                                                                                         comparing outbred mouse strains, we found that a large male-vs.-female difference in baseline tail-flick latencies can be seen in SW mice
                                                                                                                                                                                                                                         obtained from Simonsen Laboratories (Gilroy, CA), but the analogous sex difference in SW and CD-1 mice from HarlanSpragueDawley
                                                                                                                                                                                                                                         is either absent or too small to detect statistically with n = 1632 (unpublished data). This vendor effect between SW mice from two
                                                                                                                                                                                                                                         different suppliers is likely due to genetic factors, because both populations have been bred in my vivarium for several generations.
                                                                                                                                                                                                                                         Ultimately, then, the failure of some to detect sex differences may simply be due to the fact that in the subject population chosen, there is
                                                                                                                                                                                                                                         no sex difference to detect.
                                                                                                                                                                                                                                               Another intriguing sex/genotype interaction is that demonstrated by Rady and Fujimoto ( 103 ). As described above, these
                                                                                                                                                                                                                                         investigators have determined that heroin analgesia is mediated by -opioid receptors in ICR mice of both sexes but by -opioid receptors
                                                                                                                                                                                                                                         in SW mice of both sexes ( 39 ). An analysis of reciprocal (ICR  SW) F1 hybrid mice revealed that male offspring displayed an ICR-like
                                                                                                                                                                                                                                         phenotype and female offspring displayed a SW-like phenotype. That is, in F1 males, heroin analgesia was blocked by -opioid- but not -
                                                                                                                                                                                                                                         opioid-specific antagonists, whereas the reverse was true for F1 females ( 103 ). This is likely an example of sex-influenced autosomal
                                                                                                                                                                                                                                         dominant inheritance (see ref. 104 ). Further analysis of this phenomenon, if it can be replicated in inbred strains, may help to illuminate
                                                                                                                                                                                                                                         the basis of sex/genotype interactions in analgesia. Also potentially enlightening is our ongoing mapping study of supraspinal morphine
                                                                                                                                                                                                                                         analgesia in (AKR  CBA) F2 mice, focusing specifically on the identification of sex-specific QTLs. Male mice of these two strains
                                                                                                                                                                                                                                         exhibit equipotent analgesic sensitivity to morphine, whereas the of female mice differ by a factor of 35 ( 58 ).
                                                                                                                                                                                                                                                                                  ALLELIC VARIANTS OF PAIN-RELATED GENES
                                                                                                                                                                                                                                               Once the genes mediating a trait have been positively identified, a crucial task still remainsthe identification of alternate alleles of
                                                                                                                                                                                                                                         that gene giving rise to the original phenotypic difference between individuals and/or populations. This effort is again rendered more
                                                                                                                                                                                                                                         difficult when considering complex genetic traits, because the allelic variants are more likely to be single base pair changes (single-
                                                                                                                                                                                                                                         nucleotide polymorphisms) than chromosomal rearrangements or large deletions. Also, whereas the mutations giving rise to disease
                                                                                                                                                                                                                                         phenotypes are likely to occur in the coding region of a gene, allelic variation causing subtle changes in gene expression can occur outside
                                                                                                                                                                                                                                         (even far outside) the coding region. Nonetheless, some success has been reported.
                                                                                                                                                                                                                                               Opioid Receptor Genes. The coding and much of the regulatory and intronic regions of human opioid receptor genes have been
                                                                                                                                                                                                                                         sequenced (e.g., ref. 105 ). By using direct sequencing of hundreds of individuals, three separate investigations identified two common
                                                                                                                                                                                                                                         variants of the OPRM gene coding for the -opioid receptor ( 106  108 ), with allele frequencies estimated to be 6.611%. An A118G
                                                                                                                                                                                                                                         variant (i.e., an A  G substitution in nucleotide 118 of exon 1, resulting in a Asn  Asp change in amino acid residue 40) was found to
                                                                                                                                                                                                                                         be present in a lower proportion of opioid-dependent subjects than controls, whereas the C17T variant was more common in opioid-
                                                                                                                                                                                                                                         dependent subjects ( 106 , 107 ). The A118G variant was found not to be associated with susceptibility to alcohol dependence ( 108 ). An
                                                                                                                                                                                                                                         A118G -opioid receptor constructed by using site-directed mutagenesis and stably transfected into cell lines displayed higher binding
                                                                                                                                                                                                                                         affinity for -endorphin than the more common wild-type receptor ( 106 ).
                                                                                                                                                                                                                                               An allelic variant (T307C) of the OPRD gene encoding the -opioid receptor has also been found ( 109 ). Although the amino acid
                                                                                                                                                                                                                                         sequence remains unchanged by this substitution, the investigators found that heroin addicts were significantly more likely than controls to
                                                                                                                                                                                                                                         possess a CC genotype and less likely to possess a TT genotype. It was concluded that, although by unknown mechanisms, the C allele
                                                                                                                                                                                                                                         predisposes to heroin abuse. The direct relevance of any such opioid receptor variants to pain or analgesic sensitivity is as yet unpublished,
                                                                                                                                                                                                                                         although this work is no doubt underway in several laboratories.
                                                                                                                                                                                                                                               Cytochrome P450. One genetic polymorphism of well documented relevance to pain is of the gene coding for the neuronal
                                                                                                                                                                                                                                         cytochrome P450IID6 (CYP2D6; sparteine/ debrisoquine oxygenase) enzyme (see ref. 110 for review). This enzyme is in fact absent in
                                                                                                                                                                                                                                         710% of Caucasians, who are thus unable to convert codeine to morphine by O-demethylation ( 111 ). Because much evidence indicates
the authoritative version for attribution.
                                                                                                                                                                                                                                         that codeine produces analgesic effects by being biotransformed to morphine, these poor metabolizers will receive minimal therapeutic
                                                                                                                                                                                                                                         benefit from administration of codeine but are generally subject nonetheless to its side effects ( 112 , 113 ). It has been shown as well that
                                                                                                                                                                                                                                         poor metabolizers report increased pain compared with extensive metabolizers in the cold pressor test ( 114 ). An animal model of this
                                                                                                                                                                                                                                         phenomenon exists, with the female Dark Agouti (DA) rat showing a poor metabolizer phenotype ( 115 , 116 ). This well known example
                                                                                                                                                                                                                                         should serve to remind that much individual variability in drug response may be due to polymorphisms related to pharmacokinetics rather
                                                                                                                                                                                                                                         than pharmacodynamics.
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
THE GENETIC MEDIATION OF INDIVIDUAL DIFFERENCES IN SENSITIVITY TO PAIN AND ITS INHIBITION 7750
                                                                                                                                                                                                                                                                                                     FUTURE DIRECTIONS
                                                                                                                                                                                                                                               The construction of high-density genomic maps and the initial priority of the Mouse and Human Genome Projects will greatly
                                                                                                                                                                                                                                         facilitate (and already has) the identification of the 50,000100,000 mammalian genes. Given the high degree of redundancy and
                                                                                                                                                                                                                                         pleiotropy known to exist in biological systems, the determination of which genes participate in which physiological mechanisms will
                                                                                                                                                                                                                                         remain a daunting task, occupying scientists for decades to come. New, high-throughput genomic technologies (e.g., gene chips) may
                                                                                                                                                                                                                                         further accelerate the rate of discovery. It is likely that the focus in Homo sapiens will be on pathology, whereas animal models like the
                                                                                                                                                                                                                                         mouse will continue to be used to investigate more subtle questions involving the normal range of behavior. Although the use of genetic
                                                                                                                                                                                                                                         techniques naturally garners much excitement, it must be borne in mind that even variability in pain pathologies is largely determined by
                                                                                                                                                                                                                                         environmental factors. Thus, investigations into the psychosocial determinants of pain tolerance and pain behaviors must continue
                                                                                                                                                                                                                                         unabated. Nonetheless, knowledge of the genetic bases of pain-related traits may have important scientific and clinical implications,
                                                                                                                                                                                                                                         facilitating both the development of novel analgesic strategies and improved, idiosyncratic treatment of pain using conventional therapies.
                                                                                                                                                                                                                                               The author is supported by National Institutes of Health Grants DA11394 and DE12735.
                                                                                                                                                                                                                                         1. IASP Subcommittee on Toxicology ( 1979 ) Pain 6 , 249 .
                                                                                                                                                                                                                                         2. Beecher, H. K. ( 1959 ) Measurement of Subjective Responses ( Oxford Univ. Press , New York ).
                                                                                                                                                                                                                                         3. Mogil, J. S. & Grisel, J. E. ( 1998 ) Pain 77 , 107128 .
                                                                                                                                                                                                                                         4. Libman, E. ( 1934 ) J. Am. Med. Assoc. 102 , 335341 .
                                                                                                                                                                                                                                         5. Sherman, E. D. ( 1943 ) Can. Med. Assoc. J. 45 , 437441 .
                                                                                                                                                                                                                                         6. Chapman, W. P. & Jones, C. M. ( 1944 ) J. Clin. Invest. 23 , 8191 .
                                                                                                                                                                                                                                         7. Clark, J. W. & Bindra, D. ( 1956 ) Can. J. Psychol. 10 , 6976 .
                                                                                                                                                                                                                                         8. Woodrow, K. M. , Friedman, G. D. , Siegelaub, A. B. & Collen, M. F. ( 1972 ) Psychosom. Med. 34 , 548556 .
                                                                                                                                                                                                                                         9. Wolff, B. B. & Jarvik, M. E. ( 1963 ) Can. J. Psychol. 17 , 3744 .
                                                                                                                                                                                                                                         10. Wolff, B. B. & Jarvik, M. E. ( 1964 ) Am. J. Psychol. 77 , 589599 .
                                                                                                                                                                                                                                         11. Lanier, L. H. ( 1943 ) Science 97 , 4950 .
                                                                                                                                                                                                                                         12. Lasagna, L. & Beecher, H. K. ( 1954 ) J. Am. Med. Assoc. 156 , 230234 .
                                                                                                                                                                                                                                         13. Wolff, B. B. , Kantor, T. G. , Jarvik, M. E. & Laska, E. ( 1965 ) Clin. Pharmacol. Ther. 7 , 224238 .
                                                                                                                                                                                                                                         14. Brennum, J. , Kjeldsen, M. , Jensen, K. & Jensen, T. S. ( 1989 ) Pain 38 , 211217 .
                                                                                                                                                                                                                                         15. Isselee, H. , De Laat, A. , Lesaffre, E. & Lysens, R. ( 1997 ) Eur. J. Oral Sci. 105 , 583587 .
                                                                                                                                                                                                                                         16. Chen, A. C. N. , Dworkin, S. F. , Haug, J. & Gehrig, J. ( 1989 ) Pain 37 , 129141 .
                                                                                                                                                                                                                                         17. Jacobs, J. W. G. , Geenan, R. , van der Heide, A. , Rasker, J. J. & Bijlsma, J. W. J. ( 1995 ) Scand. J. Rheumatol. 24 , 243247 .
                                                                                                                                                                                                                                         18. Chen, A. C. N. , Dworkin, S. F. & Haug, J. ( 1989 ) Pain 37 , 143160 .
                                                                                                                                                                                                                                         19. Galer, B. S. , Coyle, N. , Pasternak, G. W. & Portenoy, R. K. ( 1992 ) Pain 49 , 8791 .
                                                                                                                                                                                                                                         20. Portenoy, K. M. , Foley, K. M. & Inturrisi, C. E. ( 1990 ) Pain 43 , 273286 .
                                                                                                                                                                                                                                         21. Chapman, C. R. , Hill, H. F. , Saeger, L. & Gavrin, J. ( 1990 ) Pain 43 , 4755 .
                                                                                                                                                                                                                                         22. Levine, J. D. , Gordon, N. C , Smith, R. & Fields, H. L. ( 1981 ) Pain 10 , 379389 .
                                                                                                                                                                                                                                         23. Buchsbaum, M. S. , Davis, G. C. & Bunney, W. E., Jr. ( 1977 ) Nature (London) 270 , 620622 .
                                                                                                                                                                                                                                         24. Mogil, J. S. , Sternberg, W. F. , Marek, P. , Sadowski, B. , Belknap, J. K. & Liebeskind, J. C. ( 1996 ) Proc. Natl. Acad. Sci. USA 93 , 30483055 .
                                                                                                                                                                                                                                         25. Bailey, D. W. ( 1971 ) Transplantation 11 , 325327 .
                                                                                                                                                                                                                                         26. Shuster, L. , Webster, G. W. , Yu, G. & Eleftheriou, B. E. ( 1975 ) Psychopharmacologia 42 , 249254 .
                                                                                                                                                                                                                                         27. Panocka, I. , Marek, P. & Sadowski, B. ( 1986 ) Brain Res. 397 , 152155 .
                                                                                                                                                                                                                                         28. Panocka, I. , Marek, P. & Sadowski, B. ( 1986 ) Brain Res. 397 , 156160 .
                                                                                                                                                                                                                                         29. Belknap, J. K. , Haltli, N. R. , Goebel, D. M. & Lam, M. ( 1983 ) Behav. Genet. 13 , 383396 .
                                                                                                                                                                                                                                         30. Devor, M. & Raber, P. ( 1990 ) Pain 42 , 5167 .
                                                                                                                                                                                                                                         31. Ghione, S. ( 1996 ) Hypertension 28 , 494504 .
                                                                                                                                                                                                                                         32. Festing, M. F. W. ( 1996 ) in Genetic Variants and Strains of the Laboratory Mouse , eds. Lyon, M. F. , Rastan, S. & Brown, S. D. M. ( Oxford
                                                                                                                                                                                                                                              Univ. Press , Oxford ), Vol. 3 , pp. 15371576 .
                                                                                                                                                                                                                                         33. Belknap, J. K. & OToole, L. A. ( 1991 ) in The Genetic Basis of Alcohol and Drug Actions , eds. Harris, R. A. & Crabbe, J. C. ( Plenum , New
                                                                                                                                                                                                                                              York ), pp. 225252 .
                                                                                                                                                                                                                                         34. Frischknecht, H.-R. , Siegfried, B. & Waser, P. G. ( 1988 ) Experientia 44 , 473481 .
                                                                                                                                                                                                                                         35. Helmstetter, F. J. & Fanselow, M. S. ( 1987 ) Behav. Neurosci. 101 , 735737 .
                                                                                                                                                                                                                                         36. Mogil, J. S. & Belknap, J. K. ( 1997 ) Pharmacol. Biochem. Behav. 56 , 6166 .
                                                                                                                                                                                                                                         37. Urca, G. , Segev, S. & Sarne, Y. ( 1985 ) Brain Res. 329 , 109116 .
                                                                                                                                                                                                                                         38. Vaccarino, A. L. , Tasker, R. A. R. & Melzack, R. ( 1988 ) Neurosci. Lett. 84 , 103107 .
                                                                                                                                                                                                                                         39. Rady, J. J. , Roerig, S. C. & Fujimoto, J. M. ( 1991 ) J. Pharmacol. Exp. Ther. 256 , 448457 .
                                                                                                                                                                                                                                         40. Fujimoto, J. M. , Rady, J. J. & Elmer, G. I. ( 1998 ) Soc. Neurosci. Abstr. 24 , 770 .
                                                                                                                                                                                                                                         41. Vaccarino, A. L. & Couret, L. C., Jr. ( 1995 ) Pain 63 , 385389 .
                                                                                                                                                                                                                                         42. Lee, D. H. , Chung, K. & Chung, J. M. ( 1997 ) Neuroreport 8 , 34533456 .
                                                                                                                                                                                                                                         43. West, W. L. , Yeomans, D. C. & Proudfit, H. K. ( 1993 ) Brain Res. 626 , 127135 .
                                                                                                                                                                                                                                         44. Clark, F. M. & Proudfit, H. K. ( 1992 ) Brain Res. 591 , 4453 .
                                                                                                                                                                                                                                         45. Bachiocco, V. , Scesi, M. , Morselli, A. M. & Carli, G. ( 1993 ) Clin. J. Pain 9 , 266271 .
                                                                                                                                                                                                                                         46. Edwards, P. W. , Zeichner, A. , Kuczmierczyk, A. R. & Boczkowski, J. ( 1985 ) Pain 21 , 379384 .
                                                                                                                                                                                                                                         47. Violon, A. & Giurgea, D. ( 1984 ) Pain 18 , 199203 .
                                                                                                                                                                                                                                         48. Pollard, C. A. ( 1985 ) Psychol. Rep. 57 , 813814 .
                                                                                                                                                                                                                                         49. Lester, N. , Lefebvre, J. & Keefe, F. ( 1994 ) Clin. J. Pain 10 , 282289 .
                                                                                                                                                                                                                                         50. Martin, N. , Boomsma, D. & Machin, G. ( 1997 ) Nat. Genet. 17 , 387392 .
                                                                                                                                                                                                                                         51. Ziegler, D. K. , Hur, Y. M. , Bouchard, T. J., Jr. , Hassanein, R. S. & Barter, R. ( 1998 ) Headache 38 , 417422 .
                                                                                                                                                                                                                                         52. Honkasalo, M.-L. , Kaprio, J. , Winter, T. , Heikkila, K. , Sillanpaa, M. & Koskenvuo, M. ( 1995 ) Headache 35 , 7078 .
                                                                                                                                                                                                                                         53. Larsson, B. , Bille, B. & Pedersen, N. L. ( 1995 ) Headache 35 , 513519 .
                                                                                                                                                                                                                                         54. Treloar, S. A. , Martin, N. G. & Heath, A. C. ( 1998 ) Behav. Genet. 28 , 107116 .
                                                                                                                                                                                                                                         55. Bengtsson, B. & Thorson, J. ( 1991 ) Acta Genet. Med. Gemmellol. (Roma) 40 , 8390 .
                                                                                                                                                                                                                                         56. Heikkila, J. K. , Koskenvuo, M. , Heliovaara, M. , Kurppa, K. , Riihimaki, H. , Heikkila, K. , Rita, H. & Videman, T. ( 1989 ) Ann. Med. 21 , 393
                                                                                                                                                                                                                                              398 .
the authoritative version for attribution.
THE GENETIC MEDIATION OF INDIVIDUAL DIFFERENCES IN SENSITIVITY TO PAIN AND ITS INHIBITION 7751
                                                                                                                                                                                                                                         68. Indo, Y. , Tsurata, Y. , Karim, M. A. , Ohta, K. , Kawano, T. , Mitsubuchi, H. , Tonoki, H. , Awaya, Y. & Matsuda, I. ( 1996 ) Nat. Genet. 13 , 485488 .
                                                                                                                                                                                                                                         69. Nicholson, G. A. , Dawkins, J. L. , Blair, I. P. , Kennerson, M. L. , Gordon, M. J. , Cherryson, A. K. , Nash, J. & Bananis, T. ( 1996 ) Nat. Genet. 13 , 101
                                                                                                                                                                                                                                            104 .
                                                                                                                                                                                                                                         70. Blair, I. P. , Hulme, D. , Dawkins, J. L. & Nicholson, G. A. ( 1998 ) Genomics 51 , 277281 .
                                                                                                                                                                                                                                         71. Davar, G. , Shalish, C , Blumenfeld, A. & Breakefield, X. O. ( 1996 ) Pain 67 , 1351359 .
                                                                                                                                                                                                                                         72. Ophoff, R. A. , Terwindt, G. M. , Vergouwe, M. N. , van Eijk, R. , Oefner, P. J. , Hoffman, S. M. G. , Lamerdin, J. E. , Mohrenweiser, H. W. , Bulman, D.
                                                                                                                                                                                                                                            E. , Ferrari, M. , et al. ( 1996 ) Cell 87 , 543552 .
                                                                                                                                                                                                                                         73. Joutel, A. , Bousser, M.-G. , Biousse, V. , Labauge, P. , Chabriat, H. , Nibbio, A. , Maciazek, J. , Meyer, B. , Bach, M. A. , Weissenbach, J. , et al. ( 1993 )
                                                                                                                                                                                                                                            Nat. Genet. 5 , 4045 .
                                                                                                                                                                                                                                         74. Baloh, R. W. , Yue, Q. , Jen, J. C. & Nelson, S. F. ( 1998 ) Soc. Neurosci. Abstr. 24 , 1266 .
                                                                                                                                                                                                                                         75. Peroutka, S. J. ( 1998 ) Clin. Neurosci. 5 , 3437 .
                                                                                                                                                                                                                                         76. Nyholt, D. R. , Dawkins, J. L. , Brimage, P. J. , Goadsby, P. J. , Nicholson, G. A. & Griffiths, L. R. ( 1998 ) Hum. Mol. Genet. 7 , 459463 .
                                                                                                                                                                                                                                         77. Mailis, A. & Wade, J. ( 1994 ) Clin. J. Pain 10 , 210217 .
                                                                                                                                                                                                                                         78. Khan, M. A. , Kushner, I. , Ballou, S. P. & Braun, W. E. ( 1979 ) Lancet 1 , 921922 .
                                                                                                                                                                                                                                         79. Burda, C. D. , Cox, F. R. & Osborne, P. ( 1986 ) Clin. Exper. Rheumatol. 4 , 355357 .
                                                                                                                                                                                                                                         80. Postacchini, F. , Lami, R. & Pugliese, O. ( 1988 ) Spine 13 , 14031406 .
                                                                                                                                                                                                                                         81. Gora-Maslak, G. , McClearn, G. E. , Crabbe, J. C. , Phillips, T. J. , Belknap, J. K. & Plomin, R. ( 1991 ) Psychopharmacology 104 , 413424 .
                                                                                                                                                                                                                                         82. Belknap, J. K. , Dubay, C. , Crabbe, J. C. & Buck, K. J. ( 1996 ) in Handbook of Psychiatric Genetics , eds. Blum, K. & Noble, E. P. ( CRC , New York ),
                                                                                                                                                                                                                                            pp. 435453 .
                                                                                                                                                                                                                                         83. Taylor, B. A. ( 1978 ) in Origin of Inbred Mice , ed. Morse, H. C., III ( Academic , New York ), pp. 423428 .
                                                                                                                                                                                                                                         84. Belknap, J. K. & Crabbe, J. C. ( 1992 ) Ann. N.Y. Acad. Sci. 654 , 311323 .
                                                                                                                                                                                                                                         85. Lander, E. S. & Kruglyak, L. ( 1995 ) Nat. Genet. 11 , 241247 .
                                                                                                                                                                                                                                         86. Belknap, J. K. , Mogil, J. S. , Helms, M. L. , Richards, S. P. , OToole, L. A. , Bergeson, S. E. & Buck, K. J. ( 1995 ) Life Sci. 57 , PL117124 .
                                                                                                                                                                                                                                         87. Hain, H. S. , Belknap, J. K. , Hen, R. & Mogil, J. S. ( 1998 ) J. Pharmacol. Exp. Ther. , in press .
                                                                                                                                                                                                                                         88. Pasternak, G. W. ( 1993 ) Clin. Neuropharmacol. 16 , 118 .
                                                                                                                                                                                                                                         89. Matthes, H. W. D. , Maldonado, R. , Simonin, F. , Valverde, O. , Slowe, S. , Kitchen, I. , Befort, K. , Dierich, A. , Le Meur, M. , Dolle, P. , et al. ( 1996 )
                                                                                                                                                                                                                                            Nature (London) 383 , 819823 .
                                                                                                                                                                                                                                         90. Sora, I. , Takahashi, N. , Funada, M. , Ujike, H. , Revay, R. S. , Donovan, D. M. , Miner, L. L. & Uhl, G. R. ( 1997 ) Proc. Natl. Acad. Sci. USA 94 , 15441549
                                                                                                                                                                                                                                                  .
                                                                                                                                                                                                                                         91. Berrettini,  W. H. , Ferraro, T. N. , Alexander, R. C. , Buchberg, A. M. & Vogel, W. H. ( 1994 ) Nat. Genet. 7 , 5458 .
                                                                                                                                                                                                                                         92. Alhaider, A. A. & Wilcox, G. L. ( 1993 ) J. Pharmacol. Exp. Ther. 265 , 378385 .
                                                                                                                                                                                                                                         93. Glennon, R. A. & Dukat, M. ( 1991 ) Pharmacol. Biochem. Behav. 40 , 10091017 .
                                                                                                                                                                                                                                         94. Mogil, J. S. , Richards, S. P. , O Toole, L. A. , Helms, M. L. , Mitchell, S. R. & Belknap, J. K. ( 1997 ) Pain 70 , 267277 .
                                                                                                                                                                                                                                         95. Lewis, J. W. , Cannon, J. T. & Liebeskind, J. C. ( 1980 ) Science 208 , 623625 .
                                                                                                                                                                                                                                         96. Mogil, J. S. , Richards, S. P. , OToole, L. A. , Helms, M. L. , Mitchell, S. R. , Kest, B. & Belknap, J. K. ( 1997 ) J. Neurosci. 17 , 79958002 .
                                                                                                                                                                                                                                         97. Mogil, J. S. , Sternberg, W. F. , Kest, B. , Marek, P. & Liebeskind, J. C. ( 1993 ) Pain 253 , 1725 .
                                                                                                                                                                                                                                         98. Kavaliers, M. & Choleris, E. ( 1997 ) Brain Res. 768 , 3036 .
                                                                                                                                                                                                                                         99. Melo, J. A. , Shendure, J. , Pociask, K. & Silver, L. M. ( 1996 ) Nat. Genet. 13 , 147153 .
                                                                                                                                                                                                                                         100. Berkley, K. J. ( 1997 ) Behav. Brain Sci. 20 , 371380 .
                                                                                                                                                                                                                                         101. Kest, B. , Sarton, E. , Mogil, J. S. & Dahan, A. ( 1998 ) in Physiology and Pharmacology of Cardio-respiratory Control , eds. Dahan, A. , Teppema, L. J. &
                                                                                                                                                                                                                                            van Beek, E. J. ( Kluwer, Dordrecht , The Netherlands ), pp. 7582 .
                                                                                                                                                                                                                                         102. Bartok, R. E. & Craft, R. M. ( 1997 ) J. Pharmacol. Exp. Ther. 282 , 769778 .
                                                                                                                                                                                                                                         103. Rady, J. J. & Fujimoto, J. M. ( 1997 ) Pharmacogenetics 7 , 429433 .
                                                                                                                                                                                                                                         104. McClearn, G. E. ( 1995 ) Recent Dev. Alcohol. 12 , 217221 .
                                                                                                                                                                                                                                         105. Wendel, B. & Hoehe, M. R. ( 1998 ) J. Mol. Med. 76 , 525532 .
                                                                                                                                                                                                                                         106. Bond, C. , LaForge, K. S. , Tian, M. , Melia, D. , Zhang, S. , Borg, L. , Gong, J. , Schluger, J. , Strong, J. A. , Leal, S. M. , et al. ( 1998 ) Proc. Natl. Acad.
                                                                                                                                                                                                                                            Sci. USA 95 , 96089613 .
                                                                                                                                                                                                                                         107. Berrettini, W. H. , Hoehe, M. R. , Ferrada, T. N. & Gottheil, E. ( 1997 ) Addict. Biol. 2 , 303308 .
                                                                                                                                                                                                                                         108. Bergen, A. W. , Kokoszka, J. , Peterson, R. , Long, J. C. , Virkkunen, M. , Linnoila, M. & Goldman, D. ( 1997 ) Mol. Psychiatry 2 , 490494 .
                                                                                                                                                                                                                                         109. Mayer, P. , Rochlitz, H. , Rauch, E. , Rommelspacher, H. , Hasse, H. E. , Schmidt, S. & Hollt, V. ( 1997 ) NeuroReport 8 , 25472550 .
                                                                                                                                                                                                                                         110. Meyer, U. A. ( 1994 ) J. Pharm . Pharmacol. 46 , 409415 .
                                                                                                                                                                                                                                         111. Alvan, G. , Bechtel, P. , Iselius, L. & Gundert-Remy, U. ( 1990 ) Eur. J. Clin. Pharmacol. 39 , 533537 .
                                                                                                                                                                                                                                         112. Caraco, Y. , Sheller, J. & Wood, A. J. J. ( 1996 ) J. Pharmacol. Exp. Ther. 278 , 11651174 .
                                                                                                                                                                                                                                         113. Poulsen, L. , Brosen, K. , Arendt-Nielsen, L. , Gram, L. F. , Elbaek, K. & Sindrup, S. H. ( 1996 ) Eur. J. Clin. Pharmacol. 51 , 289295 .
                                                                                                                                                                                                                                         114. Sindrup, S. H. , Poulsen, L. , Brosen, K. , Arendt-Nielsen, L. & Gram, L. F. ( 1993 ) Pain 53 , 335349 .
                                                                                                                                                                                                                                         115. Mikus, G. , Somogyi, A. A. , Bochner, F. & Eichelbaum, M. ( 1991 ) Biochem. Pharmacol. 41 , 757762 .
                                                                                                                                                                                                                                         116. Cleary, J. , Mikus, G. , Somogyi, A. A. & Bochner, F. ( 1994 ) J. Pharmacol. Exp. Ther. 271 , 15281534 .
                                                                                                                                                                                                                                         117. Sudakov, S. K. , Goldberg, S. R. , Borisova, E. V. , Surkova, L. A. , Turina, I. V. , Rusakov, D. J. & Elmer, G. I. ( 1993 ) Psychopharmacology 112 , 183188 .
                                                                                                                                                                                                                                         118. Woolfolk, D. R. & Holtzman, S. G. ( 1995 ) Pharmacol. Biochem. Behav. 51 , 699703 .
                                                                                                                                                                                                                                         119. Stohr, T. , Wermeling, D. S. , Szuran, T. , Pliska, V. , Domeney, A. , Welzl, H. , Weiner, I. & Feldon, J. ( 1998 ) Pharmacol. Biochem. Behav. 59 , 799805 .
                                                                                                                                                                                                                                         120. Tilson, H. A. & Rech, R. H. ( 1974 ) Psychopharmacologia 35 , 4560 .
                                                                                                                                                                                                                                         121. Gschossmann, J. M. , Cheng, S. H. , Martinez, V. , Tache, Y. & Mayer, E. A. ( 1996 ) Digest. Dis. Sci. J. , in press.
                                                                                                                                                                                                                                         122. Rosenthale, M. E. ( 1970 ) Arch. Int. Pharmacodyn. Ther. 188 , 1422 .
                                                                                                                                                                                                                                         123. Zidek, Z. & Perlik, F. ( 1971 ) J. Pharm. Pharmacol. 23 , 389340 .
                                                                                                                                                                                                                                         124. Wiesenfeld, Z. & Hallin, R. G. ( 1981 ) Physiol. Behav. 27 , 735740 .
                                                                                                                                                                                                                                         125. Seltzer, Z. & Shir, Y. ( 1991 ) J. Basic Clin . Physiol. Pharmacol. 2 , 1761 .
                                                                                                                                                                                                                                         126. Carr, M. M. , Best, T. J. , Mackinnon, S. E. & Evans, P. L ( 1992 ) Ann. Plast. Surg. 28 , 538544 .
                                                                                                                                                                                                                                         127. Kasson, B. G. & George, R. ( 1984 ) Life Sci. 34 , 16271634 .
                                                                                                                                                                                                                                         128. Hoffmann, O. , Plesan, A. & Wiesenfeld-Hallin, Z. ( 1998 ) Brain Res. 806 , 232237 .
                                                                                                                                                                                                                                         129. Kunos, G. , Mosqueda-Garcia, R. , Mastrianni, J. A. & Abbott, F. V. ( 1987 ) Can. J. Physiol. Pharmacol. 65 , 16241632 .
                                                                                                                                                                                                                                         130. Tiurina, I. V. , Rusakov, D. I. & Sudakov, S. K. ( 1995 ) Eksp. Klin. Farmakol. 58 , 2224 .
                                                                                                                                                                                                                                         131. Borisova, E. V. & Sudakov, S. K. ( 1995 ) Zh. Vyssh. Nerv. Deiat. Im. I. P. Pavlova 45 , 11741181 .
                                                                                                                                                                                                                                         132. Meller, S. T. , Lewis, S. J. , Brody, M. J. & Gebhart, G. F. ( 1992 ) Brain Res. 587 , 8894 .
                                                                                                                                                                                                                                         133. Rosecrans, J. A. , Robinson, S. E. , Johnson, J. H. , Mokler, D. J. & Hong, J.-S. ( 1986 ) Brain Res. 382 , 7180 .
                                                                                                                                                                                                                                         134. Zhang, L.-X. , Li, X.-L. , Wang, L. & Han, J.-S. ( 1997 ) Brain Res. 745 , 158164 .
                                                                                                                                                                                                                                         135. Eidelberg, E. , Erspamer, R. , Kreinick, C. J. & Harris, J. ( 1975 ) Eur. J. Pharmacol. 32 , 329336 .
                                                                                                                                                                                                                                         136. Ramabadran, K. , Michaud, G. & Jacob, J. J. C. ( 1982 ) Ind. J. Exp. Biol. 20 , 7476 .
the authoritative version for attribution.
                                                                                                                                                                                                                                             This paper was presented at the National Academy of Sciences colloquium The Neurobiology of Pain, held December 1113,
                                                                                                                                                                                                                                         1998, at the Arnold and Mabel Beckman Center in Irvine, CA.
                                                                                                                                                                                                                                         sponses and in their modulation by opiates, and (iii) that allelic variants at the OR locus are strong candidates for contributing to these
                                                                                                                                                                                                                                         differences in mice and attractive candidates for producing such effects in humans.
                                                                                                                                                                                                                                               Humans differ in their individual responses to pain and to opiate drugs. Recent studies of twins document that individual differences
                                                                                                                                                                                                                                         in several types of pain are likely to have substantial genetic determinants. Genetic components to susceptibility to migraine pain are
                                                                                                                                                                                                                                         documented in studies of thousands of twin pairs, although family studies document substantial genetic heterogeneity in this disorder ( 16
                                                                                                                                                                                                                                          18 ). Studies of concordance for self-reported menstrual pain also identify substantial genetic components ( 19 ). Interestingly, the
                                                                                                                                                                                                                                         heritabilities documented in these human studies (0.5) fit nicely with those identified in murine strain-comparison and quantitative trait
                                                                                                                                                                                                                                         locus studies ( 20 , 21 ).
                                                                                                                                                                                                                                               Humans also differ from one another in OR densities. Binding studies to postmortem brain samples and in vivo positron-emission
                                                                                                                                                                                                                                         tomography radioligand analyses both suggest 3050% or even larger ranges of individual human differences in OR densities. For
                                                                                                                                                                                                                                         example, Pfeiffer et al. ( 22 ) reported that median OR binding in human frontal cortex was 2.3 pmol/g (SD = 0.52). Frost and coworkers
                                                                                                                                                                                                                                         ( 23 , 24 ) noted that a measure of thalamic OR binding with [11C]carfentanil was 3.8 pmol/g (SD = 1.4). If individuals in the upper third
                                                                                                                                                                                                                                         of the population are characterized by these data, they should express >45% ( 22 ) or >74% ( 23 , 24 ) more ORs than the individuals in
                                                                                                                                                                                                                                         the lower third of the population. Mouse studies document that genetic differences of this magnitude in OR expression can influence both
                                                                                                                                                                                                                                         baseline nociception and morphine responses, as noted above. Elucidation of the genetic bases for these differences in receptor expression
                                                                                                                                                                                                                                         would thus represent a substantial advance in our understanding of individual differences in nociceptive behaviors and drug responses.
                                                                                                                                                                                                                                               Levels of expression of many, if not most, human genes differ from individual to individual. Many of these differences are thought to
                                                                                                                                                                                                                                         be based on differences in the cis-acting DNA sequences that normally act to provide cell-type-specific, appropriately regulated gene
                                                                                                                                                                                                                                         expression ( 25 ). Many of these DNA promoter and enhancer sequences are typically found in the 5 ends of genes and often serve as
                                                                                                                                                                                                                                         recognition sites for regulatory DNA binding proteins.
                                                                                                                                                                                                                                               Searches for the functional polymorphisms that contribute to these individual differences in gene expression can involve several
                                                                                                                                                                                                                                         steps. Cloning appropriate genomic sequences and characterizing the site(s) for transcriptional initiation so that 5 flanking and other
                                                                                                                                                                                                                                         potential regulatory regions can be determined with confidence represents an important initial step. Identifying polymorphisms in these
                                                                                                                                                                                                                                         regions provides a second series of challenges ( 26 ). Seeking relationships between these polymorphisms and differences in levels of gene
                                                                                                                                                                                                                                         expression is a third step. We can then ask whether the identified polymorphic sequences predict differences not only in levels of OR
                                                                                                                                                                                                                                         expression but also in opiate responses.
                                                                                                                                                                                                                                               The information currently available in GenBank describes 2 kilobases (kb) of murine and 0.2 kb of human OR genomic sequences
                                                                                                                                                                                                                                         5 to the OR translational start site ( Fig. 1 ). Studies of rapid amplification of cDNA 5 ends have suggested to other workers that two
                                                                                                                                                                                                                                         nearby regions provide the sites at which primer extension products terminate, which are thus potential transcriptional initiation sites
                                                                                                                                                                                                                                         (793 and 268 bp from the translational start site; refs. 27 and 28 ). Sequences from each of these two regions can support some
                                                                                                                                                                                                                                         expression of reporter genes in heterologous cell-expression systems. These sequences can even have enhanced expression in the SHY5Y
                                                                                                                                                                                                                                         cells that normally express OR at modest levels.
                                                                                                                                                                                                                                               None of the reported primer extension products have the modified bases characteristic of mRNA capping, however. None of these
                                                                                                                                                                                                                                         sequences provide the 5 untranslated-region length characteristic of most long mRNAs with relatively short coding sequences (see
                                                                                                                                                                                                                                         below). These 268-bp and 793-bp sequences might thus serve as true promoter/enhancer regions. If so, then human polymorphisms in
                                                                                                                                                                                                                                         these regions should be sought out, as only a moderate number have been reported thus far.
                                                                                                                                                                                                                                               Initial searches in our laboratory, as well as more extensive work by Goldman and coworkers ( 29 ) and by L. Yu (personal
                                                                                                                                                                                                                                         communication), have failed to identify common human OR protein coding-sequence variants that dramatically change the receptors
                                                                                                                                                                                                                                         function, although a modest alteration in affinity for the opioid peptide -endorphin has been noted by Yu and coworkers ( 30 ).
                                                                                                                                                                                                                                               These data are in accord with studies that document no convincing individual differences in OR affinities among humans. The data
                                                                                                                                                                                                                                         also fit with the substantial OR coding sequence conservation among species ( 2 , 4 , 22 ). Such information suggests that genetic
                                                                                                                                                                                                                                         components may be unlikely to provide commonly encountered individual differences through functionally different OR protein
                                                                                                                                                                                                                                         sequences. The information contrasts with the abundant data, noted above, documenting frequent individual differences in levels of OR
                                                                                                                                                                                                                                         expression.
                                                                                                                                                                                                                                               Studies by Ko et al. ( 27 ) and by Liang and Carr ( 28 ) indicate that searches for possible promoter-region sequences must include
                                                                                                                                                                                                                                         the sequences located between 268 and 793 bp 5 to the translational initiation sites tentatively identified by these workers. However,
                                                                                                                                                                                                                                         recently, we have also developed interesting results from comparisons of murine and human 5 flanking sequences. These data could also
                                                                                                                                                                                                                                         suggest other sites at which to seek potential promoter-region polymorphisms in humans. Scatterplot comparisons of these species OR
                                                                                                                                                                                                                                         sequences clearly show the area of high cross-species conservation at the 268/793-bp region identified by Ko et al. ( 27 ) and by Liang
                                                                                                                                                                                                                                         and Carr ( 28 ). These analyses also find another highly conserved region that seems to extend from 2,500 to 4,500 bp 5 to the
                                                                                                                                                                                                                                         translational start ( Fig. 2 ). Conceivably,
                                                                                                                                                                                                                                           FIG. 1. Human  opioid gene structure. Exons (Exn) are indicated by boxes. The codon (ATG) and stop codon (TAA for OR and
                                                                                                                                                                                                                                           TAG for the less abundant variant XDR1A) are indicated, as are Alu repeats. (GT)n, dinucleotide repeat; (GTT)n, trinucleotide
                                                                                                                                                                                                                                           repeat; SSR, short simple repeats; SNP, sample single nucleotide polymorphisms (54 G/T; 17 C/T; 118 A/G; 440 C/G; 12 C/G; 912
                                                                                                                                                                                                                                           CG/GC); dashed line, sequence to be elucidated.
the authoritative version for attribution.
                                                                                                                                                                                                                                         THE  OPIATE RECEPTOR AS A CANDIDATE GENE FOR PAIN: POLYMORPHISMS, VARIATIONS IN EXPRESSION,                                             7754
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                         each of these regions could represent unusually highly conserved promoter/enhancer sequences. Polymorphisms from these two conserved
                                                                                                                                                                                                                                         regions would thus be likely candidates for marking functional, level-of-expression allelic OR variants. Alternatively, these highly
                                                                                                                                                                                                                                         conserved regions could also reflect additional exon sequences. Under this scenario, even more 5 sequences would suggest more
                                                                                                                                                                                                                                         prominent candidates for contributing to OR regulation.
                                                                                                                                                                                                                                           FIG. 2. Scatterplot of the nucleotide sequence comparisons between the mouse (x axis) and human (y axis) -OR 5 flanking
                                                                                                                                                                                                                                           sequences, with the translational initiation site at the upper right. Oblique lines represent regions of sequence conservation as
                                                                                                                                                                                                                                           described in the text.
                                                                                                                                                                                                                                               We and others have also identified a number of interesting OR gene polymorphic markers. We identified a human polymorphism:
                                                                                                                                                                                                                                         an MspI restriction fragment length polymorphism ( 2 ). We used PCR to amplify the DNA and have sequenced >1 kb of DNA containing
                                                                                                                                                                                                                                         the 268/793-bp region from 12 unrelated human individuals (volunteers who gave informed consent for studies conducted for the
                                                                                                                                                                                                                                         Intramural Research Program of the National Institute on Drug Abuse in Baltimore; Z.W. and G.R.U., unpublished observations).
                                                                                                                                                                                                                                         Sequence comparisons identify repetitive sequences in these regions that were not polymorphic in initial screens. Two other types of
                                                                                                                                                                                                                                         sequence variation have been identified. More than 20 single nucleotide polymorphisms have been identified in these sequences (see ref.
                                                                                                                                                                                                                                         31 ). We are working to confirm these sequences and will study their frequencies in larger samples to establish their utility for correlations
                                                                                                                                                                                                                                         with receptor-expression densities and opiate-drug responses. Altering 5 untranslated mRNA sequences could readily explain different
                                                                                                                                                                                                                                         levels of OR mRNA stability or even translational efficacy and could contribute to the expression of differing levels of this protein in
                                                                                                                                                                                                                                         different individuals or cell types.
                                                                                                                                                                                                                                               We also have identified a polymorphic repetitive element in >8 kb of murine 5 flanking sequence (I.S. and G.R.U., unpublished
                                                                                                                                                                                                                                         observations). This murine polymorphism lies 2 kb 5 to the translational start site, close to sequences recently identified as candidate OR
                                                                                                                                                                                                                                         promoter/enhancer elements (see below).
                                                                                                                                                                                                                                               Because data from comparisons of more 5 human and mouse OR genomic sequences also suggest the presence of additional exon
                                                                                                                                                                                                                                         (s) and more 5 sites for transcriptional initiation or highly conserved regulatory regions, searches in more 5 genomic regions also make
                                                                                                                                                                                                                                         sense. Workers are currently undertaking approaches consisting of cloning additional 5 genomic sequence, searching for simple sequence
                                                                                                                                                                                                                                         repeat and single nucleotide polymorphisms, characterizing the individual differences in these polymorphic sites, and applying these
                                                                                                                                                                                                                                         polymorphisms to seek correlations with OR expression levels and nociceptive responses. Most genes promoters have much of their
                                                                                                                                                                                                                                         functional anatomy within several thousand base pairs of their transcriptional initiation sites. However, other genes have promoter regions
                                                                                                                                                                                                                                         that extend for >10 or even >20 kb. Analyses of further OR genomic 5 flanking sequences could make great contributions to
                                                                                                                                                                                                                                         understanding this gene and would be quite likely to identify many of its important regulatory elements.
                                                                                                                                                                                                                                               Information about OR gene polymorphisms that can predict the likelihood of high or low levels of  expression in an individual
                                                                                                                                                                                                                                         could allow drug treatments to be individualized. These data could aid in selecting analgesic agents and in optimizing dose ranges. They
                                                                                                                                                                                                                                         could thus improve pain management for individuals with acute or long-term pain problems. These data could suggest new therapeutic
                                                                                                                                                                                                                                         specificities and efficacies to even this well established opiate drug class that remains a major weapon for amelioration of pain states.
the authoritative version for attribution.
                                                                                                                                                                                                                                         THE  OPIATE RECEPTOR AS A CANDIDATE GENE FOR PAIN: POLYMORPHISMS, VARIATIONS IN EXPRESSION,                                                              7755
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
                                                                                                                                                                                                                                              Thus, the extensive work required to identify such markers should be worthwhile.
                                                                                                                                                                                                                                         1. Wang, J. B. , Imai, Y. , Eppler, C. M , Gregor, P. , Spivak, C. E. & Uhl, G. R. ( 1993 ) Proc. Natl. Acad. Sci. USA 90 , 1023010234 .
                                                                                                                                                                                                                                         2. Wang, J. B. , Johnson, P. S. , Persico, A. M. , Hawkins, A. L. , Griffin, C. A. & Uhl, G. R. ( 1994 ) FEBS Lett. 338 , 217222 .
                                                                                                                                                                                                                                         3. Kaufman, D. L. , Keith, D. J. , Anton, B. , Tian, J. , Magendzo, K. , Newman, D. , Tran, T. H. , Lee, D. S. , Wen, C. , Xia, Y. R. , et al. ( 1995 ) J.
                                                                                                                                                                                                                                              Biol. Chem. 270 , 1587715883 .
                                                                                                                                                                                                                                         4. Sora, L , Takahashi, N. , Funada, M. , Ujike, H. , Revay, R. , Donovan, D. , Miner, L. & Uhl, G. R. ( 1997 ) Proc. Natl. Acad. Sci. USA 94 , 1544
                                                                                                                                                                                                                                              1549 .
                                                                                                                                                                                                                                         5. Matthes, H. W. , Maldonado, R. , Simonin, F. , Valverde, O. , Slowe, S. , Kitchen, I. , Befort, K. , Dierich, A. , Le, M. M. , Dolle, P. , et al. ( 1996 )
                                                                                                                                                                                                                                              Nature (London) 383 , 819823 .
                                                                                                                                                                                                                                         6. Sora, I. , Funada, M. & Uhl, G. R. ( 1997 ) Eur. J. Pharmacol. 324 , R1R2 .
                                                                                                                                                                                                                                         7. Sora, I. , Li, X. F. , Funada, M. , Kinsey, S. & Uhl, G. R. ( 1999 ) Eur. J. Pharmacol 366 , R3R5 .
                                                                                                                                                                                                                                         8. Baran, A. , Shuster, L. , Eleftheriou, B. E. & Bailey, D. W. ( 1975 ) Life Sci. 17 , 633640 .
                                                                                                                                                                                                                                         9. Moskowitz, A. S. , Terman, G. W. , Carter, K. R. , Morgan, M. J. & Liebeskind, J. C. ( 1985 ) Brain Res. 361 , 4651 .
                                                                                                                                                                                                                                         10. Vaught, J. L. , Mathiasen, J. R. & Raffa, R. B. ( 1988 ) J. Pharmacol. Exp. Ther. 245 , 1316 .
                                                                                                                                                                                                                                         11. Brase, D. A. , Loh, H. H. & Way, E. L. ( 1977 ) J. Pharmacol. Exp. Ther. 201 , 368374 .
                                                                                                                                                                                                                                         12. Lander, E. S. & Botstein, D. ( 1989 ) Genetics 121 , 185199 .
                                                                                                                                                                                                                                         13. Berrettini, W. H. , Alexander, R. , Ferraro, T. N. & Vogel, W. H. ( 1994 ) Psychiatr. Genet. 4 , 8186 .
                                                                                                                                                                                                                                         14. Berrettini, W. H. , Ferraro, T. N. , Alexander, R. C. , Buchberg, A. M. & Vogel, W. H. ( 1994 ) Nat. Genet. 7 , 5458 .
                                                                                                                                                                                                                                         15. Belknap, J. K. , Mogil, J. S. , Helms, M. L. , Richards, S. P. , O Toole, L. A. , Bergeson, S. E. & Buck, K. J. ( 1995 ) Life Sci. 57 , PL117PL128 .
                                                                                                                                                                                                                                         16. Nyholt, D. R. , Lea, R. A. , Goadsby, P. J. , Brimage, P. J. & Griffiths, L. R. ( 1998 ) Neurology 50 , 14281432 .
                                                                                                                                                                                                                                         17. Peroutka, S. J. ( 1998 ) Clin. Neurosci. 5 , 3437 .
                                                                                                                                                                                                                                         18. Ziegler, D. K. , Hur, Y. M. , Bouchard, T. J. , Hassanein, R. S. & Barter, R. ( 1998 ) Headache 38 , 417422 .
                                                                                                                                                                                                                                         19. Treloar, S. A. , Martin, N. G. & Heath, A. C. ( 1998 ) Behav. Genet. 28 , 107116 .
                                                                                                                                                                                                                                         20. Mogil, J. S. , Kest, B. , Sadowski, B. & Belknap, J. K. ( 1996 ) J. Pharmacol. Exp. Ther. 276 , 532544 .
                                                                                                                                                                                                                                         21. Mogil, J. S. , Richards, S. P. , OToole, L. A. , Helms, M. L. , Mitchell, S. R. , Kest, B. & Belknap, J. K. ( 1997 ) J. Neurosci. 17 , 79958002 .
                                                                                                                                                                                                                                         22. Pfeiffer, A. , Pasi, A. , Mehraein, P. & Herz, A. ( 1982 ) Brain Res. 248 , 8796 .
                                                                                                                                                                                                                                         23. Frost, J. J. , Mayberg, H. S. , Fisher, R. S. , Douglass, K. H. , Dannals, R. F. , Links, J. M. , Wilson, A. A. , Ravert, H. T. , Rosenbaum, A. E. &
                                                                                                                                                                                                                                              Snyder, S. H. ( 1988 ) Ann. Neurol. 23 , 231237 .
                                                                                                                                                                                                                                         24. Frost, J. J. , Douglass, K. H. , Mayberg, H. S. , Dannals, R. F. , Links, J. M. , Wilson, A. A. , Ravert, H. T. , Crozier, W. C. & Wagner, H. J. ( 1989 )
                                                                                                                                                                                                                                              J. Cereb. Blood Flow Metab. 9 , 398409 .
                                                                                                                                                                                                                                         25. Uhl, G. R. , Gold, L. H. & Risch, N. ( 1997 ) Proc. Natl. Acad. Sci. USA 94 , 27852786 .
                                                                                                                                                                                                                                         26. Collins, F. S. , Guyer, M. S. & Charkravarti, A. ( 1997 ) Science 278 , 15801581 .
                                                                                                                                                                                                                                         27. Ko, J. L. , Minnerath, S. R. & Loh, H. H. ( 1997 ) Biochem. Biophys. Res. Commun. 234 , 351357 .
                                                                                                                                                                                                                                         28. Liang, Y. & Carr, L. G. ( 1997 ) Brain Res. 769 , 372374 .
                                                                                                                                                                                                                                         29. Bergen, A. W. , Kokoszka, J. , Peterson, R. , Long, J. C. , Virkkunen, M. , Linnoila, M. & Goldman, D. ( 1997 ) Mol. Psychiatry 2 , 490494 .
                                                                                                                                                                                                                                         30. Bond, C , LaForge, K. S. , Tian, M. , Melia, D. , Zhang, S. , Borg, L. , Gong, J. , Schluger, J. , Strong, J. A. , Leal, S. M. , et al. ( 1998 ) Proc. Natl.
                                                                                                                                                                                                                                              Acad. Sci. USA 95 , 96089613 .
                                                                                                                                                                                                                                         31. Wendel, B. & Hoehe, M. R. ( 1998 ) J. Mol. Med. 76 , 525532 .
the authoritative version for attribution.
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
the authoritative version for attribution.
                                                                                                                                                                                                                                  NOCICEPTION, AND OPIATE RESPONSES
                                                                                                                                                                                                                                  THE  OPIATE RECEPTOR AS A CANDIDATE GENE FOR PAIN: POLYMORPHISMS, VARIATIONS IN EXPRESSION,
                                                                                                                                                                                                                                                                                      7756
lengths, word breaks, heading styles, and other typesetting-specific formatting, however, cannot be retained, and some typographic errors may have been accidentally inserted. Please use the print version of this publication as
About this PDF file: This new digital representation of the original work has been recomposed from XML files created from the original paper book, not from the original typesetting files. Page breaks are true to the original; line
NAS COLLOQUIA
                                                                                                                                                                                                                                                                                                   SCHEDULED
                                                                                                                                                                                                                                            Virulence and Defense in Host-Pathogen Interactions: Common Features between Plants and Animals
                                                                                                                                                                                                                                            December 9-11, 1999
                                                                                                                                                                                                                                            Organizers: Jim Cook, Noel T. Keen,
                                                                                                                                                                                                                                            Brian J. Staskawicz, John J. Meckalanos, and Frederick M. Ausubel
                                                                                                                                                                                                                                            Incentives To Promote Human Capital
                                                                                                                                                                                                                                            December 17-19, 1999; Irvine, California
                                                                                                                                                                                                                                            Organizer: James Heckman
                                                                                                                                                                                                                                            Variation and Evolution in Plants and Microorganisms; Fifty Years after Stebbins
                                                                                                                                                                                                                                            January 27-29, 2000; Irvine, California
                                                                                                                                                                                                                                            Organizers: Francisco Ayala and Walter Fitch
                                                                                                                                                                                                                                            The Biotic Crisis and the Future of Evolution
                                                                                                                                                                                                                                            March 16-19, 2000; Irvine, California
                                                                                                                                                                                                                                            Organizers: Norman Myers, Andrew Knoll
                                                                                                                                                                                                                                            Auditory Neuroscience: Development, Transduction, and Integration
                                                                                                                                                                                                                                            May 19-21, 2000; Irvine, California
                                                                                                                                                                                                                                            Organizers: James Hudspeth and Mark Konishi
                                                                                                                                                                                                                                            Links between Recombination and Replication
                                                                                                                                                                                                                                            November 10-12, 2000; Irvine, California
                                                                                                                                                                                                                                            Organizer: Charles Radding
                                                                                                                                                                                                                                                                                                  COMPLETED
                                                                                                                                                                                                                                            Industrial Ecology
                                                                                                                                                                                                                                            May 20-21, 1991; Washington, D.C.
                                                                                                                                                                                                                                            Organizer: C. Kumar N. Patel
                                                                                                                                                                                                                                            Proceedings: February 4, 1992
                                                                                                                                                                                                                                            Images of Science: Science of Images
                                                                                                                                                                                                                                            January 13-14, 1992; Washington, D.C.
                                                                                                                                                                                                                                            Organizer: Albert Crewe
                                                                                                                                                                                                                                            Proceedings: November 3, 1993
                                                                                                                                                                                                                                            Physical Cosmology
                                                                                                                                                                                                                                            March 27-29, 1992; Irvine, California
                                                                                                                                                                                                                                            Organizer: David Schramm
                                                                                                                                                                                                                                            Proceedings: June 3, 1993
                                                                                                                                                                                                                                            Molecular Recognition
                                                                                                                                                                                                                                            September 10-11, 1992; Washington, D.C.
                                                                                                                                                                                                                                            Organizer: Ronald Breslow
                                                                                                                                                                                                                                            Proceedings: February 16, 1993
                                                                                                                                                                                                                                            Human-Machine Communication by Voice
                                                                                                                                                                                                                                            February 8-9, 1993; Irvine, California
                                                                                                                                                                                                                                            Organizer: Lawrence Rabiner
                                                                                                                                                                                                                                            Proceedings: October 24, 1995
                                                                                                                                                                                                                                            Voice Communication Between Humans and Machines:
                                                                                                                                                                                                                                            August 1994
                                                                                                                                                                                                                                            Changing Human Ecology and Behavior: Effects on Infectious Diseases
                                                                                                                                                                                                                                            September 27-28, 1993; Washington, D.C.
                                                                                                                                                                                                                                            Organizer: Bernard Roizman
                                                                                                                                                                                                                                            Proceedings: March 29, 1994
                                                                                                                                                                                                                                            Infectious Diseases in an Age of Change: January 1995
                                                                                                                                                                                                                                            The Tempo and Mode of Evolution
                                                                                                                                                                                                                                            January 27-29, 1994; Irvine, California
                                                                                                                                                                                                                                            Organizers: Francisco Ayala, Walter Fitch
                                                                                                                                                                                                                                            Proceedings: July 19, 1994
                                                                                                                                                                                                                                            Tempo and Mode in Evolution: January 1995
                                                                                                                                                                                                                                            Chemical Ecology: The Chemistry of Biotic Interaction
                                                                                                                                                                                                                                            March 25-26, 1994; Washington, D.C.
                                                                                                                                                                                                                                            Organizers: Thomas Eisner, Jerrold Meinwald
the authoritative version for attribution.