Diabetic Neuropathy and Neuropathic Pain: A (Con) Fusion of Pathogenic Mechanisms?
Diabetic Neuropathy and Neuropathic Pain: A (Con) Fusion of Pathogenic Mechanisms?
September 2020
· Volume 161
· Number 9
· Supplement 1 www.painjournalonline.com S65
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
S66
·
N.A. Calcutt 161 (2020) S65–S86 PAIN®
diabetes than to peripheral neuropathy,234 although there is 2.1.5. Pain generator site
supportive evidence associated with autonomic neuropathy.119
It is tempting to assume that if pain is perceived in the feet, then the
primary lesion site is to sensory nerves that innervate the feet.
2.1.2. Cellular targets Recent studies suggesting that pain correlates with nerve re-
generation markers in the skin30,58 have revived the old ideas that
As epineurial, perineurial, and endoneurial blood vessels are painful diabetic neuropathy may arise from the instability of
compromised in nerves of diabetic patients with early neurop- degenerating peripheral sensory neurons, ephaptic activation of
athy,222,269 a view of diabetic neuropathy emerged as repre- adjacent intact peripheral fibers, and/or activity of regenerating
senting a secondary manifestation of microvascular disease peripheral fibers.34 Hyperactive nociceptors and recruitment of
arising from hyperglycemia-induced damage to vascular endo- otherwise silent nociceptors have been recorded in subjects with
thelial cells.217 This aligns diabetic neuropathy with other painful diabetic neuropathy by microneurography257 and used to
complications of diabetes,261 and there is an association preselect subjects for clinical trial.315 However, the “irritable
between development of neuropathy and concurrent nephrop- nociceptor” phenotype (preserved small fiber function with hyper-
athy and retinopathy. The presence of resistance to ischemic algesia) formed only a small subset (6%) of a cohort comprising 191
conduction blockade also implies an early presence of ischemic subjects with painful diabetic neuropathy,348 and peripheral
hypoxia,217 although it also suggests that nerve metabolism hyperactivity may not be the only genesis of pain. A report that
adapts within acceptable tolerance limits. Whether vascular the onset of type 2 diabetes triggered symmetrical pain in both feet
insufficiency initiates peripheral neuropathy in diabetic patients of a patient who had 1 leg amputated some 44 years earlier
or impedes the capacity of cells within the nerve to withstand prompted the suggestion that the initiating lesion for diabetes-
direct glucotoxic or other insults remains an area of lively induced pain need not be at the site where pain is perceived.279 In
debate. Similarly, whether there is initial damage to Schwann support of this, there is a growing body of data emerging from
cells (primary schwannopathy)174 and to axons (primary imaging studies of diabetic subjects with painful neuropathy,
axonopathy)97 or independent and/or interdependent mecha- indicating that there is CNS dysfunction and pathology in regions
nisms of damage to each cell type122 has also been an area of associated with pain processing,310,312 whereas preclinical and
continuous investigation.349 clinical studies also suggest spinal involvement.225 In addition, there is
a growing appreciation that the genesis of neuropathic pain states
evolves over time, progressing from peripheral to central sites. Painful
2.1.3. Degenerative neuropathy diabetic neuropathy may therefore incorporate multiple generator
That numbness is usually first perceived in the toes, along with the sites whose relative dominance waxes and wanes as concurrent
early loss of epidermal fibers in the lower extremities, suggests degenerative pathology progresses. This complexity has implications
a length-dependent neuropathy associated with an inability to for selecting clinical trial populations that may be more heterogenous
maintain the regions of the axon that are most distant from the cell than previously recognized20 and result in the infrequent (number
body—not unlike the travails of Napoleon when invading Russia needed to treat: NNT . 5) and unpredictable efficacy of current pain
in the winter of 1812.197 However, reports that depletion of medications used to treat painful diabetic neuropathy.112
sensory nerves in the subbasal plexus of the cornea is an equally
sensitive marker for early neuropathy in diabetic patients indicate
that distal regions of the axon are most vulnerable, irrespective of 2.2. Glucotoxic mechanisms
the length.8,274 Longer axons may be particularly vulnerable to The occurrence of neuropathy in both forms of diabetes and the
accumulation of focal lesions due to size alone, but systemic success of intensive glycemic control regimens to slow pro-
insults do not appear to discriminate based on the absolute gression of diabetic neuropathy in type 1 diabetes267 naturally
axonal length. focused attention on hyperglycemia as the initiating event of
diabetic neuropathy. Glucose enters the peripheral nerve and
2.1.4. Painful vs painless neuropathy brain through insulin-independent glucose transporters (GLUTs).
GLUT-1 is the major glucose transporter of the microvascular and
Why approximately only a third of diabetic patients with perineurial components of the blood:brain and blood:nerve
degenerative neuropathy develop pain2 remains enigmatic. barriers,335 with GLUT-3 localized to peripheral neurons.338
Studies have sought clinical, structural, functional, or meta- Insulin-dependent GLUT-4 is restricted to select neuronal
bolic biomarkers that segregate subjects otherwise well populations of the brain.19 The search for detrimental con-
matched in presentation of diabetes and neuropathy into sequences of hyperglycemia has focused on modifications to the
those with or without pain.319,332 Recent examples are shown protein structure and function by direct glycation or enzyme-
in Table 1.30,34,58,59,76,90,98,113,114,139,176,177,195,215,223, mediated glycosylation along with excess flux through glucose
225,241,271–274,283,310,312,313,318,327,333,341,348,357,360,384,414
However, metabolism pathways (Fig. 1).
interpretation of such studies is complicated by the heteroge-
neity of the pain state caused by diabetes, and there have been
few attempts to identify biomarkers that identify specific pain 2.2.1. Models of diabetic neuropathy
subcategories identified among diabetic patients.20,348 Per- In vitro studies allow direct environmental manipulation of
haps the best examples to date are associations of burning individual cell types and are valuable for identifying plausible
pain with gain-of-function mutations in subunits of the Nav1.7 pathogenic mechanisms, with the recognized caveats that these
ion channel,11,29 although this represents a rare subgroup are traumatically excised tissues in artificial environments—
within those with painful diabetic neuropathy. Identification of neurons enter an axonal injury and regeneration phenotype while
features unique to subjects with pain could reveal potential Schwann cells return to their nonmyelinating form.117 Perhaps
pathogenic mechanisms specific for pain, with the usual the most sophisticated studies use cells derived from adult
caveat that any implied causality must be proven. control and diabetic animals maintained under conditions that
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
September 2020
· Volume 161
· Number 9
· Supplement 1 www.painjournalonline.com S67
Table 1
Potential biomarkers for painful vs painless diabetic neuropathy.
Type Tissue Biomarker for painful neuropathy Representative publications
Structural (PNS) Nerve biopsy Small fiber damage and regeneration 34,215,220
Skin biopsy Increased regeneration and/or axonal swellings 30,58,59,113
Cornea Increased corneal nerve loss 176,177,225,274
Increased corneal nerve branching 272
Structural (CNS) Cortex Atrophy of the somatosensory cortex 312
Functional (PNS) Nerve Increased epineurial blood flow 98
Skin Impaired stimulus-evoked blood flow 273
Skin Increased laser Doppler image (LDI) flare (small fiber function) 195
Sensory systems Severe hypoalgesia 283,348
Functional (CNS) Spinal cord Impaired rate-dependent depression of the H wave 225
Periaqueductal gray Dysfunction of descending inhibitory systems 310
Thalamus Hyperperfusion 313
Anterior cingulate cortex Hyperperfusion 384
Limbic/striatal structures Increased responses to stimuli 360
Others Blood Increased C-reactive protein (CRP) and soluble Intercellular 90
Adhesion Molecule 1 (slCAM-1)
Increased TNFa 241,271
Increased iNOS 271
Reduced vitamin D 318
Reduced Glo-1 327
Physiology Sex (female) 139,357
Increased body mass index 414
Autonomic dysfunction 76,114,333,341
Studies in which no change was detected are shown in italics.
reflect the in vivo insulin and glycemic environment from which provide most data that underpin current hypotheses regarding the
they were derived133 and coculture neurons and Schwann cells pathogenesis of diabetic neuropathy and neuropathic pain. Both
to facilitate myelination.342,364 species can be used to model prediabetes and type 1 or type 2
Of the animal models, diabetic cats exhibit nerve pathology that diabetes using dietary, chemical, or genetic initiating events. There
most closely reflects the human condition, with prominent de- are variations in the neuropathy phenotype and rate of progression
myelination and axonal degeneration.238 Rats and mice are the between specific models, species, strains, laboratories, and
most commonly used animal models of diabetic neuropathy and assays.26 The provenance of new disorders identified in
Figure 1. Mechanisms of glucotoxicity in diabetic neuropathy: Hyperglycemia has been considered central to the pathogenesis of diabetic neuropathy, and
multiple mechanisms have been proposed from both preclinical studies and clinical observations (see text for details). AGE, advanced glycation end product; PDK,
pyruvate dehydrogenase kinase; PKC, protein kinase C; RAGE, receptor for AGE; ROS, reactive oxygen species; TCA, tricarboxylic acid cycle.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
S68
·
N.A. Calcutt 161 (2020) S65–S86 PAIN®
Figure 2. Rat plantar skin stained with antibody to PGP9.5 and viewed by bright field (panel A) or fluorescence (panel B) microscopy reveals dermal nerves (yellow arrows)
projecting across the dermal:epidermal junction and into the epidermis (purple/blue counterstained nuclei) where they form profiles of intra-epidermal nerve fibers (red arrows).
Note that PGP9.5 also stains epidermal Langerhans cells (red circles). Diabetic rodents and humans (panel C) show early reductions in IENF density that are associated with
both sensory loss and pain.23,274 Confocal images of the corneal sub-basal nerve plexus of a live mouse (panel D, showing inferior whorl) and human (panel E). Reduced
corneal nerve morphometric parameters are detected within weeks of onset of diabetes in rodents52 and in early stages of clinical diabetic neuropathy.274 A cross section of
sciatic nerve from a STZ-diabetic rat (panel F), with an endoneurial blood vessel at the centre of field (black circle), lacks overt evidence of the axonal degeneration or
demyelination common in nerve biopsies from diabetic patients. Apparently misshapen axons (black arrows) represent normal paranodal regions of the nerve fiber while
multiple myelin profiles illustrate normal Schmidt-Lanterman incisures (red arrows). Mild axonal fixation artifact is illustrated by the black star. Morphometric analysis identifies
reduced mean axonal diameter in the absence of significant fiber loss, indicative of axonal atrophy or impaired maturation. Technical details of procedures used to generate
these images and representative data showing the effects of diabetes are published.167 Images provided by Ms. Katie Frizzi, Ms. Lucie Guernsey and Ms. Alex Marquez.
streptozotocin (STZ)-diabetic rodents, the most commonly used hyperglycemia-driven flux through the polyol pathway results in
model of type 1 diabetes, should be established to address accumulation of the intermediates (sorbitol and fructose) along with
concerns about acute STZ neurotoxicity.15,259,294 This can be done shifts in the redox balance of the associated cofactors nicotinamide
using concurrent 3-O-methylglucose injection,80,225 using insulin to adenine dinucleotide phosphate and nicotinamide adenine dinucle-
reverse established disorders,225 and by validating disorders using otide (NADPH and NADH). Downstream consequences potentially
genetic or dietary models.402 Rodents are frequently studied over 4 include local osmotic stress due to sorbitol accumulation,250 fructose-
to 12 weeks of diabetes and are best viewed as modeling early driven advanced glycation end product (AGE) formation12 and
nerve dysfunction in the absence of overt pathology,378 as structural subsequent receptor for AGE (RAGE) signaling, oxidative75 and
pathology in nerve trunks (Fig. 2) is limited to reduced axonal caliber nitrosative stress,72 and loss of neurotrophic support.236 As reviewed
with late (4 months1) myelin thinning, occasional segmental elsewhere,250 preclinical studies show impressive efficacy of aldose
demyelination, and minor fiber loss.157,158,182,268,288,396 This can reductase inhibitors (ARIs) against many indices of diabetic neurop-
be viewed as a boon, as molecular and biochemical changes in athy and neuropathic pain that culminated in a number of clinical trials.
nerves may be interpreted as preceding, perhaps precipitating, To date, ARI treatment has not shown sufficiently convincing efficacy
degenerative neuropathy. Unfortunately, it also limits any guaran- in clinical trials to support approval by most regulatory agencies,
tees of the translation of therapies developed using these models. although epalrestat is approved in Japan. Whether this reflects
Recognition that diabetic rodents develop early loss of sensory a pathogenic mechanism that is pertinent only to diabetic rodents,
nerve terminals in the epidermis of the plantar skin (frequently suboptimal drug properties for human use, or poor clinical trial design
termed intraepidermal nerve fibers [IENFs])23 and reduced sensory and inappropriate endpoints remains the subject of unresolved
nerve density in the cornea52 that accompany indices of both debate.250 Polyol pathway research is currently out of vogue, but the
sensory loss and hyperalgesia167 to mirror the human condition274 impressive preclinical efficacy of ARIs must either indicate a major
have revived hopes that rodent models can be used to study the contribution to downstream pathways that damage nerves or illustrate
early damage to small sensory neurons that is a feature of diabetic a disconcerting gulf between preclinical models and the human
neuropathy (Fig. 2). disease.
2.2.2. Glucose metabolism by the polyol pathway 2.2.3. Nonenzymatic glycation and the advanced glycation
Early observations that tissues that contained the polyol pathway end product–receptor for advanced glycation end product
axis
enzymes, aldose reductase and sorbitol dehydrogenase, were prone
to diabetic complications prompted extensive research into their The posttranslational modification of cellular proteins caused by
potential pathogenic role.95 Within the peripheral nerve, aldose nonenzymatic attachment of glucose to amino acids, causing
reductase is localized to endothelial and Schwann cells164 and reversible progression from Schiff base to Amadori products and
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
September 2020
· Volume 161
· Number 9
· Supplement 1 www.painjournalonline.com S69
then irreversible formation of AGEs, has intermittently recurred as phosphate, and dihydroxyacetone phosphate) and cleared by the
a mechanism of potential glucotoxicity in many organs, including glyoxalase pathway. Both excess glycolysis and impaired activity of
nerves.350,373 Advanced glycation end products are present glyoxalase pathway enzymes may contribute to accumulation of
throughout the peripheral nerve,339 and the initial focus was on methylglyoxal in diabetes, which reacts with proteins to form AGEs
the modification of components of the axonal cytoskeleton that (see above). Mice overexpressing glyoxalase 1 do not develop
could interrupt axonal transport and axial and radial growth229 indices of degenerative diabetic neuropathy.155 A gain-of-function
along with the modification of the basement membrane and property has also been proposed from studies showing that binding
extracellular matrix proteins that could impede neuronal re- of methylglyoxal to the Nav1.8 voltage-gated sodium channel in
generation after injury.94,188 More recently, glycosylation of ion sensory neurons increases excitability,25 whereas the potential for
channels has been implicated in painful diabetic neuropa- methylglyoxal to produce pain in diabetes may also be mediated
thy,25,256,388 as discussed below. through agonism of the transient receptor potential A1 (TRPA1)
Identification of a RAGE on the surface of neurons, Schwann channel in the peripheral nerve190 and spinal cord125 and induction
cells, and vascular endothelial cells375 aligned nerves with other of the integrated stress response.21 Recent studies have confirmed
organs prone to damage during chronic diabetes.299 In other the pronociceptive properties of methylglyoxal in humans,92 and
tissues, AGE binding to RAGE activates NADPH oxidase with elevated plasma methylglyoxal has been identified as a risk factor for
subsequent release of reactive oxygen species, whereas RAGE neuropathy in patients with type 2 diabetes.13 Approaches in
signaling through NF-kB modifies gene expression, promoting reducing methylglyoxal levels or blocking downstream consequen-
inflammation and dysregulation of the survival/apoptosis equilib- ces are in development.
rium. There is evidence that similar toxic events occur in nerves,356
with recent in vitro studies demonstrating that RAGE signaling
2.2.5. Mitochondrial overdrive or idling?
potentiates transient receptor potential V1 (TRPV1)-mediated
calcium signals and contributes to painful neuropathy.24,198 It has been argued that increased substrate-driven glycolysis with
Preclinical studies of agents with anti-AGE/RAGE properties such subsequent Krebs’ cycle activity and oxidative phosphorylation
as aminoguanidine and B vitamins show some efficacy in rodent (OXPHOS) in mitochondria will result in the formation of free radicals
models of diabetic neuropathy395 and neuropathic pain,169 that may not be adequately buffered so that there is oxidative
whereas a small-scale clinical trial of the vitamin B1 derivative damage to local structures. This hypothesis was developed in
benfotiamine suggested improvement in pain.337 However, these endothelial cells exposed to short periods of hyperglycemia in
agents have other potential mechanisms of action, and it should vitro.245 There is little evidence that substrate-driven “overdrive” of
also be noted that RAGE signaling is reported to have beneficial mitochondrial OXPHOS persists and is toxic to nerves. Exposing
effects in nerves such as promoting neurite outgrowth304 and that Schwann cells to acute hyperglycemia causes an increase in free
RAGE deletion attenuated indices of neuropathy in diabetic mice.84 radical buffering capacity and does not increase reactive oxygen
species production.372 In neurons from diabetic rodents, basal
respiration is unchanged and maximal mitochondrial respiratory
2.2.4. Glycolysis
capacity reduced, not increased.60,408 This is accompanied by
Hyperglycemia-driven increases in intermediates of glycolysis have reduced expression and activity of the mitochondrial proteome7,61
been linked to diabetic complications, including neuropathy. For and dysregulation of mitochondrial biogenesis,49 with aberrant
example, metabolism of fructose 6-phosphate by the hexosamine fission/fusion dynamics.301 It has therefore been proposed that
(glucosamine) pathway produces UDP-N-acetylglucosamine, nutrient excess promotes the downregulation of mitochondrial
which is highly reactive with proteins, most notably transcription function and increasing reliance on glycolysis.105 This metabolic shift
factors, in a process called O-GlcNAcylation. This pathway has supports normal neuronal function during hyperglycemia but may
been particularly linked with cardiovascular complications of limit energy-intensive processes such as dynamic plasticity of
diabetes,263 and a recent study indicates that O-GlcNAcylation peripheral terminals in the epidermis and the capacity to respond to
regulates remyelination of peripheral neurons after injury so that other insults. Efficacy of diverse activators of the adenosine
hyperglycemia-driven abnormal O-GlcNAcylation has the potential monophosphate activated protein kinase/peroxisome proliferator-
to impact nerve structure and function in diabetes.187 activated receptor gamma coactivator 1 alpha (AMPK/PGC1a)
In vascular tissue, glucose-derived diacylglycerol (through pathway, a nutrient sensor system that regulates mitochondrial
glyceraldehyde 3-phosphate and phosphatidic acid) is a substrate activity and dynamics, in restoring mitochondrial respiration and
for protein kinase C b (PKC b), and excess glucose drives elevated preventing or reversing multiple indices of neuropathy and
PKC b activity, which in turn promotes increased vascular neuropathic pain in diabetic rodents4,43,298,400 supports this
permeability and dysfunction.120 The association of diabetic concept.
neuropathy with microvascular disease led to interest in the There is an emerging appreciation that not all cells within the
therapeutic potential of PKC b inhibitors. After supportive pre- nervous system use glucose for adenosine triphosphate (ATP)
clinical studies,45 a clinical trial of ruboxistaurin in diabetic patients production in a similar manner. For example, neurons of the CNS
with neuropathy showed some improvement in skin blood flow, the express high levels of pyruvate dehydrogenase (PDH), which
NTSS-6 questionnaire, which quantified frequency and intensity of controls entry of pyruvate into the Krebs cycle and drives
aching, burning, prickling, lancinating pain, numbness, and OXPHOS, whereas astrocytes have high levels of lactate
allodynia, and in quality of life.48 However, there was no significant dehydrogenase, which converts pyruvate to lactate.201 Utilization
effect on other measures of large and small fiber neuropathy, of glycolysis-derived pyruvate in astrocytes is also limited by the
diminishing enthusiasm in this therapeutic approach. suppression of PDH complex activity through phosphorylation by
There is an increasing focus on the role of the intermediate pyruvate dehydrogenase kinase (PDK). Consequently, these
glycation product methylglyoxal in diabetic complications, including relatively quiescent glial cells rely primarily on glycolysis-derived
neuropathy and neuropathic pain. Methylglyoxal is formed by ATP and indeed may provide lactate to neurons as energy
nonenzymatic dephosphorylation of triose phosphate intermediates substrate.172 By contrast, electrically active neurons keep the
of glycolysis (fructose 1,6-bisphosphate, glyceraldehyde 3- PDH gateway open and use the more efficient generation of ATP
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
S70
·
N.A. Calcutt 161 (2020) S65–S86 PAIN®
by OXPHOS.135 The metabolic dependence of neurons on glia is notable are studies in which trace insulin was injected into the
highlighted by the report that selective damage to Schwann cell footpad,130 infused into the spinal intrathecal space,355 or applied
mitochondria results in a neuropathy with damage to both myelin topically to the eye52 of STZ-diabetic rodents. In each case,
and axons.368 Diabetes has been reported to increase PDK treatment prevented functional and structural indices of neurop-
expression and activity in peripheral neurons and glia, supporting athy without impacting hyperglycemia, indicating that provided
the idea that during persistent hyperglycemia, neurons suppress there is adequate insulin-derived trophic support, hyperglycemia is
entry to OXPHOS and rely on glycolysis for ATP production.276 not sufficient to induce neuropathy.
Conversely, PDK deficiency attenuated multiple indices of A mechanism of direct insulin action on neurons may involve
neuropathy in diabetic mice including overexpression of TRPV1 mitochondria. Insulin increases mitochondrial inner membrane
and neuropathic pain. How hyperglycemia impacts the distinct potential when applied directly to sensory neurons derived from
metabolic profiles of neurons and Schwann cells has yet to be normal rats in vitro.149 Moreover, the reduced inner membrane
widely explored, but such studies may provide insight into cell- potential, protein expression, and bioenergetics profile of
specific mechanisms of glucotoxicity. mitochondria from sensory neurons of STZ-diabetic rodents are
restored when animals received trace insulin supplementation
that also impacted functional and structural indices of diabetic
2.3. Impaired insulin signaling
neuropathy without impacting hyperglycemia.3,61,149 This ability
The correlation between glycemic control and neuropathy reported to protect against both mitochondrial dysfunction and the
in the DCCT study is not overwhelming, although a follow-up study neuropathy phenotype also extends to IGF-1,4 with the apparent
failed to show reversal of established neuropathy upon instigation redundancy perhaps reflecting the importance of the system to
of improved glycemic control.226,267 Similar studies of glycemic cells with consistently elevated energy demands.
control in type 2 diabetic subjects did not replicate the DCCT Insulin secretion is accompanied by equimolar release of C-
findings for neuropathy,289,417 and indices of neuropathy are peptide, the other product of proinsulin cleavage. Although
detected in patients with prediabetes (elevated fasting glucose initially considered inert, there is evidence that C-peptide has
levels and/or impaired glucose tolerance) and metabolic syndrome biological actions in a variety of tissues, including peripheral
(representing a combination of risk factors for progression to overt nerves,322 possibly through an insulin-sensitizing action. As C-
diabetes—central obesity, high triacylglycerides and low-density peptide prevents and reverses multiple indices of neuropathy409
lipoprotein (LDL)-cholesterol, low high-density lipoprotein (HDL)- and neuropathic pain179 in animal models of type 1 diabetes170
cholesterol, hypertension, and hyperglycemia).264 A recent study and showed efficacy against some manifestations of diabetic
that subdivided a cohort of 1105 recently diagnosed diabetics into neuropathy in a clinical trial,377 it should perhaps be aligned in
5 groups based on multiple metabolic parameters found that tandem with insulin when considering primary pathogenic
peripheral neuropathy was most prevalent in those with severe mechanisms of diabetic neuropathy and neuropathic pain.
insulin-deficient diabetes.405 Taken together, these observations
have driven interest in nonglucotoxic insults that may act either
2.4. Dyslipidemia
alone or in concert with hyperglycemia.126
Diabetes can be viewed as a disease of impaired insulin Major risk factors for developing diabetic neuropathy reflect
signaling because of insulinopenia and/or insulin resistance. exposure to impaired insulin signaling/hyperglycemia (age, duration
Insulin is structurally similar to the liver-derived peptides, insulin- of diabetes, and long-term glycemic control), followed by vascular
like growth factors 1 and 2 (IGF-1 and IGF-2), and shares their dysfunction (hypertension and smoking) and dyslipidemia (obesity,
growth factor–like properties. Insulin receptors are found on elevated plasma cholesterol, and triacylglycerols).31 Clinical evi-
peripheral neurons,35 signal through Akt,128 and activation dence linking changes in specific plasma lipids to neuropathy is
promotes growth of normal sensory111 and motor394 neurons. mixed, with studies that both demonstrated, and failed to
Conversely, sequestration of local insulin causes peripheral demonstrate, associations between elevated triacylglycerols, ele-
neuropathy in normal rats.35 Schwann cells also express insulin vated LDL-cholesterol or reduced HDL-cholesterol, and neuropa-
and IGF-1 receptors, and their depletion results in a peripheral thy. Clinical efficacy of lipid-lowering agents such as statins and
neuropathy phenotype with injury to both Schwann cells and fibrates against neuropathy is promising but limited.82,244,278,369
axons.134 Loss of insulin-mediated trophic support represents Nevertheless, there is growing interest in how dyslipidemia may
a primary pathogenic mechanism of diabetic neuropathy that is damage peripheral nerves to produce degenerative and painful
independent of hyperglycemia. This applies to both type 1 and neuropathy that is driven by recent preclinical studies.
type 2 diabetes, as studies in type 2 diabetic rodents indicate that Indices of neuropathy and neuropathic pain are detected in
insulin signaling is impaired in the peripheral nerve,127 so that the rodents fed high fat diet to induce insulin resistance and
nervous system can be considered insulin resistant. dyslipidemia but not overt hyperglycemia,80,81,252,371 although
A number of preclinical studies have demonstrated a role for the neuropathy phenotype may be species and strain specific.14
insulin deficiency in diabetic neuropathy and neuropathic pain. Many of these disorders are prevented or reversed by treating
Animals injected with STZ at doses that significantly reduce insulin prediabetic, type 1 or type 2 diabetic rodents with diets high in n-3
production but do not cause hyperglycemia go on to develop polyunsaturated fatty acids to adjust the plasma ratio of n-3:n-6
hyperalgesia in the paw pressure test.292–294 Insulin-resistant but polyunsaturated fatty acid.68–70,199,316 Adjusting high fat diets to
normoglycemic models of prediabetes also develop neuropathy,80 increase the proportion of monounsaturated fatty acids shows
and the onset of allodynia to von Frey filaments parallels the onset similar efficacy,300 and there is a growing suspicion that long-
of insulin resistance but precedes the onset of hyperglycemia in chain saturated fatty acids may therefore be a proneuropathic
a model of type 2 diabetes.295 Conversely, insulin-deficient diabetic entity in dyslipidemia.247 Downstream pathogenic mechanisms
rodents treated with trace amounts of systemic insulin for more may include disruption of mitochondrial function and transport in
than 1 year to maintain body weight without impacting systemic sensory neurons.301 Dyslipidemia thus joins glucotoxicity and
hyperglycemia showed attenuation of large fiber conduction impaired insulin signaling as a potential driver of mitochondrial
slowing and did not progress to paw heat hypoalgesia.37 Most dysfunction to cause peripheral neuropathy (Fig. 3).
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
September 2020
· Volume 161
· Number 9
· Supplement 1 www.painjournalonline.com S71
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
S72
·
N.A. Calcutt 161 (2020) S65–S86 PAIN®
Table 2
miRNA implicated in the pathogenesis of diabetic neuropathy.
miRNA species Impact of diabetes Model (tissue) Effective therapeutic manipulations Representative publications
Multiple Multiple upregulated STZ-mouse (DRG) Mimics/knock-out impact multiple indices of 57
Multiple downregulated degenerative and painful neuropathy
Multiple Multiple upregulated STZ-rat (DRG) Not done 131
Multiple downregulated
Multiple Multiple upregulated STZ-rat (nerve) Some expression changes normalized by taurine 317
Multiple downregulated
miR-25 Downregulated db/db-mouse (nerve) Precursor reduced inflammation markers 410
miR-29c Upregulated db/db-mouse (DRG, nerve) knock-out improved neurite growth in vitro 163
miR-34c Upregulated STZ-mouse (trigeminal) Antagomir improved corneal nerve growth 147
miR-96 Downregulated high fat diet/STZ-rat (nerve) Exercise increased miR, and reduced Nav1.3 and 5
thermal pain
miR-106a Downregulated STZ-mouse (DRG) Not done 392
miR-146a Upregulated STZ-rat (nerve) Not done 403
Downregulated db/db-mouse (nerve) Mimics improved neuropathy/pain 214
STZ-rat (nerve) Not done 104
db/db-mouse (nerve) Inducer improved neuropathy/pain 379
miR-155 Downregulated STZ-rat (nerve) Not done 184
Upregulated STZ-rat (nerve) Antagomir improved neuropathy 53
miR-181a Upregulated STZ-mouse (trigeminal) Antagomir improved corneal nerve growth 148
miR-182 Downregulated STZ-mouse (trigeminal) Agomir increased corneal nerve density 382
proteins.38 There are also reports of diabetes-induced increased nerve macrophages67,246 and spinal microglia.361 The absence
expression of trophic factors, as occurs with brain-derived of large increases in inflammatory cell numbers is not surprising
neurotrophic factor (BDNF)107 and both hypoxia-inducible because of the physical constraints on tissue expansion imposed
factor 1 (HIF-1) and its target gene product, vascular endothelial by the epineurium and spinal column. Glial cells of the peripheral
growth factor (VEGF),50,305 that are attributed to responses to nervous system (PNS) and CNS play many roles associated with
injury or compensation for loss of other trophic support systems. peripheral inflammatory cells after nerve injury, including the
VEGF delivery improves indices of neuropathy in diabetic release of cytokines and chemokines91 and clearance of myelin
rodents,150,290,308 although the role of BDNF is more complex. debris.161 As recently reviewed in detail elsewhere,287 this
Acute spinal delivery of BDNF causes tactile allodynia in normal neuroinflammatory system is increasingly recognized as being
rats, and sequestration of endogenous spinal BDNF alleviates dysregulated in diabetes.
tactile allodynia and restores H-wave rate-dependent depression There are multiple reports of changes in both proinflammatory
(RDD) (see above) in diabetic rats.205 By contrast, chronic spinal and anti-inflammatory cytokines and chemokines in the nervous
delivery of BDNF to diabetic rats alleviated hyperalgesia.206 In system of diabetic rodents. The most widely studied are
some cases, altered neurotrophic support has been linked to increases in the proinflammatory cytokines, IL-1b, IL-6, tumor
downstream consequences of hyperglycemia. For example, necrosis factor alpha (TNFa), and interferon-g,418 along with
hyperglycemia-driven increased flux through the polyol pathway changes to downstream transcription regulators such as NF-kB
leads to reduced nerve levels of the Schwann cell–derived factors and Nrf2.196 Expression of bradykinin B1 and B2 receptors by
NGF254 and CNTF,236 and ARI treatment prevents diabetes- microglia and neurons is also increased in the spinal cord of
induced elevated Trk-C receptor mRNA expression.320 However, diabetic rodents,46,343 and receptor antagonists alleviate indices
the pathogenesis of most neurotrophic factor deficits in diabetic of pain.343 Pathogenic consequences include induction of
nerves remains unknown, and clinical trials using trophic factors enzymes such as cyclooxygenase-2 (COX-2)181,282 and isoforms
have not been promising,16,102,389 with the exception of early of nitric oxide synthase415 that can drive tissue damage through
HGF trials against neuropathic pain6 and a neurotrophic eryth- ischemic hypoxia and oxidative/nitrosative stress, thereby
ropoietin analog against pain and corneal nerve loss.33 contributing to both degenerative neuropathy and initiation or
amplification of neuropathic pain.115 For example, induction of
COX-2 and subsequent release of prostaglandins from spinal
2.5.4. Cytokines and inflammatory pathways
oligodendrocytes282 and release of proinflammatory cytokines343
There is little evidence from human studies that diabetic and BDNF240 from activated microglia have been linked to
neuropathy represents a typical inflammatory neuropathy. spinally mediated pain in diabetic rodents. Increased chemokine/
Chronic inflammatory demyelinating polyneuropathy occurs in receptor signaling, including CCL1/CCR8,420 CXCL12/
diabetic patients but is readily distinguished from diabetic CXCR4,159 CXCL13/CXCR5,213 and others,419 is also linked to
symmetrical polyneuropathy.277 Animal models of diabetes also neuropathic pain in diabetic rodents. The primary pathogenesis of
lack marked inflammatory infiltrates, although there are reports of many of the reported changes to the neuroinflammatory system
transient increases in the number and activation of peripheral remains to be determined, although hyperglycemia-driven flux
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
September 2020
· Volume 161
· Number 9
· Supplement 1 www.painjournalonline.com S73
through the polyol pathway may be involved in some sensory neurons from diabetic animals, whereas knockdown of
aspects.156,282,334 Therapeutic approaches around manipulation PTEN improved the otherwise impaired nerve regeneration after
of the neuroinflammatory system tested in diabetic rodents range crush injury in STZ-diabetic mice.325
from microglial inactivators,240,418 COX inhibitors,266,282 TNFa
inactivators397 or inhibitors,89 overexpression of anti-
2.5.6. Membrane pumps
inflammatory cytokines,347 antagonists to proinflammatory cyto-
kines,138 chemokine-neutralizing antibodies,291 and chemokine NCV slowing in short-term diabetic rodents that lack overt damage
receptor antagonists.175,231 Efficacy of stem cells137,385 and to axons or myelin led to interest in changes to nodal ion pumps
natural products78,166,253 against aspects of diabetic neuropathy that facilitate saltatory conduction in myelinated fibers. Chief of
may also be at least partly due to their anti-inflammatory these was the Na1/K1 ATPase, given its role in maintaining and
properties. To date, clinical trials have not been successful.175 restoring resting membrane potential. Reduced maximal pump
activity in membrane fragments associated with reduced protein
expression was widely studied as a potential cause of NCV slowing
2.5.5. Death and survival pathways
and is downstream of hyperglycemia-driven polyol pathway
Dysregulation of cytoplasmic calcium has been implicated in activity.124 However, Na1/K1 ATPase pump activity is not impaired
many neurodegenerative diseases, given its role in triggering in intact endoneurial preparations from diabetic rodents,216 so the
autophagic, necrotic, and apoptotic pathways.51,123 Steady- physiological relevance is unclear. Increased expression and
state cytoplasmic calcium concentrations are regulated by activity of the Na1/H1 pump has also been reported in the nerve
pumps located in mitochondria and the endoplasmic reticulum, of diabetic rodents, and inhibition of the pump reversed functional
and these organelles serve as calcium stores. Steady-state and structural indices of neuropathy and neuropathic pain.219
cytoplasmic calcium concentrations are increased in sensory Overactivity of this pump increases cytoplasmic pH, glucose
neurons from diabetic rodents,367 and this is associated with uptake, and glycolysis, thereby having the potential to trigger
impaired calcium reuptake into the endoplasmic reticulum by the multiple pathogenic mechanisms.
sarcoendoplasmic reticulum Ca21 ATPase (SERCA) pump.411 In the spinal cord, reduced expression of the potassium
Subsequent depletion of calcium in the sarcoplasmic reticulum chloride cotransporter 2 (KCC2) pump, which maintains the
produces endoplasmic reticulum (ER) stress, which can pre- chloride gradient across neuronal membranes, has been linked
cipitate cell death,306 and this mechanism has been integrated with loss of GABAergic inhibitory function and neuropathic pain in
into the schema of potential pathogenic mechanisms of diabetic diabetic rats.204 KCC2 expression is suppressed by the neuro-
neuropathy.248 It is not yet known whether mitochondrial calcium trophic factor BDNF, and increased BDNF levels in central
pumps, such as the mitochondrial calcium uniporter complex and projections of primary afferents in diabetic rats suggest that the
voltage-dependent anion channel, are dysfunctional in diabetes. primary lesion may be of peripheral origin,204 although it has also
Reports describing the marked expression of components of been linked to activated spinal microglia.240 The electrophysio-
apoptotic death pathways in the peripheral nerve of short-term logical consequence of the disrupted spinal GABAergic inhibitory
diabetic rodents302,307 were initially difficult to reconcile with the tone in diabetic rodents is loss of RDD of the H wave,205 which is
slowly evolving loss of neurons and axons in these models and secondary to impaired insulin signaling rather than hyperglyce-
clinical descriptions of a distal degenerative neuropathy. Later mia.225 Loss of RDD may serve as a biomarker for identifying
work reported that increased caspase-3 in nerves was not a subset of diabetic humans in whom painful neuropathy includes
associated with structural features of apoptosis such as nuclear a contribution from spinal disinhibition.225
fragmentation and that DNA-repair mechanisms were activated,
suggesting that the peripheral nerve uses endogenous defence
2.5.7. Transient receptor potential channels
mechanisms to block progression from caspase-3 activation to
apoptosis.57 Overactivity of the DNA-repair enzyme poly(ADP- Transient receptor potential (TRP) channels are a family of non-
ribose) polymerase (PARP) has itself been linked to a mechanism selective cation-permeable channels that transduce diverse extra-
of nerve injury.218 Elevated PARP and other survival and repair cellular stimuli into acute and chronic neuronal responses through
markers such as heat shock protein 27 (HSP27)280,416 and influx of calcium.173 TRPA, TRPV, and TRPM family members are
growth-associated protein 43 (GAP43)130 in the nerve of diabetic modulated by endocannabinoids, which may contribute to the
rodents has encouraged the view that the peripheral nerve is analgesic properties of these substances.242 There is substantial
exposed to chronic stress arising from one or more of the preclinical evidence that dysregulation or dysfunction of TRP
mechanisms described previously but can largely tolerate and channels may contribute to neuropathic pain in diabetes.
repair metabolic injuries, paralleling to the way that it survives and The TRPV1 channel, known for transducing the burning
repairs physical injury. Therapeutic approaches that strengthen sensation of capsaicin, is activated by a range of physiological
endogenous survival and repair mechanisms include over- and pathological stimuli, including heat, low pH, proinflammatory
expression of HSP 27192 and inhibition of HSP90 to induce molecules, and endocannabinoids. TRPV1 currents are also
HSP70, a molecular chaperone protein with multiple neuro- enhanced by insulin and IGF-1366 and by the TRPM8 receptor.258
protective properties including protecting mitochondrial function An initial report144 indicated that membrane-bound TRPV1
and reducing inflammation and oxidative stress.220 HSP90 protein is increased in the dorsal root ganglia (DRG) from diabetic
inhibitors reverse multiple indices of neuropathy in diabetic rats, along with the channel phosphorylation state and both
rodents and are currently in clinical development.103 Conversely, capsaicin and proton-activated currents, whereas TRPV1 protein
the emerging appreciation that there are endogenous systems expression increased in large sensory neurons and decreased in
that constrain nerve growth has provided opportunity to intervene small sensory neurons. The pattern of TRPV1 protein expression
and thereby promote nerve growth and regeneration pathways.93 and whole-cell currents in the DRG and spinal cord of diabetic
For example, the tumor suppressor molecule PTEN (phospha- rodents paralleled progression from heat hyperalgesia to
tase and tensin homolog deleted on chromosome 10) inhibits the hypoalgesia,259 and increased TRPV1 expression at peripheral
PI3K-pAkt neuronal growth pathway194 and is upregulated in and central terminals of primary afferents has been implicated in
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
S74
·
N.A. Calcutt 161 (2020) S65–S86 PAIN®
allodynia to von Frey filaments.74 Agents that reduce TRPV1 other indices of peripheral neuropathy were largely attributed to
expression, antagonize TRPV1, or ablate it also alleviate thermal indirect effects through improved blood flow.28
hyperalgesia and tactile allodynia in diabetic rats.17,208 Involve-
ment in diabetic neuropathy beyond indices of pain is suggested
by a report that TRPV1 agonists given to normal mice produce 2.5.9. Voltage-gated potassium channels and
hyperpolarization-activated and cyclic nucleotide–gated
multiple indices of small fiber neuropathy including IENF loss.203 channels
Upstream events that may drive TRPV1-mediated pain include
increased insulin, RAGE and PKC activity,24,366 and hypoxia.285 Given the importance of potassium channels in regulating axonal
Little is known about other TRPV family members, although excitability, there are relatively few studies implicating them in the
a recent study reported that a selective TRPV4 channel pathogenesis of diabetic neuropathy and neuropathic pain.
antagonist blocked mechanical, but not cold, allodynia in diabetic Expression of the voltage-gated Kv channel subunits Kv1.2 and
mice.86 Kv1.6, but not Kv1.1, was reduced in small neuronal cell bodies of
Early indications of a role for the irritant-sensing TRPA1 the DRG in STZ-diabetic rats coincident with reduced K1
channel in diabetes-induced hyperalgesia came from studies currents, and these changes were linked to enhanced C-fiber
with TRPA1 antagonists that alleviated allodynia to von Frey excitability and hyperalgesia.380 Most recently, voltage-gated Kv7
filaments and mechanical hyperalgesia.386,387 Cold allodynia in (KCNQ) channels, which produce a slow noninactivating outward
diabetic mice has also been attributed to TRPA1 activity.142 K1 current also called the M current due to its modulation by
Diabetes enhances channel activity without inducing TRPA1 muscarinic antagonists,390 have been examined. There was
protein expression in the DRG of diabetic animals.286 As decreased mRNA and protein for the Kv7.2, Kv7.3, and Kv7.5
discussed previously, a pathogenic mechanism linking hypergly- channels in the DRG of STZ-diabetic rats accompanied by
cemia with TRPA1 channel activation through methylglyoxal reduced M current density and increased neuronal excitability.404
binding to the channel has been proposed,99,191 and there is also A Kv7 channel activator reduced neuronal excitability and
a recent report linking TRPA1-mediated hyperalgesia in diabetic alleviated allodynia to von Frey filaments and thermal
rodents to local hydrogen sulfide.286 hyperalgesia.
Protein for the cold/menthol-sensing TRPM8 channel is elevated Hyperpolarization-activated and cyclic nucleotide–gated
in the DRG of STZ-diabetic rats with concurrent cold allodynia.401 channels (HCN1-4) are a distinct category of voltage-gated ion
Conversely, agonist-activated TRPM8 currents are decreased in channels whose threshold potentials are regulated by cyclic
the DRG of STZ-diabetic mice258 and in normal DRG cells after nucleotides and that have been implicated in neuropathic pain
exposure to methylglyoxal.66 Although this does not appear to be states.359 Inhibition of HCNs1-4 or ablation of HCN2 alleviated
consistent with a role for methylglyoxal in diabetes-evoked pain allodynia to von Frey filaments but not thermal hypoalgesia in
(see above), it has been argued that loss of TRPM8 activity diabetic mice.358 Expression of HCN1-4 protein was not altered
enhances other TRP channel activities associated with neuropathic in the peripheral nerve of diabetic mice, and it was speculated that
pain.258 There is clearly room for additional studies in this area. a measured increase in intracellular cAMP might activate HCN2
and thus increase primary afferent firing.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
September 2020
· Volume 161
· Number 9
· Supplement 1 www.painjournalonline.com S75
of NAv1.3,344 by induction of miR-96 to reduce Nav1.3 NMDA NR1 and NR2B subunits also show an increased
expression,5 and by blockers of Nav1.7383 and Nav1.8.365 Topical phosphorylation (activation)79,154 secondary to elevated protein
lidocaine has shown efficacy against painful diabetic neuropa- tyrosine phosphatase activity.340 NR2B expression is also
thy391 and is used off-label.9 A clinical trial of a Nav1.7 blocker in increased in a model of prediabetes.340 These findings are
subjects with painful diabetic neuropathy showed only minor consistent with the efficacy of spinally delivered NK-1 and NMDA
effects,227 perhaps reflecting the cohort of subjects in whom pain antagonists against allodynia and hyperalgesia in diabetic
could be due to a variety of diabetes-related mechanisms. rodents.39,71,79,296 Unfortunately, side-effect–free targeting of
Although the pathogenesis of altered expression and/or function spinal excitatory receptors as a strategy to treat painful diabetic
of VGSC is not clear, other than relatively rare gain-of-function neuropathy has been largely unsuccessful to date.
mutations and a link to hyperglycemia through increased Of the spinal inhibitory receptors, GABAA expression is
glycolysis and methylglyoxal for modification of Nav1.8 activity,25 unchanged by diabetes,168 but inhibitory function is diminished
there is an interesting suggestion that mutations in Nav1.7 may be secondary to reduced KCC2 activity (see above), whereas
a primary cause of both painful diabetic neuropathy and diabetes GABAB expression is reduced.381 Both basal and stimulus-
itself because of the location of this channel on both primary evoked spinal GABA levels are increased in diabetic rats224 and
sensory neurons and pancreatic b cells.143 may contribute to pain through dysfunctional GABAA receptors,
as GABAA antagonists alleviate allodynia and hyperalgesia,168
whereas activation of GABAB receptors shows the expected
2.5.11. Neurotransmitters and receptors
inhibitory effects.212 Multiple serotonergic receptor agonists have
Purinergic P2X receptors are ligand (ATP)-gated nonselective cation been shown to alleviate indices of pain in animal models of
channels located on neurons, Schwann cells, and microglia.36 There diabetes, including agonists or indirect activators of 5-HT1A,162
is increased P2X2R and P2X3R expression and current density in 5HT2A/C,235 and 5HT7362 receptors. Although spinal expression
the DRG of STZ-diabetic rats and mice and increased P2X4R of 5HT2A receptors is unchanged by diabetes,265 efficacy of
expression by satellite glial cells.346 Increased gene expression of duloxetine, a selective serotonin reuptake inhibitor approved for
P2X3R in diabetic rats has been linked to demethylation of the p2x3r use against pain in diabetic patients,9 is through activation of
gene.406 Involvement of P2XR in pain is suggested by reports that these receptors in the spinal cord of diabetic rats.235 Muscarinic
peripheral and intrathecal delivery of antagonists alleviated tactile M2 receptors are increased in the spinal cord of diabetic rats55
and thermal hyperalgesia in diabetic animals.233,346,393,406 Increased and facilitate the antinociceptive actions of cholinergic agonists
activity of receptors located at synapses and on adjacent microglia operating through GABAB receptors.54
has the potential to enhance primary afferent input and spinal Although increased expression and/or activity of ion channels
sensitization of sensory processing. and receptors may lead to hypersensitive or destabilized primary
The spinal cord of diabetic rodents shows increased glutamate afferents and inappropriate electrical activity, the contribution of
and substance P ligand binding178,207 and increased mRNA for peripherally drive to pain in diabetes may be offset by progression
subunits of glutamatergic NMDA and AMPA receptors.353 The to a degenerative neuropathy phenotype. No matter how
Figure 4. A plethora of potential pain precipitating pathways? Animal models of diabetic neuropathy develop molecular and functional disorders throughout the
neuraxis that have the potential to generate or amplify pain and thus serve as targets for therapeutic intervention. The majority of these disorders have yet to be
validated in the human condition. TRP, transient receptor potential.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
S76
·
N.A. Calcutt 161 (2020) S65–S86 PAIN®
electrically active a primary afferent becomes, it is effectively silent 10).112 Refinement of models of neuropathic pain and of assays
if it cannot release adequate neurotransmitter at the spinal dorsal towards those that incorporate more complex cognitive functions
horn. For example, in diabetic rats, there is an early reduction in may improve the predictive value of preclinical studies87 and have
the synthesis,88 transport,354 and stimulus-evoked spinal release begun to be used in models of diabetes.376 There is also
of both neuropeptide44,118 and amino acid224 excitatory neuro- a growing appreciation that pain in diabetic patients falls into
transmitters that are concurrent with enhanced pain-associated distinct subtypes,20 potentially reflecting different dominant
behavior in the same animals. There is also a progression from pathogenic mechanisms and thus responsiveness to targeted
increased, to loss of, synaptic markers in the spinal cord of therapeutic interventions. Drugs not statistically effective against
diabetic rats.165,210 Pain generator sites may evolve over time, pain in an unrefined cohort have been shown to be effective in
with initial pain driven by a hypersensitive or hyperactive periphery a subgroup defined by pain mechanism,85 and clinical trial
but progressing to spinal and CNS generator/maintenance sites designs are beginning to incorporate patient stratification based
as the primary afferent input fades with first neurochemical, then on the likely mechanism of both pain generation and the agent
physical, degeneration. Longitudinal clinical studies in subjects under investigation.315 Together, these advances may allow the
with diabetic neuropathy, using such techniques as micro- identification and development of translatable therapies that are
neurography,315 RDD,225 and magnetic resonance imaging,312 tested against the mechanistically appropriate population and
to track activity of generator sites over time may be of value. open an encouraging gateway into the world of personalized
medicine.
2.6. Summary
A PubMed search at 7 PM Pacific Standard TIme on February 14, Conflict of interest statement
2020, using the search phrase “diabetic neuropathy treatment” N.A. Calcutt is a paid consultant for Torrey Industries Inc and has
returned 17,238 hits—a lot of words for a condition with no equity interest in WinSanTor, Inc and CerSci Inc, these being
approved therapy. It is challenging to reconcile the plethora of commercial entities that may potentially benefit from research
biochemical and molecular changes in the nervous system of and insights in this area. The terms of these arrangements have
diabetic rodents described previously with their mildly dysfunc- been reviewed and approved by the University of California, San
tional neuropathy phenotype and limited nerve pathology. It is Diego, in accordance with its conflict of interest policies.
also remarkable that so many highly selective interventions
against specific pathways are completely effective in preventing
or reversing indices of neuropathy and pain, despite presumably Acknowledgements
continued operation of multiple other documented pathways.
The sheer volume of effective interventions must raise concerns The author thanks Dr. Corinne Jolivalt for helpful discussions and
about the relevance of preclinical models and assays to the appropriately brutal editing.
clinical condition. Experimental models of diabetic neuropathy Supported by NIH award DK104512.
are perhaps best viewed as hypothesis-generating tools that offer
a veritable cornucopia of potential pathogenic events and Article history:
plausible targets for therapeutic intervention against neuro- Received 5 March 2020
degeneration and pain (Fig. 4). Relatively few of the biochemical Received in revised form 5 May 2020
and molecular changes described previously have been con- Accepted 7 May 2020
firmed in humans and, when they are, it is not easy to determine
whether altered protein expression and/or activity has physio-
logical and pathological consequences or is itself a consequence References
of neuropathy. [1] Abbate SL, Atkinson MB, Breuer AC. Amount and speed of fast axonal
The challenge before us remains the same as it was over half transport in diabetes. Diabetes 1991;40:111–17.
[2] Abbott CA, Malik RA, van Ross ER, Kulkarni J, Boulton AJ. Prevalence
a century ago following the advent of ARIs189—namely, trans- and characteristics of painful diabetic neuropathy in a large community-
lating mechanisms and interventions identified in preclinical based diabetic population in the U.K. Diabetes Care 2011;34:2220–4.
models of diabetes into viable therapies to prevent and reverse [3] Aghanoori MR, Smith DR, Roy Chowdhury S, Sabbir MG, Calcutt NA,
diabetic neuropathy and neuropathic pain. Agents that appear Fernyhough P. Insulin prevents aberrant mitochondrial phenotype in
sensory neurons of type 1 diabetic rats. Exp Neurol 2017;297:148–57.
promising in preclinical studies have consistently failed in clinical
[4] Aghanoori MR, Smith DR, Shariati-Ievari S, Ajisebutu A, Nguyen A,
trials against degenerative diabetic neuropathy, and there has Desmond F, Jesus CHA, Zhou X, Calcutt NA, Aliani M, Fernyhough P.
been plenty of subsequent finger pointing, from allegations of Insulin-like growth factor-1 activates AMPK to augment mitochondrial
poor drug design and unrepresentative animal models to flawed function and correct neuronal metabolism in sensory neurons in type 1
clinical trial designs and outcome measures.106,221 Encourag- diabetes. Mol Metab 2019;20:149–65.
[5] Aghdam AM, Shahabi P, Karimi-Sales E, Ghiasi R, Sadigh-Eteghad S,
ingly, recognition and analysis of prior failings221 has prompted Mahmoudi J, Alipour MR. Swimming exercise induced reversed
the development of focused in vitro models to aid mechanistic expression of miR-96 and its target gene NaV1.3 in diabetic peripheral
and drug screening studies,117 animal models that more closely neuropathy in rats. Chin J Physiol 2018;61:124–9.
resemble the human diabetic condition,402 refinement of clinical [6] Ajroud-Driss S, Christiansen M, Allen JA, Kessler JA. Phase 1/2 open-
label dose-escalation study of plasmid DNA expressing two isoforms of
protocols,32 and introduction of outcome measures such as
hepatocyte growth factor in patients with painful diabetic peripheral
nerve fiber density in the skin and cornea that highlight small fiber neuropathy. Mol Ther 2013;21:1279–86.
neuropathy.221,274 In contrast to degenerative neuropathy, there [7] Akude E, Zherebitskaya E, Chowdhury SK, Smith DR, Dobrowsky RT,
are a number of therapies approved by regulatory agencies to Fernyhough P. Diminished superoxide generation is associated with
alleviate pain in diabetes and others that are used off-label.9,18 respiratory chain dysfunction and changes in the mitochondrial
proteome of sensory neurons from diabetic rats. Diabetes 2011;60:
However, it is notable that none were developed to target 288–97.
diabetes-specific mechanisms, although efficacy is both un- [8] Alam U, Jeziorska M, Petropoulos IN, Asghar O, Fadavi H, Ponirakis G,
predictable and restricted to small subsets of patients (NNT . 5- Marshall A, Tavakoli M, Boulton AJM, Efron N, Malik RA. Diagnostic
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
September 2020
· Volume 161
· Number 9
· Supplement 1 www.painjournalonline.com S77
utility of corneal confocal microscopy and intra-epidermal nerve fibre [27] Biessels GJ, Despa F. Cognitive decline and dementia in diabetes
density in diabetic neuropathy. PLoS One 2017;12:e0180175. mellitus: mechanisms and clinical implications. Nat Rev Endocrinol
[9] Alam U, Sloan G, Tesfaye S. Treating pain in diabetic neuropathy: 2018;14:591–604.
current and developmental drugs. Drugs 2020;80:363–84. [28] Biessels GJ, ter Laak MP, Hamers FP, Gispen WH. Neuronal Ca21
[10] Ali S, Driscoll HE, Newton VL, Gardiner NJ. Matrix metalloproteinase-2 is disregulation in diabetes mellitus. Eur J Pharmacol 2002;447:201–9.
downregulated in sciatic nerve by streptozotocin induced diabetes and/ [29] Blesneac I, Themistocleous AC, Fratter C, Conrad LJ, Ramirez JD, Cox
or treatment with minocycline: implications for nerve regeneration. Exp JJ, Tesfaye S, Shillo PR, Rice ASC, Tucker SJ, Bennett DLH. Rare
Neurol 2014;261:654–65. NaV1.7 variants associated with painful diabetic peripheral neuropathy.
[11] Alsaloum M, Estacion M, Almomani R, Gerrits MM, Bonhof GJ, Ziegler PAIN 2018;159:469–80.
D, Malik R, Ferdousi M, Lauria G, Merkies IS, Faber CG, Dib-Hajj S, [30] Bonhof GJ, Strom A, Puttgen S, Ringel B, Bruggemann J, Bodis K,
Waxman SG, Propane Study G. A gain-of-function sodium channel Mussig K, Szendroedi J, Roden M, Ziegler D. Patterns of cutaneous
beta2-subunit mutation in painful diabetic neuropathy. Mol Pain 2019; nerve fibre loss and regeneration in type 2 diabetes with painful and
15:1744806919849802. painless polyneuropathy. Diabetologia 2017;60:2495–503.
[12] Amani S, Fatima S. Glycation with fructose: the bitter side of nature’s [31] Braffett BH, Gubitosi-Klug RA, Albers JW, Feldman EL, Martin CL, White
own sweetner. Curr Diabetes Rev 2020. doi: 10.2174/ NH, Orchard TJ, Lopes-Virella M, Lachin JM, Pop-Busui R; Group DES.
1389450121666200204115751 [epub ahead of print]. Risk factors for diabetic peripheral neuropathy and cardiovascular
[13] Andersen ST, Witte DR, Dalsgaard EM, Andersen H, Nawroth P, autonomic neuropathy in the diabetes control and complications trial/
Fleming T, Jensen TM, Finnerup NB, Jensen TS, Lauritzen T, Feldman epidemiology of diabetes interventions and complications (DCCT/EDIC)
EL, Callaghan BC, Charles M. Risk factors for incident diabetic study. Diabetes 2020;69:1000–10.
polyneuropathy in a cohort with screen-detected type 2 diabetes [32] Bril V. The perfect clinical trial. Int Rev Neurobiol 2016;127:27–41.
followed for 13 years: ADDITION-Denmark. Diabetes Care 2018;41: [33] Brines M, Dunne AN, van Velzen M, Proto PL, Ostenson CG, Kirk RI,
1068–75. Petropoulos IN, Javed S, Malik RA, Cerami A, Dahan A. ARA 290,
[14] Anderson NJ, King MR, Delbruck L, Jolivalt CG. Role of insulin signaling a nonerythropoietic peptide engineered from erythropoietin, improves
impairment, adiponectin and dyslipidemia in peripheral and central metabolic control and neuropathic symptoms in patients with type 2
neuropathy in mice. Dis Model Mech 2014;7:625–33. diabetes. Mol Med 2015;20:658–66.
[15] Andersson DA, Filipovic MR, Gentry C, Eberhardt M, Vastani N, Leffler A, [34] Brown MJ, Martin JR, Asbury AK. Painful diabetic neuropathy. A
Reeh P, Bevan S. Streptozotocin stimulates the ion channel TRPA1 morphometric study. Arch Neurol 1976;33:164–71.
directly: involvement of peroxynitrite. J Biol Chem 2015;290:15185–96. [35] Brussee V, Cunningham FA, Zochodne DW. Direct insulin signaling of
[16] Apfel SC. Nerve growth factor for the treatment of diabetic neuropathy: neurons reverses diabetic neuropathy. Diabetes 2004;53:1824–30.
what went wrong, what went right, and what does the future hold? Int [36] Burnstock G. Purines and sensory nerves. Handb Exp Pharmacol 2009:
Rev Neurobiol 2002;50:393–413. 333–92.
[17] Araya EI, Nones CFM, Ferreira LEN, Kopruszinski CM, Cunha JMD, [37] Calcutt NA. Modeling diabetic sensory neuropathy in rats. Methods Mol
Chichorro JG. Role of peripheral and central TRPV1 receptors in facial Med 2004;99:55–65.
heat hyperalgesia in streptozotocin-induced diabetic rats. Brain Res [38] Calcutt NA, Allendoerfer KL, Mizisin AP, Middlemas A, Freshwater JD,
2017;1670:146–55.
Burgers M, Ranciato R, Delcroix JD, Taylor FR, Shapiro R, Strauch K,
[18] Ardeleanu V, Toma A, Pafili K, Papanas N, Motofei I, Diaconu CC, Rizzo
Dudek H, Engber TM, Galdes A, Rubin LL, Tomlinson DR. Therapeutic
M, Stoian AP. Current pharmacological treatment of painful diabetic
efficacy of sonic hedgehog protein in experimental diabetic neuropathy.
neuropathy: a narrative review. Medicina (Kaunas) 2020;56:pii: E25.
J Clin Invest 2003;111:507–14.
[19] Ashrafi G, Wu Z, Farrell RJ, Ryan TA. GLUT4 mobilization supports
[39] Calcutt NA, Chaplan SR. Spinal pharmacology of tactile allodynia in
energetic demands of active synapses. Neuron 2017;93:606–15.e3.
diabetic rats. Br J Pharmacol 1997;122:1478–82.
[20] Baron R, Maier C, Attal N, Binder A, Bouhassira D, Cruccu G, Finnerup
[40] Calcutt NA, Freshwater JD, Mizisin AP. Prevention of sensory disorders
NB, Haanpaa M, Hansson P, Hullemann P, Jensen TS, Freynhagen R,
in diabetic Sprague-Dawley rats by aldose reductase inhibition or
Kennedy JD, Magerl W, Mainka T, Reimer M, Rice AS, Segerdahl M,
treatment with ciliary neurotrophic factor. Diabetologia 2004;47:
Serra J, Sindrup S, Sommer C, Tolle T, Vollert J, Treede RD. Peripheral
718–24.
neuropathic pain: a mechanism-related organizing principle based on
[41] Calcutt NA, Jorge MC, Yaksh TL, Chaplan SR. Tactile allodynia and
sensory profiles. PAIN 2017;158:261–72.
formalin hyperalgesia in streptozotocin-diabetic rats: effects of insulin,
[21] Barragan-Iglesias P, Kuhn J, Vidal-Cantu GC, Salinas-Abarca AB,
Granados-Soto V, Dussor GO, Campbell ZT, Price TJ. Activation of the aldose reductase inhibition and lidocaine. PAIN 1996;68:293–9.
[42] Calcutt NA, Muir D, Powell HC, Mizisin AP. Reduced ciliary
integrated stress response in nociceptors drives methylglyoxal-induced
pain. PAIN 2019;160:160–71. neuronotrophic factor-like activity in nerves from diabetic or galactose-
[22] Bauer CS, Nieto-Rostro M, Rahman W, Tran-Van-Minh A, Ferron L, fed rats. Brain Res 1992;575:320–4.
Douglas L, Kadurin I, Sri Ranjan Y, Fernandez-Alacid L, Millar NS, [43] Calcutt NA, Smith DR, Frizzi K, Sabbir MG, Chowdhury SK, Mixcoatl-
Dickenson AH, Lujan R, Dolphin AC. The increased trafficking of the Zecuatl T, Saleh A, Muttalib N, Van der Ploeg R, Ochoa J, Gopaul A,
calcium channel subunit alpha2delta-1 to presynaptic terminals in Tessler L, Wess J, Jolivalt CG, Fernyhough P. Selective antagonism of
neuropathic pain is inhibited by the alpha2delta ligand pregabalin. muscarinic receptors is neuroprotective in peripheral neuropathy. J Clin
J Neurosci 2009;29:4076–88. Invest 2017;127:608–22.
[23] Beiswenger KK, Calcutt NA, Mizisin AP. Epidermal nerve fiber [44] Calcutt NA, Stiller C, Gustafsson H, Malmberg AB. Elevated substance-
quantification in the assessment of diabetic neuropathy. Acta P-like immunoreactivity levels in spinal dialysates during the formalin test
Histochem 2008;110:351–62. in normal and diabetic rats. Brain Res 2000;856:20–7.
[24] Bestall SM, Hulse RP, Blackley Z, Swift M, Ved N, Paton K, Beazley- [45] Cameron NE, Cotter MA. Effects of protein kinase Cbeta inhibition on
Long N, Bates DO, Donaldson LF. Sensory neuronal sensitisation neurovascular dysfunction in diabetic rats: interaction with oxidative
occurs through HMGB-1-RAGE and TRPV1 in high-glucose conditions. stress and essential fatty acid dysmetabolism. Diabetes Metab Res Rev
J Cell Sci 2018;131. 2002;18:315–23.
[25] Bierhaus A, Fleming T, Stoyanov S, Leffler A, Babes A, Neacsu C, Sauer [46] Campos MM, Ongali B, Thibault G, Neugebauer W, Couture R.
SK, Eberhardt M, Schnolzer M, Lasitschka F, Neuhuber WL, Kichko TI, Autoradiographic distribution and alterations of kinin B(2) receptors in
Konrade I, Elvert R, Mier W, Pirags V, Lukic IK, Morcos M, Dehmer T, the brain and spinal cord of streptozotocin-diabetic rats. Synapse 2005;
Rabbani N, Thornalley PJ, Edelstein D, Nau C, Forbes J, Humpert PM, 58:184–92.
Schwaninger M, Ziegler D, Stern DM, Cooper ME, Haberkorn U, [47] Carroll SL, Byer SJ, Dorsey DA, Watson MA, Schmidt RE. Ganglion-
Brownlee M, Reeh PW, Nawroth PP. Methylglyoxal modification of specific patterns of diabetes-modulated gene expression are
Nav1.8 facilitates nociceptive neuron firing and causes hyperalgesia in established in prevertebral and paravertebral sympathetic ganglia prior
diabetic neuropathy. Nat Med 2012;18:926–33. to the development of neuroaxonal dystrophy. J Neuropathol Exp Neurol
[26] Biessels GJ, Bril V, Calcutt NA, Cameron NE, Cotter MA, Dobrowsky R, 2004;63:1144–54.
Feldman EL, Fernyhough P, Jakobsen J, Malik RA, Mizisin AP, Oates PJ, [48] Casellini CM, Barlow PM, Rice AL, Casey M, Simmons K, Pittenger G,
Obrosova IG, Pop-Busui R, Russell JW, Sima AA, Stevens MJ, Schmidt Bastyr EJ III, Wolka AM, Vinik AI. A 6-month, randomized, double-
RE, Tesfaye S, Veves A, Vinik AI, Wright DE, Yagihashi S, Yorek MA, masked, placebo-controlled study evaluating the effects of the protein
Ziegler D, Zochodne DW. Phenotyping animal models of diabetic kinase C-beta inhibitor ruboxistaurin on skin microvascular blood flow
neuropathy: a consensus statement of the diabetic neuropathy study and other measures of diabetic peripheral neuropathy. Diabetes Care
group of the EASD (Neurodiab). J Peripher Nerv Syst 2014;19:77–87. 2007;30:896–902.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
S78
·
N.A. Calcutt 161 (2020) S65–S86 PAIN®
[49] Chandrasekaran K, Anjaneyulu M, Choi J, Kumar P, Salimian M, Ho CY, [71] Coudore-Civiale M, Courteix C, Boucher M, Fialip J, Eschalier A.
Russell JW. Role of mitochondria in diabetic peripheral neuropathy: Evidence for an involvement of tachykinins in allodynia in streptozocin-
influencing the NAD(1)-dependent SIRT1-PGC-1alpha-TFAM pathway. induced diabetic rats. Eur J Pharmacol 2000;401:47–53.
Int Rev Neurobiol 2019;145:177–209. [72] Cowell RM, Russell JW. Nitrosative injury and antioxidant therapy in the
[50] Chavez JC, Almhanna K, Berti-Mattera LN. Transient expression of management of diabetic neuropathy. J Investig Med 2004;52:33–44.
hypoxia-inducible factor-1 alpha and target genes in peripheral nerves [73] Craner MJ, Klein JP, Renganathan M, Black JA, Waxman SG. Changes
from diabetic rats. Neurosci Lett 2005;374:179–82. of sodium channel expression in experimental painful diabetic
[51] Chemaly ER, Troncone L, Lebeche D. SERCA control of cell death and neuropathy. Ann Neurol 2002;52:786–92.
survival. Cell Calcium 2018;69:46–61. [74] Cui YY, Xu H, Wu HH, Qi J, Shi J, Li YQ. Spatio-temporal expression and
[52] Chen DK, Frizzi KE, Guernsey LS, Ladt K, Mizisin AP, Calcutt NA. functional involvement of transient receptor potential vanilloid 1 in
Repeated monitoring of corneal nerves by confocal microscopy as an diabetic mechanical allodynia in rats. PLoS One 2014;9:e102052.
index of peripheral neuropathy in type-1 diabetic rodents and the effects [75] Cunha JM, Jolivalt CG, Ramos KM, Gregory JA, Calcutt NA, Mizisin AP.
of topical insulin. J Peripher Nerv Syst 2013;18:306–15. Elevated lipid peroxidation and DNA oxidation in nerve from diabetic
[53] Chen J, Li C, Liu W, Yan B, Hu X, Yang F. miRNA-155 silencing reduces rats: effects of aldose reductase inhibition, insulin, and neurotrophic
sciatic nerve injury in diabetic peripheral neuropathy. J Mol Endocrinol factors. Metabolism 2008;57:873–81.
2019;63:227–38. [76] D’Amato C, Morganti R, Di Gennaro F, Greco C, Marfia GA, Spallone V.
[54] Chen SR, Pan HL. Spinal GABAB receptors mediate antinociceptive A novel association between nondipping and painful diabetic
actions of cholinergic agents in normal and diabetic rats. Brain Res polyneuropathy. Diabetes Care 2014;37:2640–2.
2003;965:67–74. [77] Dalmolin GD, Silva CR, Rigo FK, Gomes GM, Cordeiro Mdo N,
[55] Chen SR, Pan HL. Up-regulation of spinal muscarinic receptors and Richardson M, Silva MA, Prado MA, Gomez MV, Ferreira J.
increased antinociceptive effect of intrathecal muscarine in diabetic rats. Antinociceptive effect of Brazilian armed spider venom toxin Tx3-3 in
J Pharmacol Exp Ther 2003;307:676–81. animal models of neuropathic pain. PAIN 2011;152:2224–32.
[56] Cheng C, Kobayashi M, Martinez JA, Ng H, Moser JJ, Wang X, Singh V, [78] Daugherty DJ, Marquez A, Calcutt NA, Schubert D. A novel curcumin
Fritzler MJ, Zochodne DW. Evidence for epigenetic regulation of gene derivative for the treatment of diabetic neuropathy. Neuropharmacology
expression and function in chronic experimental diabetic neuropathy. 2018;129:26–35.
J Neuropathol Exp Neurol 2015;74:804–17. [79] Daulhac L, Maffre V, Mallet C, Etienne M, Privat AM, Kowalski-Chauvel
[57] Cheng C, Zochodne DW. Sensory neurons with activated caspase-3 A, Seva C, Fialip J, Eschalier A. Phosphorylation of spinal N-methyl-d-
survive long-term experimental diabetes. Diabetes 2003;52:2363–71. aspartate receptor NR1 subunits by extracellular signal-regulated kinase
[58] Cheng HT, Dauch JR, Porzio MT, Yanik BM, Hsieh W, Smith AG, in dorsal horn neurons and microglia contributes to diabetes-induced
Singleton JR, Feldman EL. Increased axonal regeneration and swellings painful neuropathy. Eur J Pain 2011;15:169.e1–e12.
in intraepidermal nerve fibers characterize painful phenotypes of diabetic [80] Davidson E, Coppey L, Lu B, Arballo V, Calcutt NA, Gerard C, Yorek M.
neuropathy. J Pain 2013;14:941–7. The roles of streptozotocin neurotoxicity and neutral endopeptidase in
[59] Cheung A, Podgorny P, Martinez JA, Chan C, Toth C. Epidermal axonal murine experimental diabetic neuropathy. Exp Diabetes Res 2009;
swellings in painful and painless diabetic peripheral neuropathy. Muscle 2009:431980.
[81] Davidson EP, Coppey LJ, Calcutt NA, Oltman CL, Yorek MA. Diet-
Nerve 2015;51:505–13.
induced obesity in Sprague-Dawley rats causes microvascular and
[60] Chowdhury SK, Smith DR, Fernyhough P. The role of aberrant
neural dysfunction. Diabetes Metab Res Rev 2010;26:306–18.
mitochondrial bioenergetics in diabetic neuropathy. Neurobiol Dis
[82] Davis TM, Yeap BB, Davis WA, Bruce DG. Lipid-lowering therapy and
2013;51:56–65.
peripheral sensory neuropathy in type 2 diabetes: the Fremantle
[61] Chowdhury SK, Zherebitskaya E, Smith DR, Akude E, Chattopadhyay S,
Diabetes Study. Diabetologia 2008;51:562–6.
Jolivalt CG, Calcutt NA, Fernyhough P. Mitochondrial respiratory chain
[83] de Anda-Jauregui G, Guo K, McGregor BA, Feldman EL, Hur J. Pathway
dysfunction in dorsal root ganglia of streptozotocin-induced diabetic rats
crosstalk perturbation network modeling for identification of connectivity
and its correction by insulin treatment. Diabetes 2010;59:1082–91.
changes induced by diabetic neuropathy and pioglitazone. BMC Syst
[62] Christianson JA, Riekhof JT, Wright DE. Restorative effects of
Biol 2019;13:1.
neurotrophin treatment on diabetes-induced cutaneous axon loss in
[84] de la Hoz CL, Cheng C, Fernyhough P, Zochodne DW. A model of
mice. Exp Neurol 2003;179:188–99.
chronic diabetic polyneuropathy: benefits from intranasal insulin are
[63] Ciccacci C, Latini A, Greco C, Politi C, D’Amato C, Lauro D, Novelli G,
modified by sex and RAGE deletion. Am J Physiol Endocrinol Metab
Borgiani P, Spallone V. Association between a MIR499A polymorphism
2017;312:E407–19.
and diabetic neuropathy in type 2 diabetes. J Diabetes Complicat 2018; [85] Demant DT, Lund K, Vollert J, Maier C, Segerdahl M, Finnerup NB,
32:11–17. Jensen TS, Sindrup SH. The effect of oxcarbazepine in peripheral
[64] Ciccacci C, Morganti R, Di Fusco D, D’Amato C, Cacciotti L, Greco C, neuropathic pain depends on pain phenotype: a randomised, double-
Rufini S, Novelli G, Sangiuolo F, Marfia GA, Borgiani P, Spallone V. blind, placebo-controlled phenotype-stratified study. PAIN 2014;155:
Common polymorphisms in MIR146a, MIR128a and MIR27a genes 2263–73.
contribute to neuropathy susceptibility in type 2 diabetes. Acta Diabetol [86] Dias FC, Alves VS, Matias DO, Figueiredo CP, Miranda ALP, Passos GF,
2014;51:663–71. Costa R. The selective TRPV4 channel antagonist HC-067047
[65] Cimen SG, Cimen S, Kessaris N, Kahveci E, Tuzuner A. Challenges of attenuates mechanical allodynia in diabetic mice. Eur J Pharmacol
pancreas transplantation in developing countries, exploring the Turkey 2019;856:172408.
example. World J Transpl 2019;9:158–64. [87] Dickenson AH, Patel R. Translational issues in precision medicine in
[66] Ciobanu AC, Selescu T, Gasler I, Soltuzu L, Babes A. Glycolytic neuropathic pain. Can J Pain 2020;4:30–8.
metabolite methylglyoxal inhibits cold and menthol activation of the [88] Diemel LT, Brewster WJ, Fernyhough P, Tomlinson DR. Expression of
transient receptor potential melastatin type 8 channel. J Neurosci Res neuropeptides in experimental diabetes; effects of treatment with nerve
2016;94:282–94. growth factor or brain-derived neurotrophic factor. Brain Res Mol Brain
[67] Conti G, Scarpini E, Baron P, Livraghi S, Tiriticco M, Bianchi R, Vedeler Res 1994;21:171–5.
C, Scarlato G. Macrophage infiltration and death in the nerve during the [89] Dogrul A, Gul H, Yesilyurt O, Ulas UH, Yildiz O. Systemic and spinal
early phases of experimental diabetic neuropathy: a process administration of etanercept, a tumor necrosis factor alpha inhibitor,
concomitant with endoneurial induction of IL-1beta and p75NTR. blocks tactile allodynia in diabetic mice. Acta Diabetol 2011;48:135–42.
J Neurol Sci 2002;195:35–40. [90] Doupis J, Lyons TE, Wu S, Gnardellis C, Dinh T, Veves A. Microvascular
[68] Coppey L, Davidson E, Shevalye H, Obrosov A, Yorek M. Effect of early reactivity and inflammatory cytokines in painful and painless peripheral
and late interventions with dietary oils on vascular and neural diabetic neuropathy. J Clin Endocrinol Metab 2009;94:2157–63.
complications in a type 2 diabetic rat model. J Diabetes Res 2019; [91] Dubovy P, Jancalek R, Kubek T. Role of inflammation and cytokines in
2019:5020465. peripheral nerve regeneration. Int Rev Neurobiol 2013;108:173–206.
[69] Coppey LJ, Davidson EP, Obrosov A, Yorek MA. Enriching the diet with [92] Dull MM, Riegel K, Tappenbeck J, Ries V, Strupf M, Fleming T, Sauer
menhaden oil improves peripheral neuropathy in streptozotocin- SK, Namer B. Methylglyoxal causes pain and hyperalgesia in human
induced type 1 diabetic rats. J Neurophysiol 2015;113:701–8. through C-fiber activation. PAIN 2019;160:2497–507.
[70] Coppey LJ, Holmes A, Davidson EP, Yorek MA. Partial replacement with [93] Duraikannu A, Krishnan A, Chandrasekhar A, Zochodne DW. Beyond
menhaden oil improves peripheral neuropathy in high-fat-fed low-dose trophic factors: exploiting the intrinsic regenerative properties of adult
streptozotocin type 2 diabetic rat. J Nutr Metab 2012;2012:950517. neurons. Front Cell Neurosci 2019;13:128.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
September 2020
· Volume 161
· Number 9
· Supplement 1 www.painjournalonline.com S79
[94] Duran-Jimenez B, Dobler D, Moffatt S, Rabbani N, Streuli CH, [118] Garrett NE, Malcangio M, Dewhurst M, Tomlinson DR. alpha-Lipoic acid
Thornalley PJ, Tomlinson DR, Gardiner NJ. Advanced glycation end corrects neuropeptide deficits in diabetic rats via induction of trophic
products in extracellular matrix proteins contribute to the failure of support. Neurosci Lett 1997;222:191–4.
sensory nerve regeneration in diabetes. Diabetes 2009;58:2893–903. [119] Gastol J, Kapusta P, Polus A, Pitera E, Biela M, Wolkow P, Pawlinski L,
[95] Dvornik E, Simard-Duquesne N, Krami M, Sestanj K, Gabbay KH, Kiec-Wilk B. Epigenetic mechanism in search for the pathomechanism
Kinoshita JH, Varma SD, Merola LO. Polyol accumulation in of diabetic neuropathy development in diabetes mellitus type 1 (T1DM).
galactosemic and diabetic rats: control by an aldose reductase Endocrine 2020;68:235–40.
inhibitor. Science 1973;182:1146–8. [120] Geraldes P, King GL. Activation of protein kinase C isoforms and its
[96] Dyck PJ, Karnes JL, Daube J, O’Brien P, Service FJ. Clinical and impact on diabetic complications. Circ Res 2010;106:1319–31.
neuropathological criteria for the diagnosis and staging of diabetic [121] Gibbons CH, Freeman R. Treatment-induced neuropathy of diabetes:
polyneuropathy. Brain 1985;108(pt 4):861–80. an acute, iatrogenic complication of diabetes. Brain 2015;138(pt 1):
[97] Dyck PJ, Lais A, Karnes JL, O’Brien P, Rizza R. Fiber loss is primary and 43–52.
multifocal in sural nerves in diabetic polyneuropathy. Ann Neurol 1986; [122] Goncalves NP, Vaegter CB, Andersen H, Ostergaard L, Calcutt NA,
19:425–39. Jensen TS. Schwann cell interactions with axons and microvessels in
[98] Eaton SE, Harris ND, Ibrahim S, Patel KA, Selmi F, Radatz M, Ward JD, diabetic neuropathy. Nat Rev Neurol 2017;13:135–47.
Tesfaye S. Increased sural nerve epineurial blood flow in human subjects [123] Green DR, Llambi F. Cell death signaling. Cold Spring Harb Perspect
with painful diabetic neuropathy. Diabetologia 2003;46:934–9. Biol 2015;7:pii: a006080.
[99] Eberhardt MJ, Filipovic MR, Leffler A, de la Roche J, Kistner K, Fischer [124] Greene DA, Lattimer SA, Sima AA. Are disturbances of sorbitol,
MJ, Fleming T, Zimmermann K, Ivanovic-Burmazovic I, Nawroth PP, phosphoinositide, and Na1-K1-ATPase regulation involved in
Bierhaus A, Reeh PW, Sauer SK. Methylglyoxal activates nociceptors pathogenesis of diabetic neuropathy? Diabetes 1988;37:688–93.
through transient receptor potential channel A1 (TRPA1) a possible [125] Griggs RB, Santos DF, Laird DE, Doolen S, Donahue RR, Wessel CR, Fu
mechanism of metabolic neuropathies. J Biol Chem 2012;287: W, Sinha GP, Wang P, Zhou J, Brings S, Fleming T, Nawroth PP, Susuki
28291–306. K, Taylor BK. Methylglyoxal and a spinal TRPA1-AC1-Epac cascade
[100] Eid S, Sas KM, Abcouwer SF, Feldman EL, Gardner TW, Pennathur S, facilitate pain in the db/db mouse model of type 2 diabetes. Neurobiol
Fort PE. New insights into the mechanisms of diabetic complications: Dis 2019;127:76–86.
[126] Grisold A, Callaghan BC, Feldman EL. Mediators of diabetic
role of lipids and lipid metabolism. Diabetologia 2019;62:1539–49.
[101] Eid SA, El Massry M, Hichor M, Haddad M, Grenier J, Dia B, Barakat R, neuropathy: is hyperglycemia the only culprit? Curr Opin Endocrinol
Diabetes Obes 2017;24:103–11.
Boutary S, Chanal J, Aractingi S, Wiesel P, Szyndralewiez C, Azar ST,
[127] Grote CW, Groover AL, Ryals JM, Geiger PC, Feldman EL, Wright DE.
Boitard C, Zaatari G, Eid AA, Massaad C. Targeting the NADPH
Peripheral nervous system insulin resistance in ob/ob mice. Acta
oxidase-4 and liver X receptor signaling axis preserve Schwann cell
Neuropathol Commun 2013;1:15.
integrity in diabetic mice. Diabetes 2020;69:448–64.
[128] Grote CW, Ryals JM, Wright DE. In vivo peripheral nervous system
[102] Eisenstein M. Sangamo’s lead zinc-finger therapy flops in diabetic
insulin signaling. J Peripher Nerv Syst 2013;18:209–19.
neuropathy. Nat Biotechnol 2012;30:121–3.
[129] Gu Y, Qiu ZL, Liu DZ, Sun GL, Guan YC, Hei ZQ, Li X. Differential gene
[103] Emery SM, Dobrowsky RT. Promoting neuronal tolerance of diabetic
expression profiling of the sciatic nerve in type 1 and type 2 diabetic
stress: modulating molecular chaperones. Int Rev Neurobiol 2016;127:
mice. Biomed Rep 2018;9:291–304.
181–210.
[130] Guo G, Kan M, Martinez JA, Zochodne DW. Local insulin and the rapid
[104] Feng YH, Chen L, Luo Q, Wu M, Chen YH, Shi XH. Involvement of
regrowth of diabetic epidermal axons. Neurobiol Dis 2011;43:414–21.
microRNA-146a in diabetic peripheral neuropathy through the
[131] Guo G, Liu Y, Ren S, Kang Y, Duscher D, Machens HG, Chen Z.
regulation of inflammation. Drug Des Dev Ther 2018;12:171–7. Comprehensive analysis of differentially expressed microRNAs and
[105] Fernyhough P. Mitochondrial dysfunction in diabetic neuropathy:
mRNAs in dorsal root ganglia from streptozotocin-induced diabetic rats.
a series of unfortunate metabolic events. Curr Diab Rep 2015;15:89. PLoS One 2018;13:e0202696.
[106] Fernyhough P, Calcutt NA. New directions in diabetic neuropathy: [132] Gylfadottir SS, Christensen DH, Nicolaisen SK, Andersen H, Callaghan
evolution or extinction? Int Rev Neurobiol 2016;127:229–34. BC, Itani M, Khan KS, Kristensen AG, Nielsen JS, Sindrup SH, Andersen
[107] Fernyhough P, Diemel LT, Brewster WJ, Tomlinson DR. Altered NT, Jensen TS, Thomsen RW, Finnerup NB. Diabetic polyneuropathy
neurotrophin mRNA levels in peripheral nerve and skeletal muscle of and pain, prevalence, and patient characteristics: a cross-sectional
experimentally diabetic rats. J Neurochem 1995;64:1231–7. questionnaire study of 5,514 patients with recently diagnosed type 2
[108] Fernyhough P, Diemel LT, Hardy J, Brewster WJ, Mohiuddin L, diabetes. PAIN 2020;161:574–83.
Tomlinson DR. Human recombinant nerve growth factor replaces [133] Habash T, Saleh A, Roy Chowdhury SK, Smith DR, Fernyhough P. The
deficient neurotrophic support in the diabetic rat. Eur J Neurosci 1995;7: proinflammatory cytokine, interleukin-17A, augments mitochondrial
1107–10. function and neurite outgrowth of cultured adult sensory neurons
[109] Fernyhough P, Diemel LT, Tomlinson DR. Target tissue production and derived from normal and diabetic rats. Exp Neurol 2015;273:177–89.
axonal transport of neurotrophin-3 are reduced in streptozotocin- [134] Hackett AR, Strickland A, Milbrandt J. Disrupting insulin signaling in
diabetic rats. Diabetologia 1998;41:300–6. Schwann cells impairs myelination and induces a sensory neuropathy.
[110] Fernyhough P, Schmidt RE. Neurofilaments in diabetic neuropathy. Int Glia 2020;68:963–78.
Rev Neurobiol 2002;50:115–44. [135] Halim ND, McFate T, Mohyeldin A, Okagaki P, Korotchkina LG, Patel
[111] Fernyhough P, Willars GB, Lindsay RM, Tomlinson DR. Insulin and MS, Jeoung NH, Harris RA, Schell MJ, Verma A. Phosphorylation status
insulin-like growth factor I enhance regeneration in cultured adult rat of pyruvate dehydrogenase distinguishes metabolic phenotypes of
sensory neurones. Brain Res 1993;607:117–24. cultured rat brain astrocytes and neurons. Glia 2010;58:1168–76.
[112] Finnerup NB, Sindrup SH, Jensen TS. The evidence for [136] Hampton KK, Alani SM, Wilson JI, Price DE. Resistance to ischaemic
pharmacological treatment of neuropathic pain. PAIN 2010;150: conduction failure in chronic hypoxaemia and diabetes. J Neurol
573–81. Neurosurg Psychiatry 1989;52:1303–5.
[113] Galosi E, La Cesa S, Di Stefano G, Karlsson P, Fasolino A, Leone C, [137] Han JW, Choi D, Lee MY, Huh YH, Yoon YS. Bone marrow-derived
Biasiotta A, Cruccu G, Truini A. A pain in the skin. Regenerating nerve mesenchymal stem cells improve diabetic neuropathy by direct
sprouts are distinctly associated with ongoing burning pain in patients modulation of both angiogenesis and myelination in peripheral nerves.
with diabetes. Eur J Pain 2018;22:1727–34. Cell Transpl 2016;25:313–26.
[114] Gandhi RA, Marques JL, Selvarajah D, Emery CJ, Tesfaye S. Painful [138] Hangping Z, Ling H, Lijin J, Wenting Z, Xiaoxia L, Qi Z, Xiaoming Z,
diabetic neuropathy is associated with greater autonomic dysfunction Qingchun L, Yiming L, Qian X, Ji H, Bin L, Shuo Z. The preventive effect
than painless diabetic neuropathy. Diabetes Care 2010;33:1585–90. of IL-1beta antagonist on Diabetic Peripheral Neuropathy. Endocr
[115] Ganesh Yerra V, Negi G, Sharma SS, Kumar A. Potential therapeutic Metab Immune Disord Drug Targets 2020. doi: 10.2174/
effects of the simultaneous targeting of the Nrf2 and NF-kappaB 1871530319666191022114139 [epub ahead of print].
pathways in diabetic neuropathy. Redox Biol 2013;1:394–7. [139] Hebert HL, Veluchamy A, Torrance N, Smith BH. Risk factors for
[116] Garcia-Caballero A, Gadotti VM, Chen L, Zamponi GW. A cell-permeant neuropathic pain in diabetes mellitus. PAIN 2017;158:560–8.
peptide corresponding to the cUBP domain of USP5 reverses [140] Hellweg R, Hartung HD. Endogenous levels of nerve growth factor (NGF)
inflammatory and neuropathic pain. Mol Pain 2016;12. are altered in experimental diabetes mellitus: a possible role for NGF in
[117] Gardiner NJ, Freeman OJ. Can diabetic neuropathy be modeled in the pathogenesis of diabetic neuropathy. J Neurosci Res 1990;26:
vitro? Int Rev Neurobiol 2016;127:53–87. 258–67.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
S80
·
N.A. Calcutt 161 (2020) S65–S86 PAIN®
[141] Hinder LM, Murdock BJ, Park M, Bender DE, O’Brien PD, Rumora AE, [164] Jiang Y, Calcutt NA, Ramos KM, Mizisin AP. Novel sites of aldose
Hur J, Feldman EL. Transcriptional networks of progressive diabetic reductase immunolocalization in normal and streptozotocin-diabetic
peripheral neuropathy in the db/db mouse model of type 2 diabetes: an rats. J Peripher Nerv Syst 2006;11:274–85.
inflammatory story. Exp Neurol 2018;305:33–43. [165] Jiang Y, Mizisin AP, Rearden A, Jolivalt CG. Diabetes induces changes
[142] Hiyama H, Yano Y, So K, Imai S, Nagayasu K, Shirakawa H, Nakagawa in ILK, PINCH and components of related pathways in the spinal cord of
T, Kaneko S. TRPA1 sensitization during diabetic vascular impairment rats. Brain Res 2010;1332:100–9.
contributes to cold hypersensitivity in a mouse model of painful diabetic [166] Jimenez-Osorio AS, Gonzalez-Reyes S, Pedraza-Chaverri J. Natural
peripheral neuropathy. Mol Pain 2018;14:1744806918789812. Nrf2 activators in diabetes. Clin Chim Acta 2015;448:182–92.
[143] Hoeijmakers JG, Faber CG, Merkies IS, Waxman SG. Channelopathies, [167] Jolivalt CG, Frizzi KE, Guernsey L, Marquez A, Ochoa J, Rodriguez M,
painful neuropathy, and diabetes: which way does the causal arrow Calcutt NA. Peripheral neuropathy in mouse models of diabetes. Curr
point? Trends Mol Med 2014;20:544–50. Protoc Mouse Biol 2016;6:223–55.
[144] Hong S, Wiley JW. Early painful diabetic neuropathy is associated with [168] Jolivalt CG, Lee CA, Ramos KM, Calcutt NA. Allodynia and hyperalgesia
differential changes in the expression and function of vanilloid receptor in diabetic rats are mediated by GABA and depletion of spinal
1. J Biol Chem 2005;280:618–27. potassium-chloride co-transporters. PAIN 2008;140:48–57.
[145] Hong S, Wiley JW. Altered expression and function of sodium channels [169] Jolivalt CG, Mizisin LM, Nelson A, Cunha JM, Ramos KM, Bonke D,
Calcutt NA. B vitamins alleviate indices of neuropathic pain in diabetic
in large DRG neurons and myelinated A-fibers in early diabetic
rats. Eur J Pharmacol 2009;612:41–7.
neuropathy in the rat. Biochem Biophys Res Commun 2006;339:
[170] Jolivalt CG, Rodriguez M, Wahren J, Calcutt NA. Efficacy of a long-
652–60.
acting C-peptide analogue against peripheral neuropathy in
[146] Hong SS, Morrow TJ, Paulson PE, Isom LL, Wiley JW. Early painful
streptozotocin-diabetic mice. Diabetes Obes Metab 2015;17:781–8.
diabetic neuropathy is associated with differential changes in
[171] Jolivalt CG, Vu Y, Mizisin LM, Mizisin AP, Calcutt NA. Impaired
tetrodotoxin-sensitive and -resistant sodium channels in dorsal root
prosaposin secretion during nerve regeneration in diabetic rats and
ganglion neurons in the rat. J Biol Chem 2004;279:29341–50. protection of nerve regeneration by a prosaposin-derived peptide.
[147] Hu J, Hu X, Kan T. MiR-34c participates in diabetic corneal neuropathy J Neuropathol Exp Neurol 2008;67:702–10.
via regulation of autophagy. Invest Ophthalmol Vis Sci 2019;60:16–25. [172] Juaristi I, Contreras L, Gonzalez-Sanchez P, Perez-Liebana I, Gonzalez-
[148] Hu J, Huang Y, Lin Y, Lin J. Protective effect inhibiting the expression of Moreno L, Pardo B, Del Arco A, Satrustegui J. The response to
miR-181a on the diabetic corneal nerve in a mouse model. Exp Eye Res stimulation in neurons and astrocytes. Neurochem Res 2019;44:
2020;192:107925. 2385–91.
[149] Huang TJ, Price SA, Chilton L, Calcutt NA, Tomlinson DR, Verkhratsky [173] Julius D. TRP channels and pain. Annu Rev Cell Dev Biol 2013;29:
A, Fernyhough P. Insulin prevents depolarization of the mitochondrial 355–84.
inner membrane in sensory neurons of type 1 diabetic rats in the [174] Kalichman MW, Powell HC, Mizisin AP. Reactive, degenerative, and
presence of sustained hyperglycemia. Diabetes 2003;52:2129–36. proliferative Schwann cell responses in experimental galactose and
[150] Hulse RP, Beazley-Long N, Ved N, Bestall SM, Riaz H, Singhal P, human diabetic neuropathy. Acta Neuropathol 1998;95:47–56.
Ballmer Hofer K, Harper SJ, Bates DO, Donaldson LF. Vascular [175] Kalliomaki J, Attal N, Jonzon B, Bach FW, Huizar K, Ratcliffe S, Eriksson
endothelial growth factor-A165b prevents diabetic neuropathic pain and B, Janecki M, Danilov A, Bouhassira D, Group APS. A randomized,
sensory neuronal degeneration. Clin Sci (Lond) 2015;129:741–56. double-blind, placebo-controlled trial of a chemokine receptor 2 (CCR2)
[151] Hur J, O’Brien PD, Nair V, Hinder LM, McGregor BA, Jagadish HV, antagonist in posttraumatic neuralgia. PAIN 2013;154:761–7.
Kretzler M, Brosius FC III, Feldman EL. Transcriptional networks of [176] Kalteniece A, Ferdousi M, Azmi S, Mubita WM, Marshall A, Lauria G,
murine diabetic peripheral neuropathy and nephropathy: common and Faber CG, Soran H, Malik RA. Corneal confocal microscopy detects
distinct gene expression patterns. Diabetologia 2016;59:1297–306. small nerve fibre damage in patients with painful diabetic neuropathy.
[152] Hur J, Sullivan KA, Pande M, Hong Y, Sima AA, Jagadish HV, Kretzler M, Sci Rep 2020;10:3371.
Feldman EL. The identification of gene expression profiles associated [177] Kalteniece A, Ferdousi M, Petropoulos I, Azmi S, Adam S, Fadavi H,
with progression of human diabetic neuropathy. Brain 2011;134(pt 11): Marshall A, Boulton AJM, Efron N, Faber CG, Lauria G, Soran H, Malik
3222–35. RA. Greater corneal nerve loss at the inferior whorl is related to the
[153] Ishii DN. Implication of insulin-like growth factors in the pathogenesis of presence of diabetic neuropathy and painful diabetic neuropathy. Sci
diabetic neuropathy. Brain Res Brain Res Rev 1995;20:47–67. Rep 2018;8:3283.
[154] Ismail CAN, Suppian R, Abd Aziz CB, Haris K, Long I. Increased [178] Kamei J, Ogawa M, Kasuya Y. Development of supersensitivity to
nociceptive responses in streptozotocin-induced diabetic rats and the substance P in the spinal cord of the streptozotocin-induced diabetic
related expression of spinal NR2B subunit of N-Methyl-D-Aspartate rats. Pharmacol Biochem Behav 1990;35:473–5.
receptors. Diabetes Metab J 2019;43:222–35. [179] Kamiya H, Zhang W, Ekberg K, Wahren J, Sima AA. C-Peptide reverses
[155] Jack MM, Ryals JM, Wright DE. Protection from diabetes-induced nociceptive neuropathy in type 1 diabetes. Diabetes 2006;55:3581–7.
peripheral sensory neuropathy—a role for elevated glyoxalase I? Exp [180] Kato N, Nemoto K, Nakanishi K, Morishita R, Kaneda Y, Uenoyama M,
Neurol 2012;234:62–9. Ikeda T, Fujikawa K. Nonviral gene transfer of human hepatocyte growth
[156] Jaiswal S, Mishra S, Torgal SS, Shengule S. Neuroprotective effect of factor improves streptozotocin-induced diabetic neuropathy in rats.
Diabetes 2005;54:846–54.
epalrestat mediated through oxidative stress markers, cytokines and
[181] Kellogg AP, Wiggin TD, Larkin DD, Hayes JM, Stevens MJ, Pop-Busui
TAU protein levels in diabetic rats. Life Sci 2018;207:364–71.
R. Protective effects of cyclooxygenase-2 gene inactivation against
[157] Jakobsen J. Axonal dwindling in early experimental diabetes. I. A study
peripheral nerve dysfunction and intraepidermal nerve fiber loss in
of cross sectioned nerves. Diabetologia 1976;12:539–46.
experimental diabetes. Diabetes 2007;56:2997–3005.
[158] Jakobsen J. Axonal dwindling in early experimental diabetes. II. A study
[182] Kennedy JM, Zochodne DW. Experimental diabetic neuropathy with
of isolated nerve fibres. Diabetologia 1976;12:547–53.
spontaneous recovery: is there irreparable damage? Diabetes 2005;54:
[159] Jayaraj ND, Bhattacharyya BJ, Belmadani AA, Ren D, Rathwell CA,
830–7.
Hackelberg S, Hopkins BE, Gupta HR, Miller RJ, Menichella DM. [183] Kerckhove N, Pereira B, Soriot-Thomas S, Alchaar H, Deleens R, Hieng
Reducing CXCR4-mediated nociceptor hyperexcitability reverses VS, Serra E, Lanteri-Minet M, Arcagni P, Picard P, Lefebvre-Kuntz D,
painful diabetic neuropathy. J Clin Invest 2018;128:2205–25. Maindet C, Mick G, Balp L, Lucas C, Creach C, Letellier M, Martinez V,
[160] Jeffcoate WJ, Vileikyte L, Boyko EJ, Armstrong DG, Boulton AJM. Navez M, Delbrouck D, Kuhn E, Piquet E, Bozzolo E, Brosse C, Lietar B,
Current challenges and opportunities in the prevention and Marcaillou F, Hamdani A, Leroux-Bromberg N, Perier Y, Vergne-Salle P,
management of diabetic foot ulcers. Diabetes Care 2018;41:645–52. Gov C, Delage N, Gillet D, Romettino S, Richard D, Mallet C, Bernard L,
[161] Jessen KR, Mirsky R. The success and failure of the Schwann cell Lambert C, Dubray C, Duale C, Eschalier A. Efficacy and safety of a T-
response to nerve injury. Front Cell Neurosci 2019;13:33. type calcium channel blocker in patients with neuropathic pain: a proof-
[162] Jesus CHA, Redivo DDB, Gasparin AT, Sotomaior BB, de Carvalho MC, of-concept, randomized, double-blind and controlled trial. Eur J Pain
Genaro K, Zuardi AW, Hallak JEC, Crippa JA, Zanoveli JM, da Cunha 2018;22:1321–30.
JM. Cannabidiol attenuates mechanical allodynia in streptozotocin- [184] Khamaneh AM, Alipour MR, Sheikhzadeh Hesari F, Ghadiri Soufi F. A
induced diabetic rats via serotonergic system activation through 5- signature of microRNA-155 in the pathogenesis of diabetic
HT1A receptors. Brain Res 2019;1715:156–64. complications. J Physiol Biochem 2015;71:301–9.
[163] Jia L, Wang L, Chopp M, Li C, Zhang Y, Szalad A, Zhang ZG. MiR-29c/ [185] Khoshnoodi M, Truelove S, Polydefkis M. Effect of diabetes type on
PRKCI regulates axonal growth of dorsal root ganglia neurons under long-term outcome of epidermal axon regeneration. Ann Clin Transl
hyperglycemia. Mol Neurobiol 2018;55:851–8. Neurol 2019;6:2088–96.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
September 2020
· Volume 161
· Number 9
· Supplement 1 www.painjournalonline.com S81
[186] Kim KS, Song YS, Jin J, Joe JH, So BI, Park JY, Fang CH, Kim MJ, Cho [207] Li N, Young MM, Bailey CJ, Smith ME. NMDA and AMPA glutamate
YH, Hwang S, Ro YS, Kim H, Ahn YH, Sung HJ, Sung JJ, Park SH, receptor subtypes in the thoracic spinal cord in lean and obese-diabetic
Lipton SA. Granulocyte-colony stimulating factor as a treatment for ob/ob mice. Brain Res 1999;849:34–44.
diabetic neuropathy in rat. Mol Cell Endocrinol 2015;414:64–72. [208] Li P, Xiong DL, Sun WP, Xu SY. Effects of baicalin on diabetic
[187] Kim S, Maynard JC, Strickland A, Burlingame AL, Milbrandt J. Schwann neuropathic pain involving transient receptor potential vanilloid 1 in the
cell O-GlcNAcylation promotes peripheral nerve remyelination via dorsal root ganglia of rats. Neuroreport 2018;29:1492–8.
attenuation of the AP-1 transcription factor JUN. Proc Natl Acad Sci U [209] Li Y, Ma WG, Xie CQ, Zhang M, Yin XH, Wang FF, Xu J, Shi BY.
S A 2018;115:8019–24. Identification of genes and signaling pathways associated with diabetic
[188] King RH, Llewelyn JG, Thomas PK, Gilbey SG, Watkins PJ. Diabetic neuropathy using a weighted correlation network analysis A consort
neuropathy: abnormalities of Schwann cell and perineurial basal study. Medicine 2016;95:e5443.
laminae. Implications for diabetic vasculopathy. Neuropathol Appl [210] Lin JY, Huang XL, Chen J, Yang ZW, Lin J, Huang S, Peng B.
Neurobiol 1989;15:339–55. Stereological study on the number of synapses in the rat spinal dorsal
[189] Kinoshita JH, Dvornik D, Kraml M, Gabbay KH. The effect of an aldose horn with painful diabetic neuropathy induced by streptozotocin.
reductase inhibitor on the galactose-exposed rabbit lens. Biochim Neuroreport 2017;28:319–24.
Biophys Acta 1968;158:472–5. [211] Liu GS, Shi JY, Lai CL, Hong YR, Shin SJ, Huang HT, Lam HC, Wen ZH,
[190] Koivisto A, Hukkanen M, Saarnilehto M, Chapman H, Kuokkanen K, Wei Hsu KS, Chen CH, Howng SL, Tai MH. Peripheral gene transfer of glial
cell-derived neurotrophic factor ameliorates neuropathic deficits in
H, Viisanen H, Akerman KE, Lindstedt K, Pertovaara A. Inhibiting TRPA1
diabetic rats. Hum Gene Ther 2009;20:715–27.
ion channel reduces loss of cutaneous nerve fiber function in diabetic
[212] Liu P, Yuan HB, Zhao S, Liu FF, Jiang YQ, Guo YX, Wang XL. Activation
animals: sustained activation of the TRPA1 channel contributes to the
of GABAB receptor suppresses diabetic neuropathic pain through toll-
pathogenesis of peripheral diabetic neuropathy. Pharmacol Res 2012;
like receptor 4 signaling pathway in the spinal dorsal horn. Mediators
65:149–58.
Inflamm 2018;2018:6016272.
[191] Koivisto A, Pertovaara A. Transient receptor potential ankyrin 1 (TRPA1)
[213] Liu S, Liu X, Xiong H, Wang W, Liu Y, Yin L, Tu C, Wang H, Xiang X, Xu J,
ion channel in the pathophysiology of peripheral diabetic neuropathy. Duan B, Tao A, Zhao Z, Mei Z. CXCL13/CXCR5 signaling contributes to
Scand J Pain 2013;4:129–36. diabetes-induced tactile allodynia via activating pERK, pSTAT3, pAKT
[192] Korngut L, Ma CH, Martinez JA, Toth CC, Guo GF, Singh V, Woolf CJ, pathways and pro-inflammatory cytokines production in the spinal cord
Zochodne DW. Overexpression of human HSP27 protects sensory of male mice. Brain Behav Immun 2019;80:711–24.
neurons from diabetes. Neurobiol Dis 2012;47:436–43. [214] Liu XS, Fan B, Szalad A, Jia L, Wang L, Wang X, Pan W, Zhang L, Zhang
[193] Kostyuk E, Voitenko N, Kruglikov I, Shmigol A, Shishkin V, Efimov A, R, Hu J, Zhang XM, Chopp M, Zhang ZG. MicroRNA-146a mimics
Kostyuk P. Diabetes-induced changes in calcium homeostasis and the reduce the peripheral neuropathy in type 2 diabetic mice. Diabetes
effects of calcium channel blockers in rat and mice nociceptive neurons. 2017;66:3111–21.
Diabetologia 2001;44:1302–9. [215] Llewelyn JG, Gilbey SG, Thomas PK, King RH, Muddle JR, Watkins PJ.
[194] Krishnan A, Zochodne DW. Is cytoplasmic PTEN a specific target for Sural nerve morphometry in diabetic autonomic and painful sensory
neuronal survival? Mol Neurobiol 2015;52:1758–64. neuropathy. A clinicopathological study. Brain 1991;114(pt 2):867–92.
[195] Krishnan ST, Quattrini C, Jeziorska M, Malik RA, Rayman G. Abnormal [216] Lockett MJ, Tomlinson DR. The effects of dietary treatment with
LDIflare but normal quantitative sensory testing and dermal nerve fiber essential fatty acids on sciatic nerve conduction and activity of the Na1/
density in patients with painful diabetic neuropathy. Diabetes Care K1 pump in streptozotocin-diabetic rats. Br J Pharmacol 1992;105:
2009;32:451–5. 355–60.
[196] Kumar A, Mittal R. Nrf2: a potential therapeutic target for diabetic [217] Low PA. Recent advances in the pathogenesis of diabetic neuropathy.
neuropathy. Inflammopharmacology 2017;25:393–402. Muscle Nerve 1987;10:121–8.
[197] Kushchayev SV, Belykh E, Fishchenko Y, Salei A, Teytelboym OM, [218] Lupachyk S, Shevalye H, Maksimchyk Y, Drel VR, Obrosova IG. PARP
Shabaturov L, Cruse M, Preul MC. Two bullets to the head and an early inhibition alleviates diabetes-induced systemic oxidative stress and
winter: fate permits Kutuzov to defeat Napoleon at Moscow. Neurosurg neural tissue 4-hydroxynonenal adduct accumulation: correlation with
Focus 2015;39:E3. peripheral nerve function. Free Radic Biol Med 2011;50:1400–9.
[198] Lam D, Momeni Z, Theaker M, Jagadeeshan S, Yamamoto Y, Ianowski [219] Lupachyk S, Watcho P, Shevalye H, Vareniuk I, Obrosov A, Obrosova
JP, Campanucci VA. RAGE-dependent potentiation of TRPV1 currents IG, Yorek MA. Na1/H1 exchanger 1 inhibition reverses manifestation of
in sensory neurons exposed to high glucose. PLoS One 2018;13: peripheral diabetic neuropathy in type 1 diabetic rats. Am J Physiol
e0193312. Endocrinol Metab 2013;305:E396–404.
[199] Lamping KG, Nuno DW, Coppey LJ, Holmes AJ, Hu S, Oltman CL, [220] Ma J, Pan P, Anyika M, Blagg BS, Dobrowsky RT. Modulating molecular
Norris AW, Yorek MA. Modification of high saturated fat diet with n-3 chaperones improves mitochondrial bioenergetics and decreases the
polyunsaturated fat improves glucose intolerance and vascular inflammatory transcriptome in diabetic sensory neurons. ACS Chem
dysfunction. Diabetes Obes Metab 2013;15:144–52. Neurosci 2015;6:1637–48.
[200] Latham JR, Pathirathna S, Jagodic MM, Choe WJ, Levin ME, Nelson [221] Malik RA. Wherefore art thou, O treatment for diabetic neuropathy? Int
MT, Lee WY, Krishnan K, Covey DF, Todorovic SM, Jevtovic-Todorovic Rev Neurobiol 2016;127:287–317.
[222] Malik RA, Tesfaye S, Newrick PG, Walker D, Rajbhandari SM, Siddique I,
V. Selective T-type calcium channel blockade alleviates hyperalgesia in
Sharma AK, Boulton AJ, King RH, Thomas PK, Ward JD. Sural nerve
ob/ob mice. Diabetes 2009;58:2656–65.
pathology in diabetic patients with minimal but progressive neuropathy.
[201] Laughton JD, Bittar P, Charnay Y, Pellerin L, Kovari E, Magistretti PJ,
Diabetologia 2005;48:578–85.
Bouras C. Metabolic compartmentalization in the human cortex and
[223] Malik RA, Veves A, Walker D, Siddique I, Lye RH, Schady W, Boulton AJ.
hippocampus: evidence for a cell- and region-specific localization of
Sural nerve fibre pathology in diabetic patients with mild neuropathy:
lactate dehydrogenase 5 and pyruvate dehydrogenase. BMC Neurosci
relationship to pain, quantitative sensory testing and peripheral nerve
2007;8:35. electrophysiology. Acta Neuropathol 2001;101:367–74.
[202] Lazniewska J, Rzhepetskyy Y, Zhang FX, Zamponi GW, Weiss N. [224] Malmberg AB, O’Connor WT, Glennon JC, Cesena R, Calcutt NA.
Cooperative roles of glucose and asparagine-linked glycosylation in T- Impaired formalin-evoked changes of spinal amino acid levels in diabetic
type calcium channel expression. Pflugers Arch 2016;468:1837–51. rats. Brain Res 2006;1115:48–53.
[203] Lee YC, Lu SC, Hsieh YL. Establishing a mouse model of a pure small [225] Marshall AG, Lee-Kubli C, Azmi S, Zhang M, Ferdousi M, Mixcoatl-
fiber neuropathy with the ultrapotent agonist of transient receptor Zecuatl T, Petropoulos IN, Ponirakis G, Fineman MS, Fadavi H, Frizzi K,
potential vanilloid type 1. J Vis Exp 2018;132:56651. Tavakoli M, Jeziorska M, Jolivalt CG, Boulton AJM, Efron N, Calcutt NA,
[204] Lee-Kubli C, Marshall AG, Malik RA, Calcutt NA. The H-reflex as Malik RA. Spinal disinhibition in experimental and clinical painful diabetic
a biomarker for spinal disinhibition in painful diabetic neuropathy. Curr neuropathy. Diabetes 2017;66:1380–90.
Diab Rep 2018;18:1. [226] Martin CL, Albers JW, Pop-Busui R, Group DER. Neuropathy and
[205] Lee-Kubli CA, Calcutt NA. Altered rate-dependent depression of the related findings in the diabetes control and complications trial/
spinal H-reflex as an indicator of spinal disinhibition in models of epidemiology of diabetes interventions and complications study.
neuropathic pain. PAIN 2014;155:250–60. Diabetes Care 2014;37:31–8.
[206] Li L, Yu T, Yu L, Li H, Liu Y, Wang D. Exogenous brain-derived [227] McDonnell A, Collins S, Ali Z, Iavarone L, Surujbally R, Kirby S, Butt RP.
neurotrophic factor relieves pain symptoms of diabetic rats by reducing Efficacy of the Nav1.7 blocker PF-05089771 in a randomised, placebo-
excitability of dorsal root ganglion neurons. Int J Neurosci 2016;126: controlled, double-blind clinical study in subjects with painful diabetic
749–58. peripheral neuropathy. PAIN 2018;159:1465–76.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
S82
·
N.A. Calcutt 161 (2020) S65–S86 PAIN®
[228] McGregor BA, Eid S, Rumora AE, Murdock B, Guo K, de Anda-Jauregui [251] Obradovic A, Hwang SM, Scarpa J, Hong SJ, Todorovic SM, Jevtovic-
G, Porter JE, Feldman EL, Hur J. Conserved transcriptional signatures in Todorovic V. CaV3.2 T-type calcium channels in peripheral sensory
human and murine diabetic peripheral neuropathy. Sci Rep 2018;8: neurons are important for mibefradil-induced reversal of hyperalgesia
17678. and allodynia in rats with painful diabetic neuropathy. PLoS One 2014;9:
[229] McLean WG. The role of axonal cytoskeleton in diabetic neuropathy. e91467.
Neurochem Res 1997;22:951–6. [252] Obrosova IG, Ilnytska O, Lyzogubov VV, Pavlov IA, Mashtalir N, Nadler
[230] Memon B, Abdelalim EM. Stem cell therapy for diabetes: beta cells JL, Drel VR. High-fat diet induced neuropathy of pre-diabetes and
versus pancreatic progenitors. Cells 2020;9:pii: E283. obesity: effects of “healthy” diet and aldose reductase inhibition.
[231] Menichella DM, Abdelhak B, Ren D, Shum A, Frietag C, Miller RJ. Diabetes 2007;56:2598–608.
CXCR4 chemokine receptor signaling mediates pain in diabetic [253] Oh YS. Bioactive compounds and their neuroprotective effects in
neuropathy. Mol Pain 2014;10:42. diabetic complications. Nutrients 2016;8:pii: E472.
[232] Messinger RB, Naik AK, Jagodic MM, Nelson MT, Lee WY, Choe WJ, [254] Ohi T, Saita K, Furukawa S, Ohta M, Hayashi K, Matsukura S.
Orestes P, Latham JR, Todorovic SM, Jevtovic-Todorovic V. In vivo Therapeutic effects of aldose reductase inhibitor on experimental
silencing of the Ca(V)3.2 T-type calcium channels in sensory neurons diabetic neuropathy through synthesis/secretion of nerve growth
alleviates hyperalgesia in rats with streptozocin-induced diabetic factor. Exp Neurol 1998;151:215–20.
neuropathy. PAIN 2009;145:184–95. [255] Okuse K, Chaplan SR, McMahon SB, Luo ZD, Calcutt NA, Scott BP,
[233] Migita K, Moriyama T, Koguchi M, Honda K, Katsuragi T, Takano Y, Akopian AN, Wood JN. Regulation of expression of the sensory neuron-
Ueno S. Modulation of P2X receptors in dorsal root ganglion neurons of specific sodium channel SNS in inflammatory and neuropathic pain. Mol
streptozotocin-induced diabetic neuropathy. Neurosci Lett 2009;452: Cell Neurosci 1997;10:196–207.
200–3. [256] Orestes P, Osuru HP, McIntire WE, Jacus MO, Salajegheh R, Jagodic
[234] Misra A, Bloomgarden Z. Metabolic memory: evolving concepts. MM, Choe W, Lee J, Lee SS, Rose KE, Poiro N, Digruccio MR, Krishnan
J Diabetes 2018;10:186–7. K, Covey DF, Lee JH, Barrett PQ, Jevtovic-Todorovic V, Todorovic SM.
[235] Mixcoatl-Zecuatl T, Jolivalt CG. A spinal mechanism of action for Reversal of neuropathic pain in diabetes by targeting glycosylation of
duloxetine in a rat model of painful diabetic neuropathy. Br J Pharmacol Ca(V)3.2 T-type calcium channels. Diabetes 2013;62:3828–38.
2011;164:159–69. [257] Orstavik K, Jorum E. Microneurographic findings of relevance to pain in
[236] Mizisin AP, Calcutt NA, DiStefano PS, Acheson A, Longo FM. Aldose patients with erythromelalgia and patients with diabetic neuropathy.
reductase inhibition increases CNTF-like bioactivity and protein in sciatic Neurosci Lett 2010;470:180–4.
nerves from galactose-fed and normal rats. Diabetes 1997;46:647–52. [258] Pabbidi MR, Premkumar LS. Role of transient receptor potential
[237] Mizisin AP, Calcutt NA, Tomlinson DR, Gallagher A, Fernyhough P. channels Trpv1 and Trpm8 in diabetic peripheral neuropathy.
Neurotrophin-3 reverses nerve conduction velocity deficits in J Diabetes Treat 2017;2017:pii: 029.
streptozotocin-diabetic rats. J Peripher Nerv Syst 1999;4:211–21. [259] Pabbidi RM, Cao DS, Parihar A, Pauza ME, Premkumar LS. Direct role of
[238] Mizisin AP, Nelson RW, Sturges BK, Vernau KM, Lecouteur RA, Williams
streptozotocin in inducing thermal hyperalgesia by enhanced
DC, Burgers ML, Shelton GD. Comparable myelinated nerve pathology
expression of transient receptor potential vanilloid 1 in sensory
in feline and human diabetes mellitus. Acta Neuropathol 2007;113:
neurons. Mol Pharmacol 2008;73:995–1004.
431–42.
[260] Pande M, Hur J, Hong Y, Backus C, Hayes JM, Oh SS, Kretzler M,
[239] Mizisin AP, Vu Y, Shuff M, Calcutt NA. Ciliary neurotrophic factor
Feldman EL. Transcriptional profiling of diabetic neuropathy in the BKS
improves nerve conduction and ameliorates regeneration deficits in
db/db mouse: a model of type 2 diabetes. Diabetes 2011;60:1981–9.
diabetic rats. Diabetes 2004;53:1807–12.
[261] Park S, Kang HJ, Jeon JH, Kim MJ, Lee IK. Recent advances in the
[240] Morgado C, Pereira-Terra P, Cruz CD, Tavares I. Minocycline
pathogenesis of microvascular complications in diabetes. Arch Pharm
completely reverses mechanical hyperalgesia in diabetic rats through
Res 2019;42:252–62.
microglia-induced changes in the expression of the potassium chloride
[262] Patel R, Dickenson AH. Mechanisms of the gabapentinoids and alpha 2
co-transporter 2 (KCC2) at the spinal cord. Diabetes Obes Metab 2011;
delta-1 calcium channel subunit in neuropathic pain. Pharmacol Res
13:150–9.
Perspect 2016;4:e00205.
[241] Mu ZP, Wang YG, Li CQ, Lv WS, Wang B, Jing ZH, Song XJ, Lun Y, Qiu
[263] Peterson SB, Hart GW. New insights: a role for O-GlcNAcylation in
MY, Ma XL. Association between tumor necrosis factor-alpha and
diabetic peripheral neuropathy in patients with type 2 diabetes: a meta- diabetic complications. Crit Rev Biochem Mol Biol 2016;51:150–61.
analysis. Mol Neurobiol 2017;54:983–96. [264] Petropoulos IN, Ponirakis G, Khan A, Almuhannadi H, Gad H, Malik RA.
[242] Muller C, Morales P, Reggio PH. Cannabinoid ligands targeting TRP Diagnosing diabetic neuropathy: something old, something new.
channels. Front Mol Neurosci 2018;11:487. Diabetes Metab J 2018;42:255–69.
[243] Nakae M, Kamiya H, Naruse K, Horio N, Ito Y, Mizubayashi R, Hamada [265] Pichon X, Wattiez AS, Becamel C, Ehrlich I, Bockaert J, Eschalier A,
Y, Nakashima E, Akiyama N, Kobayashi Y, Watarai A, Kimura N, Marin P, Courteix C. Disrupting 5-HT(2A) receptor/PDZ protein
Horiguchi M, Tabata Y, Oiso Y, Nakamura J. Effects of basic fibroblast interactions reduces hyperalgesia and enhances SSRI efficacy in
growth factor on experimental diabetic neuropathy in rats. Diabetes neuropathic pain. Mol Ther 2010;18:1462–70.
2006;55:1470–7. [266] Pop-Busui R, Marinescu V, Van Huysen C, Li F, Sullivan K, Greene DA,
[244] Nielsen SF, Nordestgaard BG. Statin use before diabetes diagnosis and Larkin D, Stevens MJ. Dissection of metabolic, vascular, and nerve
risk of microvascular disease: a nationwide nested matched study. conduction interrelationships in experimental diabetic neuropathy by
Lancet Diabetes Endocrinol 2014;2:894–900. cyclooxygenase inhibition and acetyl-L-carnitine administration.
[245] Nishikawa T, Edelstein D, Du XL, Yamagishi S, Matsumura T, Kaneda Y, Diabetes 2002;51:2619–28.
Yorek MA, Beebe D, Oates PJ, Hammes HP, Giardino I, Brownlee M. [267] Pop-Busui R, Martin C. Neuropathy in the DCCT/EDIC-what was done
Normalizing mitochondrial superoxide production blocks three then and what we would do better now. Int Rev Neurobiol 2016;127:
pathways of hyperglycaemic damage. Nature 2000;404:787–90. 9–25.
[246] Nukada H, McMorran PD, Baba M, Ogasawara S, Yagihashi S. [268] Powell HC, Myers RR. Axonopathy and microangiopathy in chronic
Increased susceptibility to ischemia and macrophage activation in alloxan diabetes. Acta Neuropathol 1984;65:128–37.
STZ-diabetic rat nerve. Brain Res 2011;1373:172–82. [269] Powell HC, Rosoff J, Myers RR. Microangiopathy in human diabetic
[247] O’Brien PD, Guo K, Eid SA, Rumora AE, Hinder LM, Hayes JM, neuropathy. Acta Neuropathol 1985;68:295–305.
Mendelson FE, Hur J, Feldman EL. Integrated lipidomic and [270] Price SA, Zeef LA, Wardleworth L, Hayes A, Tomlinson DR. Identification
transcriptomic analyses identify altered nerve triglycerides in mouse of changes in gene expression in dorsal root ganglia in diabetic
models of prediabetes and type 2 diabetes. Dis Model Mech 2020;13: neuropathy: correlation with functional deficits. J Neuropathol Exp
doi:10.1242/dmm.042101. Neurol 2006;65:722–32.
[248] O’Brien PD, Hinder LM, Sakowski SA, Feldman EL. ER stress in diabetic [271] Purwata TE. High TNF-alpha plasma levels and macrophages iNOS and
peripheral neuropathy: a new therapeutic target. Antioxid Redox Signal TNF-alpha expression as risk factors for painful diabetic neuropathy.
2014;21:621–33. J Pain Res 2011;4:169–75.
[249] O’Brien PD, Hur J, Hayes JM, Backus C, Sakowski SA, Feldman EL. [272] Puttgen S, Bonhof GJ, Strom A, Mussig K, Szendroedi J, Roden M,
BTBR ob/ob mice as a novel diabetic neuropathy model: neurological Ziegler D. Augmented corneal nerve fiber branching in painful compared
characterization and gene expression analyses. Neurobiol Dis 2015;73: with painless diabetic neuropathy. J Clin Endocrinol Metab 2019;104:
348–55. 6220–8.
[250] Oates PJ. Aldose reductase, still a compelling target for diabetic [273] Quattrini C, Harris ND, Malik RA, Tesfaye S. Impaired skin microvascular
neuropathy. Curr Drug Targets 2008;9:14–36. reactivity in painful diabetic neuropathy. Diabetes Care 2007;30:655–9.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
September 2020
· Volume 161
· Number 9
· Supplement 1 www.painjournalonline.com S83
[274] Quattrini C, Tavakoli M, Jeziorska M, Kallinikos P, Tesfaye S, Finnigan J, spinal cord NMDA receptor phosphorylation in a rat model of diabetic
Marshall A, Boulton AJ, Efron N, Malik RA. Surrogate markers of small neuropathic pain. J Physiol 2010;588(pt 21):4205–15.
fiber damage in human diabetic neuropathy. Diabetes 2007;56: [297] Rowe-Rendleman CL, Eichberg J. P0 phosphorylation in nerves from
2148–54. normal and diabetic rats: role of protein kinase C and turnover of
[275] Rachana KS, Manu MS, Advirao GM. Insulin influenced expression of phosphate groups. Neurochem Res 1994;19:1023–31.
myelin proteins in diabetic peripheral neuropathy. Neurosci Lett 2016; [298] Roy Chowdhury SK, Smith DR, Saleh A, Schapansky J, Marquez A,
629:110–15. Gomes S, Akude E, Morrow D, Calcutt NA, Fernyhough P. Impaired
[276] Rahman MH, Jha MK, Kim JH, Nam Y, Lee MG, Go Y, Harris RA, Park adenosine monophosphate-activated protein kinase signalling in dorsal
DH, Kook H, Lee IK, Suk K. Pyruvate dehydrogenase kinase-mediated root ganglia neurons is linked to mitochondrial dysfunction and
glycolytic metabolic shift in the dorsal root ganglion drives painful peripheral neuropathy in diabetes. Brain 2012;135(pt 6):1751–66.
diabetic neuropathy. J Biol Chem 2016;291:6011–25. [299] Ruiz HH, Ramasamy R, Schmidt AM. Advanced glycation end products:
[277] Rajabally YA, Stettner M, Kieseier BC, Hartung HP, Malik RA. CIDP and building on the concept of the “common soil” in metabolic disease.
other inflammatory neuropathies in diabetes—diagnosis and Endocrinology 2020;161:pii: bqz006.
management. Nat Rev Neurol 2017;13:599–611. [300] Rumora AE, LoGrasso G, Hayes JM, Mendelson FE, Tabbey MA, Haidar
[278] Rajamani K, Colman PG, Li LP, Best JD, Voysey M, D’Emden MC, JA, Lentz SI, Feldman EL. The divergent roles of dietary saturated and
Laakso M, Baker JR, Keech AC; Fs investigators. Effect of fenofibrate on monounsaturated fatty acids on nerve function in murine models of
amputation events in people with type 2 diabetes mellitus (FIELD study): obesity. J Neurosci 2019;39:3770–81.
a prespecified analysis of a randomised controlled trial. Lancet 2009; [301] Rumora AE, Savelieff MG, Sakowski SA, Feldman EL. Disorders of
373:1780–8. mitochondrial dynamics in peripheral neuropathy: clues from hereditary
[279] Rajbhandari SM, Jarratt JA, Griffiths PD, Ward JD. Diabetic neuropathic neuropathy and diabetes. Int Rev Neurobiol 2019;145:127–76.
pain in a leg amputated 44 years previously. PAIN 1999;83:627–9. [302] Russell JW, Sullivan KA, Windebank AJ, Herrmann DN, Feldman EL.
[280] Ramji N, Toth C, Kennedy J, Zochodne DW. Does diabetes mellitus Neurons undergo apoptosis in animal and cell culture models of
target motor neurons? Neurobiol Dis 2007;26:301–11. diabetes. Neurobiol Dis 1999;6:347–63.
[281] Ramli R, Reddy M, Oliver N. Artificial pancreas: current progress and [303] Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N,
future outlook in the treatment of type 1 diabetes. Drugs 2019;79: Colagiuri S, Guariguata L, Motala AA, Ogurtsova K, Shaw JE, Bright D,
1089–101. Williams R, Committee IDFDA. Global and regional diabetes prevalence
[282] Ramos KM, Jiang Y, Svensson CI, Calcutt NA. Pathogenesis of spinally estimates for 2019 and projections for 2030 and 2045: results from the
mediated hyperalgesia in diabetes. Diabetes 2007;56:1569–76. International Diabetes Federation Diabetes Atlas, 9(th) edition. Diabetes
[283] Raputova J, Srotova I, Vlckova E, Sommer C, Uceyler N, Birklein F, Res Clin Pract 2019;157:107843.
Rittner HL, Rebhorn C, Adamova B, Kovalova I, Kralickova Nekvapilova [304] Saleh A, Smith DR, Tessler L, Mateo AR, Martens C, Schartner E, Van
E, Forer L, Belobradkova J, Olsovsky J, Weber P, Dusek L, Jarkovsky J, der Ploeg R, Toth C, Zochodne DW, Fernyhough P. Receptor for
Bednarik J. Sensory phenotype and risk factors for painful diabetic advanced glycation end-products (RAGE) activates divergent signaling
pathways to augment neurite outgrowth of adult sensory neurons. Exp
neuropathy: a cross-sectional observational study. PAIN 2017;158:
Neurol 2013;249:149–59.
2340–53.
[305] Samii A, Unger J, Lange W. Vascular endothelial growth factor
[284] Ren YS, Qian NS, Tang Y, Liao YH, Yang YL, Dou KF, Toi M. Sodium
expression in peripheral nerves and dorsal root ganglia in diabetic
channel Nav1.6 is up-regulated in the dorsal root ganglia in a mouse
neuropathy in rats. Neurosci Lett 1999;262:159–62.
model of type 2 diabetes. Brain Res Bull 2012;87:244–9.
[306] Sano R, Reed JC. ER stress-induced cell death mechanisms. Biochim
[285] Ristoiu V, Shibasaki K, Uchida K, Zhou Y, Ton BH, Flonta ML, Tominaga
Biophys Acta 2013;1833:3460–70.
M. Hypoxia-induced sensitization of transient receptor potential vanilloid
[307] Schmeichel AM, Schmelzer JD, Low PA. Oxidative injury and apoptosis
1 involves activation of hypoxia-inducible factor-1 alpha and PKC. PAIN
of dorsal root ganglion neurons in chronic experimental diabetic
2011;152:936–45.
neuropathy. Diabetes 2003;52:165–71.
[286] Roa-Coria JE, Pineda-Farias JB, Barragan-Iglesias P, Quinonez-
[308] Schratzberger P, Walter DH, Rittig K, Bahlmann FH, Pola R, Curry C,
Bastidas GN, Zuniga-Romero A, Huerta-Cruz JC, Reyes-Garcia JG,
Silver M, Krainin JG, Weinberg DH, Ropper AH, Isner JM. Reversal of
Flores-Murrieta FJ, Granados-Soto V, Rocha-Gonzalez HI. Possible
experimental diabetic neuropathy by VEGF gene transfer. J Clin Invest
involvement of peripheral TRP channels in the hydrogen sulfide-induced 2001;107:1083–92.
hyperalgesia in diabetic rats. BMC Neurosci 2019;20:1. [309] Scott JN, Clark AW, Zochodne DW. Neurofilament and tubulin gene
[287] Robb JL, Morrissey NA, Weightman Potter PG, Smithers HE, Beall C, expression in progressive experimental diabetes: failure of synthesis and
Ellacott KLJ. Immunometabolic changes in glia—a potential role in the export by sensory neurons. Brain 1999;122(pt 11):2109–18.
pathophysiology of obesity and diabetes. Neuroscience 2019;S0303- [310] Segerdahl AR, Themistocleous AC, Fido D, Bennett DL, Tracey I. A
4522(19)30708-0. brain-based pain facilitation mechanism contributes to painful diabetic
[288] Robertson DM, Sima AA. Diabetic neuropathy in the mutant mouse polyneuropathy. Brain 2018;141:357–64.
[C57BL/ks(db/db)]: a morphometric study. Diabetes 1980;29:60–7. [311] Selvarajah D, Wilkinson ID, Emery CJ, Harris ND, Shaw PJ, Witte DR,
[289] Rodriguez-Gutierrez R, Montori VM. Glycemic control for patients with Griffiths PD, Tesfaye S. Early involvement of the spinal cord in diabetic
type 2 diabetes mellitus: our evolving faith in the face of evidence. Circ peripheral neuropathy. Diabetes Care 2006;29:2664–9.
Cardiovasc Qual Outcomes 2016;9:504–12. [312] Selvarajah D, Wilkinson ID, Fang F, Sankar A, Davies J, Boland E,
[290] Rojas DR, Tegeder I, Kuner R, Agarwal N. Hypoxia-inducible factor Harding J, Rao G, Gandhi R, Tracey I, Tesfaye S. Structural and
1alpha protects peripheral sensory neurons from diabetic peripheral functional abnormalities of the primary somatosensory cortex in diabetic
neuropathy by suppressing accumulation of reactive oxygen species. peripheral neuropathy: a multimodal MRI study. Diabetes 2019;68:
J Mol Med (Berl) 2018;96:1395–405. 796–806.
[291] Rojewska E, Zychowska M, Piotrowska A, Kreiner G, Nalepa I, Mika J. [313] Selvarajah D, Wilkinson ID, Gandhi R, Griffiths PD, Tesfaye S.
Involvement of macrophage inflammatory protein-1 family members in Microvascular perfusion abnormalities of the Thalamus in painful but
the development of diabetic neuropathy and their contribution to not painless diabetic polyneuropathy: a clue to the pathogenesis of pain
effectiveness of morphine. Front Immunol 2018;9:494. in type 1 diabetes. Diabetes Care 2011;34:718–20.
[292] Romanovsky D, Cruz NF, Dienel GA, Dobretsov M. Mechanical [314] Seneviratne KN, Peiris OA. The effect of ischaemia on the excitability of
hyperalgesia correlates with insulin deficiency in normoglycemic human sensory nerve. J Neurol Neurosurg Psychiatry 1968;31:338–47.
streptozotocin-treated rats. Neurobiol Dis 2006;24:384–94. [315] Serra J, Duan WR, Locke C, Sola R, Liu W, Nothaft W. Effects of a T-type
[293] Romanovsky D, Dobretsov M. Pressure-induced pain: early sign of calcium channel blocker, ABT-639, on spontaneous activity in C-
diabetes-associated impairment of insulin production in rats. Neurosci nociceptors in patients with painful diabetic neuropathy: a randomized
Lett 2010;483:110–13. controlled trial. PAIN 2015;156:2175–83.
[294] Romanovsky D, Hastings SL, Stimers JR, Dobretsov M. Relevance of [316] Shevalye H, Yorek MS, Coppey LJ, Holmes A, Harper MM, Kardon RH,
hyperglycemia to early mechanical hyperalgesia in streptozotocin- Yorek MA. Effect of enriching the diet with menhaden oil or daily
induced diabetes. J Peripher Nerv Syst 2004;9:62–9. treatment with resolvin D1 on neuropathy in a mouse model of type 2
[295] Romanovsky D, Walker JC, Dobretsov M. Pressure pain precedes diabetes. J Neurophysiol 2015;114:199–208.
development of type 2 disease in Zucker rat model of diabetes. Neurosci [317] Shi X, Qiu Z, Inam UL, Zhang M, Li K, Wu P, Suleman R, Aadil RM, Piao
Lett 2008;445:220–3. F. The microRNAs expression profile in sciatic nerves of diabetic
[296] Rondon LJ, Privat AM, Daulhac L, Davin N, Mazur A, Fialip J, Eschalier neuropathy rats after taurine treatment by sequencing. Adv Exp Med
A, Courteix C. Magnesium attenuates chronic hypersensitivity and Biol 2019;1155:935–47.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
S84
·
N.A. Calcutt 161 (2020) S65–S86 PAIN®
[318] Shillo P, Selvarajah D, Greig M, Gandhi R, Rao G, Wilkinson ID, Anand P, [343] Talbot S, Chahmi E, Dias JP, Couture R. Key role for spinal dorsal horn
Tesfaye S. Reduced vitamin D levels in painful diabetic peripheral microglial kinin B1 receptor in early diabetic pain neuropathy.
neuropathy. Diabet Med 2019;36:44–51. J Neuroinflammation 2010;7:36.
[319] Shillo P, Sloan G, Greig M, Hunt L, Selvarajah D, Elliott J, Gandhi R, [344] Tan AM, Samad OA, Dib-Hajj SD, Waxman SG. Virus-mediated
Wilkinson ID, Tesfaye S. Painful and painless diabetic neuropathies: knockdown of Nav1.3 in dorsal root ganglia of STZ-induced diabetic
what is the difference? Curr Diab Rep 2019;19:32. rats alleviates tactile allodynia. Mol Med 2015;21:544–52.
[320] Shimoshige Y, Enomoto R, Aoki T, Matsuoka N, Kaneko S. The [345] Tang Y, Lenzini PA, Pop-Busui R, Ray PR, Campbell H, Perkins BA,
involvement of aldose reductase in alterations to neurotrophin receptors Callaghan B, Wagner MJ, Motsinger-Reif AA, Buse JB, Price TJ,
and neuronal cytoskeletal protein mRNA levels in the dorsal root Mychaleckyj JC, Cresci S, Shah H, Doria A. A genetic locus on
ganglion of streptozotocin-induced diabetic rats. Biol Pharm Bull 2010; chromosome 2q24 predicting peripheral neuropathy risk in type 2
33:67–71. diabetes: results from the ACCORD and BARI 2D studies. Diabetes
[321] Shutov L, Kruglikov I, Gryshchenko O, Khomula E, Viatchenko- 2019;68:1649–62.
Karpinski V, Belan P, Voitenko N. The effect of nimodipine on calcium [346] Teixeira JM, Dos Santos GG, Neves AF, Athie MCP, Bonet IJM,
homeostasis and pain sensitivity in diabetic rats. Cell Mol Neurobiol Nishijima CM, Farias FH, Figueiredo JG, Hernandez-Olmos V,
2006;26:1541–57. Alshaibani S, Tambeli CH, Muller CE, Parada CA. Diabetes-induced
[322] Sima AA. C-peptide and diabetic neuropathy. Expert Opin Investig neuropathic mechanical hyperalgesia depends on P2X4 receptor
Drugs 2003;12:1471–88. activation in dorsal root ganglia. Neuroscience 2019;398:158–70.
[323] Sima AA. Encephalopathies: the emerging diabetic complications. Acta [347] Thakur V, Gonzalez M, Pennington K, Chattopadhyay M. Viral vector
Diabetol 2010;47:279–93. mediated continuous expression of interleukin-10 in DRG alleviates pain
[324] Simeoli R, Fierabracci A. Insights into the role of MicroRNAs in the onset in type 1 diabetic animals. Mol Cell Neurosci 2016;72:46–53.
and development of diabetic neuropathy. Int J Mol Sci 2019;20:pii: [348] Themistocleous AC, Ramirez JD, Shillo PR, Lees JG, Selvarajah D,
E4627. Orengo C, Tesfaye S, Rice AS, Bennett DL. The Pain in Neuropathy
[325] Singh B, Singh V, Krishnan A, Koshy K, Martinez JA, Cheng C, Almquist Study (PiNS): a cross-sectional observational study determining the
C, Zochodne DW. Regeneration of diabetic axons is enhanced by somatosensory phenotype of painful and painless diabetic neuropathy.
selective knockdown of the PTEN gene. Brain 2014;137(pt 4):1051–67. PAIN 2016;157:1132–45.
[326] Singh J, Yousuf MS, Jones KE, Shelemey PTM, Joy T, Macandili H, Kerr [349] Thomas PK. The morphological basis for alterations in nerve conduction
BJ, Zochodne DW, Sauve Y, Ballanyi K, Webber CA. Characterization of in peripheral neuropathy. Proc R Soc Med 1971;64:295–8.
the nile grass rat as a unique model for type 2 diabetic polyneuropathy. [350] Thornalley PJ. Glycation in diabetic neuropathy: characteristics,
J Neuropathol Exp Neurol 2018;77:469–78. consequences, causes, and therapeutic options. Int Rev Neurobiol
[327] Skapare E, Konrade I, Liepinsh E, Strele I, Makrecka M, Bierhaus A, 2002;50:37–57.
Lejnieks A, Pirags V, Dambrova M. Association of reduced glyoxalase 1 [351] Tibbs GR, Posson DJ, Goldstein PA. Voltage-gated ion channels in the
activity and painful peripheral diabetic neuropathy in type 1 and 2 PNS: novel therapies for neuropathic pain? Trends Pharmacol Sci 2016;
diabetes mellitus patients. J Diabetes Complications 2013;27:262–7. 37:522–42.
[328] Slager UT. Diabetic myelopathy. Arch Pathol Lab Med 1978;102:467–9. [352] Todorovic SM. Painful diabetic neuropathy: prevention or suppression?.
[329] Smith AG, Russell J, Feldman EL, Goldstein J, Peltier A, Smith S, Hamwi Int Rev Neurobiol 2016;127:211–25.
J, Pollari D, Bixby B, Howard J, Singleton JR. Lifestyle intervention for [353] Tomiyama M, Furusawa K, Kamijo M, Kimura T, Matsunaga M, Baba M.
pre-diabetic neuropathy. Diabetes Care 2006;29:1294–9. Upregulation of mRNAs coding for AMPA and NMDA receptor subunits
[330] Smith BE. Focal and entrapment neuropathies. Handb Clin Neurol and metabotropic glutamate receptors in the dorsal horn of the spinal
2014;126:31–43. cord in a rat model of diabetes mellitus. Brain Res Mol Brain Res 2005;
[331] Sopacua M, Hoeijmakers JGJ, Merkies ISJ, Lauria G, Waxman SG, 136:275–81.
Faber CG. Small-fiber neuropathy: expanding the clinical pain universe. [354] Tomlinson DR, Robinson JP, Willars GB, Keen P. Deficient axonal
J Peripher Nerv Syst 2019;24:19–33. transport of substance P in streptozocin-induced diabetic rats. Effects
[332] Spallone V, Greco C. Painful and painless diabetic neuropathy: one of sorbinil and insulin. Diabetes 1988;37:488–93.
disease or two? Curr Diab Rep 2013;13:533–49. [355] Toth C, Brussee V, Zochodne DW. Remote neurotrophic support of
[333] Spallone V, Morganti R, D’Amato C, Cacciotti L, Fedele T, Maiello MR, epidermal nerve fibres in experimental diabetes. Diabetologia 2006;49:
Marfia G. Clinical correlates of painful diabetic neuropathy and 1081–8.
relationship of neuropathic pain with sensorimotor and autonomic [356] Toth C, Martinez J, Zochodne DW. RAGE, diabetes, and the nervous
nerve function. Eur J Pain 2011;15:153–60. system. Curr Mol Med 2007;7:766–76.
[334] Srivastava SK, Yadav UC, Reddy AB, Saxena A, Tammali R, Shoeb M, [357] Truini A, Spallone V, Morganti R, Tamburin S, Zanette G, Schenone A,
Ansari NH, Bhatnagar A, Petrash MJ, Srivastava S, Ramana KV. Aldose De Michelis C, Tugnoli V, Simioni V, Manganelli F, Dubbioso R, Lauria G,
reductase inhibition suppresses oxidative stress-induced inflammatory Lombardi R, Jann S, De Toni Franceschini L, Tesfaye S, Fiorelli M,
disorders. Chem Biol Interact 2011;191:330–8. Spagnoli A, Cruccu G. A cross-sectional study investigating frequency
[335] Stark B, Carlstedt T, Cullheim S, Risling M. Developmental and lesion- and features of definitely diagnosed diabetic painful polyneuropathy.
induced changes in the distribution of the glucose transporter Glut-1 in PAIN 2018;159:2658–66.
the central and peripheral nervous system. Exp Brain Res 2000;131: [358] Tsantoulas C, Lainez S, Wong S, Mehta I, Vilar B, McNaughton PA.
74–84. Hyperpolarization-activated cyclic nucleotide-gated 2 (HCN2) ion
[336] Stino AM, Smith AG. Peripheral neuropathy in prediabetes and the channels drive pain in mouse models of diabetic neuropathy. Sci
metabolic syndrome. J Diabetes Investig 2017;8:646–55. Transl Med 2017;9:eaam6072.
[337] Stracke H, Gaus W, Achenbach U, Federlin K, Bretzel RG. Benfotiamine [359] Tsantoulas C, Mooney ER, McNaughton PA. HCN2 ion channels: basic
in diabetic polyneuropathy (BENDIP): results of a randomised, double science opens up possibilities for therapeutic intervention in neuropathic
blind, placebo-controlled clinical study. Exp Clin Endocrinol Diabetes pain. Biochem J 2016;473:2717–36.
2008;116:600–5. [360] Tseng MT, Chiang MC, Chao CC, Tseng WY, Hsieh ST. fMRI evidence
[338] Stuart CA, Wen G, Peng BH, Popov VL, Hudnall SD, Campbell GA. of degeneration-induced neuropathic pain in diabetes: enhanced limbic
GLUT-3 expression in human skeletal muscle. Am J Physiol Endocrinol and striatal activations. Hum Brain Mapp 2013;34:2733–46.
Metab 2000;279:E855–861. [361] Tsuda M, Ueno H, Kataoka A, Tozaki-Saitoh H, Inoue K. Activation of
[339] Sugimoto K, Yasujima M, Yagihashi S. Role of advanced glycation end dorsal horn microglia contributes to diabetes-induced tactile allodynia
products in diabetic neuropathy. Curr Pharm Des 2008;14:953–61. via extracellular signal-regulated protein kinase signaling. Glia 2008;56:
[340] Suo M, Wang P, Zhang M. Role of Fyn-mediated NMDA receptor 378–86.
function in prediabetic neuropathy in mice. J Neurophysiol 2016;116: [362] Ulugol A, Oltulu C, Gunduz O, Citak C, Carrara R, Shaqaqi MR, Sanchez
448–55. AM, Dogrul A. 5-HT7 receptor activation attenuates thermal
[341] Tack CJ, van Gurp PJ, Holmes C, Goldstein DS. Local sympathetic hyperalgesia in streptozocin-induced diabetic mice. Pharmacol
denervation in painful diabetic neuropathy. Diabetes 2002;51:3545–53. Biochem Behav 2012;102:344–8.
[342] Takaku S, Yako H, Niimi N, Akamine T, Kawanami D, Utsunomiya K, [363] Umeda M, Ohkubo T, Ono J, Fukuizumi T, Kitamura K. Molecular and
Sango K. Establishment of a myelinating co-culture system with a motor immunohistochemical studies in expression of voltage-dependent
neuron-like cell line NSC-34 and an adult rat Schwann cell line IFRS1. Ca21 channels in dorsal root ganglia from streptozotocin-induced
Histochem Cell Biol 2018;149:537–43. diabetic mice. Life Sci 2006;79:1995–2000.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
September 2020
· Volume 161
· Number 9
· Supplement 1 www.painjournalonline.com S85
[364] Urban MJ, Li C, Yu C, Lu Y, Krise JM, McIntosh MP, Rajewski RA, Blagg [386] Wei H, Chapman H, Saarnilehto M, Kuokkanen K, Koivisto A,
BS, Dobrowsky RT. Inhibiting heat-shock protein 90 reverses sensory Pertovaara A. Roles of cutaneous versus spinal TRPA1 channels in
hypoalgesia in diabetic mice. ASN Neuro 2010;2:e00040. mechanical hypersensitivity in the diabetic or mustard oil-treated non-
[365] Urru M, Muzzi M, Coppi E, Ranieri G, Buonvicino D, Camaioni E, Coppini diabetic rat. Neuropharmacology 2010;58:578–84.
R, Pugliese AM, Tanaka B, Estacion M, Waxman SG, Dib-Hajj SD, [387] Wei H, Viisanen H, Amorim D, Koivisto A, Pertovaara A. Dissociated
Chiarugi A. Dexpramipexole blocks Nav1.8 sodium channels and modulation of conditioned place-preference and mechanical
provides analgesia in multiple nociceptive and neuropathic pain models. hypersensitivity by a TRPA1 channel antagonist in peripheral
PAIN 2020;161:831–41. neuropathy. Pharmacol Biochem Behav 2013;104:90–6.
[366] Van Buren JJ, Bhat S, Rotello R, Pauza ME, Premkumar LS. [388] Weiss N, Black SA, Bladen C, Chen L, Zamponi GW. Surface
Sensitization and translocation of TRPV1 by insulin and IGF-I. Mol expression and function of Cav3.2 T-type calcium channels are
Pain 2005;1:17. controlled by asparagine-linked glycosylation. Pflugers Arch 2013;
[367] Verkhratsky A, Fernyhough P. Calcium signalling in sensory neurones 465:1159–70.
and peripheral glia in the context of diabetic neuropathies. Cell Calcium [389] Wellmer A, Misra VP, Sharief MK, Kopelman PG, Anand P. A double-
2014;56:362–71. blind placebo-controlled clinical trial of recombinant human brain-
[368] Viader A, Golden JP, Baloh RH, Schmidt RE, Hunter DA, Milbrandt J. derived neurotrophic factor (rhBDNF) in diabetic polyneuropathy.
Schwann cell mitochondrial metabolism supports long-term axonal J Peripher Nerv Syst 2001;6:204–10.
survival and peripheral nerve function. J Neurosci 2011;31:10128–40. [390] Wickenden AD, McNaughton-Smith G. Kv7 channels as targets for the
[369] Villegas-Rivera G, Roman-Pintos LM, Cardona-Munoz EG, Arias- treatment of pain. Curr Pharm Des 2009;15:1773–98.
Carvajal O, Rodriguez-Carrizalez AD, Troyo-Sanroman R, Pacheco- [391] Wolff RF, Bala MM, Westwood M, Kessels AG, Kleijnen J. 5% lidocaine-
Moises FP, Moreno-Ulloa A, Miranda-Diaz AG. Effects of ezetimibe/ medicated plaster vs other relevant interventions and placebo for post-
simvastatin and rosuvastatin on oxidative stress in diabetic neuropathy: herpetic neuralgia (PHN): a systematic review. Acta Neurol Scand 2011;
a randomized, double-blind, placebo-controlled clinical trial. Oxid Med 123:295–309.
Cell Longev 2015;2015:756294. [392] Wu YB, Xu DD, Zhu X, Yang GW, Ren MS. MiR-106a associated with
[370] Villeneuve LM, Natarajan R. Epigenetics of diabetic complications. diabetic peripheral neuropathy through the regulation of 12/15-LOX-
Expert Rev Endocrinol Metab 2010;5:137–48. mediated oxidative/nitrative stress. Curr Neurovasc Res 2017;14:
[371] Vincent AM, Hayes JM, McLean LL, Vivekanandan-Giri A, Pennathur S, 117–24.
Feldman EL. Dyslipidemia-induced neuropathy in mice: the role of [393] Xu GY, Li GW, Liu NG, Huang LYM. Mechanisms underlying purinergic
oxLDL/LOX-1. Diabetes 2009;58:2376–85. P2X3 receptor-mediated mechanical allodynia induced in diabetic rats.
[372] Vincent AM, Kato K, McLean LL, Soules ME, Feldman EL. Sensory Mol Pain 2011;7:60.
neurons and Schwann cells respond to oxidative stress by increasing [394] Xu QG, Li XQ, Kotecha SA, Cheng C, Sun HS, Zochodne DW. Insulin as
antioxidant defense mechanisms. Antioxid Redox Signal 2009;11: an in vivo growth factor. Exp Neurol 2004;188:43–51.
425–38. [395] Yagihashi S, Kamijo M, Baba M, Yagihashi N, Nagai K. Effect of
[373] Vlassara H, Brownlee M, Cerami A. Nonenzymatic glycosylation of aminoguanidine on functional and structural abnormalities in peripheral
peripheral nerve protein in diabetes mellitus. Proc Natl Acad Sci U S A nerve of STZ-induced diabetic rats. Diabetes 1992;41:47–52.
1981;78:5190–2. [396] Yagihashi S, Nishihira M, Baba M. Morphometrical analysis of the
[374] Voitenko NV, Kruglikov IA, Kostyuk EP, Kostyuk PG. Effect of peripheral nerve lesions in experimental diabetes rats. Tohoku J Exp
streptozotocin-induced diabetes on the activity of calcium channels in Med 1979;129:139–49.
rat dorsal horn neurons. Neuroscience 2000;95:519–24. [397] Yamakawa I, Kojima H, Terashima T, Katagi M, Oi J, Urabe H, Sanada
[375] Wada R, Yagihashi S. Role of advanced glycation end products and M, Kawai H, Chan L, Yasuda H, Maegawa H, Kimura H. Inactivation of
their receptors in development of diabetic neuropathy. Ann N Y Acad Sci TNF-alpha ameliorates diabetic neuropathy in mice. Am J Physiol
2005;1043:598–604. Endocrinol Metab 2011;301:E844–852.
[376] Wagner K, Lee KS, Yang J, Hammock BD. Epoxy fatty acids mediate [398] Yamamoto H, Shimoshige Y, Yamaji T, Murai N, Aoki T, Matsuoka N.
analgesia in murine diabetic neuropathy. Eur J Pain 2017;21:456–65. Pharmacological characterization of standard analgesics on mechanical
[377] Wahren J, Foyt H, Daniels M, Arezzo JC. Long-Acting C-peptide and allodynia in streptozotocin-induced diabetic rats. Neuropharmacology
neuropathy in type 1 diabetes: a 12-month clinical trial. Diabetes Care 2009;57:403–8.
2016;39:596–602. [399] Yamazaki S, Yamaji T, Murai N, Yamamoto H, Matsuda T, Price RD,
[378] Walker D, Carrington A, Cannan SA, Sawicki D, Sredy J, Boulton AJ, Matsuoka N. FK1706, a novel non-immunosuppressive immunophilin
Malik RA. Structural abnormalities do not explain the early functional ligand, modifies gene expression in the dorsal root ganglia during painful
abnormalities in the peripheral nerves of the streptozotocin diabetic rat. diabetic neuropathy. Neurol Res 2012;34:469–77.
J Anat 1999;195(pt 3):419–27. [400] Yerra VG, Areti A, Kumar A. Adenosine monophosphate-activated
[379] Wang L, Chopp M, Lu X, Szalad A, Jia L, Liu XS, Wu KH, Lu M, Zhang protein kinase abates hyperglycaemia-induced neuronal injury in
ZG. miR-146a mediates thymosin beta4 induced neurovascular experimental models of diabetic neuropathy: effects on mitochondrial
remodeling of diabetic peripheral neuropathy in type-II diabetic mice. biogenesis, autophagy and neuroinflammation. Mol Neurobiol 2017;54:
Brain Res 2019;1707:198–207. 2301–12.
[380] Wang XC, Wang S, Zhang M, Gao F, Yin C, Li H, Zhang Y, Hu SJ, Duan [401] Yoon H, Thakur V, Isham D, Fayad M, Chattopadhyay M. Moderate
JH. Alpha-Dendrotoxin-sensitive Kv1 channels contribute to conduction exercise training attenuates inflammatory mediators in DRG of Type 1
failure of polymodal nociceptive C-fibers from rat coccygeal nerve. diabetic rats. Exp Neurol 2015;267:107–14.
J Neurophysiol 2016;115:947–57. [402] Yorek MA. Alternatives to the streptozotocin-diabetic rodent. Int Rev
[381] Wang XL, Zhang Q, Zhang YZ, Liu YT, Dong R, Wang QJ, Guo YX. Neurobiol 2016;127:89–112.
Downregulation of GABAB receptors in the spinal cord dorsal horn in [403] Yousefzadeh N, Alipour MR, Soufi FG. Deregulation of NF-small ka,
diabetic neuropathy. Neurosci Lett 2011;490:112–15. CyrillicB-miR-146a negative feedback loop may be involved in the
[382] Wang Y, Zhao X, Wu X, Dai Y, Chen P, Xie L. microRNA-182 mediates pathogenesis of diabetic neuropathy. J Physiol Biochem 2015;71:51–8.
Sirt1-induced diabetic corneal nerve regeneration. Diabetes 2016;65: [404] Yu T, Li L, Liu H, Li H, Liu Z, Li Z. KCNQ2/3/5 channels in dorsal root
2020–31. ganglion neurons can be therapeutic targets of neuropathic pain in
[383] Wang-Fischer Y, Garyantes T. Improving the reliability and utility of diabetic rats. Mol Pain 2018;14:1744806918793229.
streptozotocin-induced rat diabetic model. J Diabetes Res 2018;2018: [405] Zaharia OP, Kuss O, Strassburger K, Burkart V, Szendroedi J, Roden M.
8054073. Diabetes clusters and risk of diabetes-associated diseases—authors’
[384] Watanabe K, Hirano S, Kojima K, Nagashima K, Mukai H, Sato T, reply. Lancet Diabetes Endocrinol 2019;7:828–9.
Takemoto M, Matsumoto K, Iimori T, Isose S, Omori S, Shibuya K, [406] Zhang HH, Hu J, Zhou YL, Qin X, Song ZY, Yang PP, Hu SF, Jiang XH,
Sekiguchi Y, Beppu M, Amino H, Suichi T, Yokote K, Uno T, Kuwabara Xu GY. Promoted interaction of nuclear factor-kappa B with
S, Misawa S. Altered cerebral blood flow in the anterior cingulate cortex demethylated purinergic P2X3 receptor gene contributes to
is associated with neuropathic pain. J Neurol Neurosurg Psychiatry neuropathic pain in rats with diabetes. Diabetes 2015;64:4272–84.
2018;89:1082–7. [407] Zhang L, Qu S, Liang A, Jiang H, Wang H. Gene expression microarray
[385] Waterman RS, Morgenweck J, Nossaman BD, Scandurro AE, analysis of the sciatic nerve of mice with diabetic neuropathy. Int J Mol
Scandurro SA, Betancourt AM. Anti-inflammatory mesenchymal stem Med 2015;35:333–9.
cells (MSC2) attenuate symptoms of painful diabetic peripheral [408] Zhang L, Zhao H, Blagg BS, Dobrowsky RT. C-terminal heat shock
neuropathy. Stem Cells Transl Med 2012;1:557–65. protein 90 inhibitor decreases hyperglycemia-induced oxidative stress
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.
S86
·
N.A. Calcutt 161 (2020) S65–S86 PAIN®
and improves mitochondrial bioenergetics in sensory neurons. [415] Zochodne DW, Verge VM, Cheng C, Hoke A, Jolley C, Thomsen K,
J Proteome Res 2012;11:2581–93. Rubin I, Lauritzen M. Nitric oxide synthase activity and expression in
[409] Zhang W, Kamiya H, Ekberg K, Wahren J, Sima AA. C-peptide improves experimental diabetic neuropathy. J Neuropathol Exp Neurol 2000;59:
neuropathy in type 1 diabetic BB/Wor-rats. Diabetes Metab Res Rev 798–807.
2007;23:63–70. [416] Zochodne DW, Verge VM, Cheng C, Sun H, Johnston J. Does diabetes
[410] Zhang Y, Song C, Liu J, Bi Y, Li H. Inhibition of miR-25 aggravates target ganglion neurones? Progressive sensory neurone involvement in
diabetic peripheral neuropathy. Neuroreport 2018;29:945–53. long-term experimental diabetes. Brain 2001;124(pt 11):2319–34.
[411] Zherebitskaya E, Schapansky J, Akude E, Smith DR, Van der Ploeg R, [417] Zoungas S, Arima H, Gerstein HC, Holman RR, Woodward M, Reaven
Solovyova N, Verkhratsky A, Fernyhough P. Sensory neurons derived P, Hayward RA, Craven T, Coleman RL, Chalmers J; Collaborators on
from diabetic rats have diminished internal Ca21 stores linked to Trials of Lowering Glucose g. Effects of intensive glucose control on
impaired re-uptake by the endoplasmic reticulum. ASN Neuro 2012;4: microvascular outcomes in patients with type 2 diabetes: a meta-
pii: e00072. analysis of individual participant data from randomised controlled trials.
[412] Zhuang HX, Wuarin L, Fei ZJ, Ishii DN. Insulin-like growth factor (IGF) Lancet Diabetes Endocrinol 2017;5:431–7.
gene expression is reduced in neural tissues and liver from rats with non- [418] Zychowska M, Rojewska E, Kreiner G, Nalepa I, Przewlocka B, Mika J.
insulin-dependent diabetes mellitus, and IGF treatment ameliorates Minocycline influences the anti-inflammatory interleukins and enhances
diabetic neuropathy. J Pharmacol Exp Ther 1997;283:366–74. the effectiveness of morphine under mice diabetic neuropathy.
[413] Ziegler D, Duan WR, An G, Thomas JW, Nothaft W. A randomized J Neuroimmunol 2013;262:35–45.
double-blind, placebo-, and active-controlled study of T-type calcium [419] Zychowska M, Rojewska E, Pilat D, Mika J. The role of some
channel blocker ABT-639 in patients with diabetic peripheral chemokines from the CXC subfamily in a mouse model of diabetic
neuropathic pain. PAIN 2015;156:2013–20. neuropathy. J Diabetes Res 2015;2015:750182.
[414] Ziegler D, Landgraf R, Lobmann R, Reiners K, Rett K, Schnell O, Strom [420] Zychowska M, Rojewska E, Piotrowska A, Kreiner G, Nalepa I, Mika J.
A. Painful and painless neuropathies are distinct and largely Spinal CCL1/CCR8 signaling interplay as a potential therapeutic target -
undiagnosed entities in subjects participating in an educational evidence from a mouse diabetic neuropathy model. Int
initiative (PROTECT study). Diabetes Res Clin Pract 2018;139:147–54. Immunopharmacol 2017;52:261–71.
Copyright © 2020 by the International Association for the Study of Pain. Unauthorized reproduction of this article is prohibited.