肌痛性脑脊髓炎 慢性疲劳综合征的神经免疫模型
肌痛性脑脊髓炎 慢性疲劳综合征的神经免疫模型
DOI 10.1007/s11011-012-9324-8
REVIEW ARTICLE
Abstract This paper proposes a neuro-immune model for may activate microglia via the vagus nerve. Elevated proin-
Myalgic Encephalomyelitis/Chronic fatigue syndrome (ME/ flammatory cytokines together with raised O&NS conspire
CFS). A wide range of immunological and neurological to produce mitochondrial damage. The subsequent ATP
abnormalities have been reported in people suffering from deficit together with inflammation and O&NS are responsi-
ME/CFS. They include abnormalities in proinflammatory ble for the landmark symptoms of ME/CFS, including post-
cytokines, raised production of nuclear factor-κB, mito- exertional malaise. Raised levels of O&NS subsequently
chondrial dysfunctions, autoimmune responses, autonomic cause progressive elevation of autoimmune activity facili-
disturbances and brain pathology. Raised levels of oxidative tated by molecular mimicry, bystander activation or epitope
and nitrosative stress (O&NS), together with reduced levels spreading. These processes provoke central nervous system
of antioxidants are indicative of an immuno-inflammatory (CNS) activation in an attempt to restore immune homeo-
pathology. A number of different pathogens have been statsis. This model proposes that the antagonistic activities
reported either as triggering or maintaining factors. Our of the CNS response to peripheral inflammation, O&NS and
model proposes that initial infection and immune activation chronic immune activation are responsible for the remitting-
caused by a number of possible pathogens leads to a state of relapsing nature of ME/CFS. Leads for future research are
chronic peripheral immune activation driven by activated suggested based on this neuro-immune model.
O&NS pathways that lead to progressive damage of self
epitopes even when the initial infection has been cleared. Keywords Chronic fatigue syndrome . Inflammation .
Subsequent activation of autoreactive T cells conspiring Cytokines . Depression . Tryptophan . Oxidative and
with O&NS pathways cause further damage and provoke nitrosative stress . Mitochondria . Autoimmune
chronic activation of immuno-inflammatory pathways. The
subsequent upregulation of proinflammatory compounds Abbreviations
ME Myalgic Encephalomyelitis
G. Morris WHO World Health Organization
Tir Na Nog, CFS Chronic fatigue syndrome
Pembrey,
PICs Pro-inflammatory cytokines
Llanelli, UK
TNFα Tumor necrosis factor
M. Maes IL-6 Interleukin-6
Maes Clinics @ TRIA, NK Natural killer
Bangkok, Thailand
O&NS Oxidative and nitrosative stress
M. Maes (*) NF-κB Nuclear factor κB
Piyavate Hospital, 2-5A 2′-5′-oligoadenylate
998 Rimklongsamsen Road, RNaseL 2-5A-dependent ribonuclease L
Bangkok 10310, Thailand
PKR Protein kinase R
e-mail: [email protected]
URL: http://scholar.google.co.th/citations? COX-2 Cyclo-oxygenase 2
hl0en&user01wzMZ7UAAAAJ&oi0sra IFNγ Interferon γ
Metab Brain Dis
Th T helper subset (Maes et al. 2012a). Moreover, the data show that
TGF-β1 Transforming growth factor post-exertional malaise is a key component of ME and
Treg T regulatory differentiates ME from CFS (Maes et al. 2012a). Thus, the
ATP Adenosine triphosphate diagnosis of ME, CFS and ME/CFS refer to different con-
LPS Lipopolysaccharide cepts, ranging from chronic fatigue together with physio-
TLR Toll-like receptor somatic symptoms (Fukuda et al. 1994), to a symptom
BBB Blood brain barrier complex where post-exertional malaise is a key factor (Maes
SPECT Single-photon emission computed tomography et al. 2012a). Patients with ME display the unique charac-
PET Positron emission tomography teristic of producing a range of objectively measurable ab-
T MRI Tesla magnetic-resonance imaging normal physiological responses to exercise and a global
HPA Hypothalamic-pituitary-adrenal worsening of symptoms following even trivial increases in
MS Multiple sclerosis cognitive or physical activity (De Becker et al. 2000; Nijs et
IDO 2,3-dioxygenase al. 2005; Lane et al. 1998). Obviously these differences in
TRYCAT Tryptophan catabolite diagnostic classifications and criteria have blurred the
NMDA N-methyl-D-aspartate results of scientific research. In this paper we will use the
NADP Nicotinamide adenine dinucleotide phosphate term ME/CFS, since most research until now has been
STAT3 Signal Transducer and Activator performed on ME/CFS and not on ME as defined recently
of Transcription 3 (Maes et al. 2012a; Carruthers et al. 2011).
Foxp3 Forkhead box P3 Recently an explanatory model has been developed in
PBMCs Peripheral blood mononuclear cells support of the WHO’s classification that considers ME to be
SNPs Single nucleotide polymorphisms a neuro-immune disorder (Maes 2009; Maes and Twisk
2010; Maes et al. 2012b). This model provides considerable
evidence that the pathophysiology of ME/CFS is associated
with immuno-inflammatory pathways, which lead to neuro-
Introduction logical aberrations, including behavioral responses, and
neuro-endocrine, autonomic and brain dysfunctions.
Myalgic Encephalomyelitis (ME) has been described in the The presence of raised levels of the pro-inflammatory cyto-
medical literature since the ‘30s. Characteristics of ME are kines (PICs), tumor necrosis factor (TNF)α, interleukin-6 (IL-
post-exertional malaise, chronic fatigue, autonomic symp- 6) and IL-1β, is a consistent finding in patients with ME/CFS
toms, hyperalgesia, malaise, neurocognitive dysfunctions, who show signs of a chronically activated and highly disregu-
and a chronic and relapsing-remitting course. The World lated immune system. These include the presence of autoim-
Health Organization (WHO) acknowledges ME as a disease mune responses, loss of natural killer (NK) function and raised
of the nervous system (World Health Organization 1992). levels of the anti-inflammatory cytokines. Studies investigating
Much later, in the ‘80s and ‘90s, the diagnosis of chronic the pathophysiology of ME/CFS have consistently revealed a
fatigue syndrome (CFS) was introduced. The definition of wide range of aberrations in oxidative and nitrosative stress
CFS is primarily based upon the presence of chronic fatigue (O&NS) pathways and lowered antioxidant defenses. Perhaps
lasting more than 6 months. According to a commonly used unsurprisingly then evidence of mitochondrial damage has
diagnostic classification of CFS (Fukuda et al. 1994), chron- been reported in patients with ME/CFS. Studies have reported
ic fatigue must be accompanied by four or more of the activation of intracellular signaling molecules, e.g. nuclear
following symptoms: substantial impairment in short-term factor κB (NF-κB) and the 2′-5′-oligoadenylate (2-5A)/2-5A-
memory or concentration; sore throat; tender lymph nodes; dependent ribonuclease L (RNaseL) and the protein kinase R
muscle pain; multi-joint pain without swelling or redness; (PKR) pathways. A new immuno-inflammatory pathway in
headache of a new type, pattern or severity; unrefreshing ME/CFS that may cause neuro-immune dysfunctions and even
sleep; and post-exertional malaise lasting more than 24 h. neurological disease is bacterial translocation.
ME and CFS are therefore different diagnostic concepts Here we propose a key role for changes in immuno-
(Maes et al. 2012a; Twisk and Maes 2009). inflammatory pathways and resultant neuro-inflammation, in
In an attempt to define a more homogenous patient pop- conjunction with aberrations in O&NS pathways, in produc-
ulation, a panel of ME/CFS experts has published consensus ing the wide range of symptoms reported and abnormalities
criteria for ME (Carruthers et al. 2011). Using multivariate identified in people with ME/CFS. This paper proposes that an
statistical analyses, Maes et al. (2012a) showed that the interaction between an activated immune system, O&NS, and
diagnosis of CFS according to Fukuda’s criteria (Fukuda et mitochondrial damage, explains the symptoms of ME/CFS.
al. 1994) defines a heterogeneous population of individuals This is an area of active research and much needs to be
with chronic fatigue, of whom individuals with ME are a confirmed by new results. As such there are gaps, which we
Metab Brain Dis
have filled with established knowledge on immune and neuro- CD8+T cells (Landay et al. 1991), and higher percentages of
immune mechanisms and directions for future research. CD8+T cells and higher numbers of CD8+T cell class 11
activation markers (Klimas et al. 1990). We have reviewed
that a Th2 response with increased levels of IL-4 or IL-5 may
Pathophysiological findings in ME/CFS occur in part of the patients (Maes et al. 2012b; Fletcher et al.
2009). Other studies show signs of immunosuppression, in-
Immuno-inflammatory pathways in ME/CFS cluding lowered NK cell activity and raised levels of immu-
noregulatory cytokines, IL-10 and transforming growth factor
Activation of immuno-inflammatory pathways is a consistent (TGF)-β1 (Chao et al. 1991; Ogawa et al. 1998; Fletcher et al.
finding in ME/CFS (Moss et al. 1999; Cannon et al. 1997). 2010; Bennett et al. 1997; Nakamura et al. 2010). All in all,
Increased levels of PICs, such as IL-1β, TNFα and IL-6 have while a chronic, low-grade inflammatory response accompa-
been reported in people with ME/CFS (Maes et al. 2012a; nies ME/CFS, some patient groups feature elevations of Th1,
Broderick et al. 2010). The levels of IL-1 and TNFα show a Th2, Treg and maybe Th17 cytokines. Some ME/CFS patients
positive significant correlation with fatigue, autonomic symp- show a mixed response with activation of Th1 and Th2
toms and flu like symptoms (Maes et al. 2012b). Other markers cytokines. Th1 and Th2 subsets are indeed not mutually
of inflammation, e.g. lysozyme and polymorph nuclear neutro- exclusive, as Th17 and Treg subsets, but together facilitate
phil elastase, are also elevated (Maes et al. 2012b). The levels recovery from infection (Bot et al. 2004; Afzali et al. 2007).
of the latter correlate significantly and positively with levels of For example, IL-4 and Th2 immune responses monitor Th1
memory and cognitive impairment and phenomenological ex- responses and maintain homeostasis (Bot et al. 2004). It may
perience of infection. Increased levels of acute phase reactants, be that the different cytokine profiles in ME/CFS depend on
e.g. C-reactive protein and the α2 protein fraction obtained subgroups or staging of the disease, e.g. acute flares versus
during protein electrophoresis, further underscore the presence partial remission. In ‘Mechanistic explanations for the chronic
of an inflammatory response (Buchwald et al. 1997; Maes et al. and relapsing–remitting nature of ME/CFS’ we will discuss
2005a; Spence et al. 2008). Also the production of cyclo- how Th1 versus Th2 dominance and Treg functions may play
oxygenase 2 (COX-2) is significantly elevated in patients suf- a role in the relapsing–remitting nature of ME/CFS.
fering from ME/CFS (Maes et al. 2007b). The elevated levels A number of factors predispose to chronic inflammation.
of COX-2 are additionally related to key symptoms of ME/ IgG1 deficiencies often result in a decreased level of total IgG
CFS, such as a subjective experience of infection, hyperalgesia, (hypogammaglobulinemia). Along with IgG1, the IgG3 sub-
fatigue, and neurocognitive disorders (Maes et al. 2007b). class is most frequently present in the antibody response to
Neopterin, an indicant of cell-mediated immune activa- protein antigens. Decreased IgG3 levels are frequently associ-
tion is significantly increased in ME/CFS patients (Maes et ated with IgG1 deficiency (Herrod 1993). IgG1 and IgG3
al. 2012b). The same study showed that severity of fatigue, deficiency has been associated with a history of recurrent
autonomic and flu like symptoms also correlate positively infectious. IgG1 and IgG3 levels are reduced in ME/CFS (Bassi
with serum neopterin concentrations. Some but not all stud- et al. 2008; Hilgers and Frank 1994). Another feature which
ies found increased production of interferon (IFN)γ in ME/ predisposes to chronic inflammation is the decreased ratio of
CFS (review: Maes and Twisk 2010). Increased levels of IL- ω3/ω6 polyunsaturated fatty acids (PUFAs) in patients with
12 were detected by Fletcher et al. (2009), suggesting a ME/CFS (Maes et al. 2005b). Omega-3 fatty acids have anti-
Thelper (Th)1-like response in some patients. No significant inflammatory properties, whereas omega-6 fatty acids have
differences in the Th17 cytokines, IL-17 and IL-23, were pro-inflammatory effects (Simopoulos 2002). A genetic predis-
observed in patients with ME/CFS as compared with con- position is repeatedly observed in patients with ME/CFS. For
trols (Fletcher et al. 2009). Nevertheless, associations be- example, a significant increase in TNF–857 TT and CT geno-
tween fatigue scores and IL-17 levels were established in types, likely to be associated with increased production of
patients with primary Sjögren's syndrome (Haldorsen et al. TNFα and IFNγ; human leucocyte antigen class II allele
2011). This finding together with increased levels of PICs, HLA-DQA1*01; C/T polymorphism leading to His161Arg
including IL-1, IL-6 and TNFα, further suggest that a Th17 substitution in IL-17F; and IFN-γ +874T/A and IL-10 -592C/
response may occur in some patients with chronic fatigue. In A polymorphisms (review: Maes and Twisk 2010).
addition, the lowered expression of CD69 activation
markers on CD4+ T cells in ME/CFS (Mihaylova et al. O&NS pathways in ME/CFS
2007) may further skew towards Th17 responses. Indeed,
CD69 promotes some pathways that inhibit Th17 cell dif- ME/CFS is accompanied by increased reactive oxygen and
ferentiation (Martín and Sánchez-Madrid 2011). nitrogen species (ROS/RNS) and activated O&NS pathways
A state of immune activation is further evidenced by ele- (Jammes et al. 2005; Meeus et al. 2009; Maes et al. 2006b,
vated expression of surface markers CD38+, HLA-DR on 2009b). Frequently reported abnormalities include O&NS
Metab Brain Dis
damage to proteins, lipids and DNA (Maes et al. 2009b; activation by viral products, lipopolysaccharide (LPS), a
Maes and Twisk 2010). Patients with ME/CFS additionally constituent of the outer membrane of gram negative bacte-
show increased IgM-mediated responses to different NO- ria, PICs and free radicals, NF-κβ is translocated from the
adducts, indicating chronically increased nitric oxide (NO) cytoplasma to the nucleus where it binds DNA promoter
production (Maes et al. 2006b), and increased production of sequences and induces transcriptional activation of PICs,
inducible NO synthase (iNOS) (Maes et al. 2007b). In- such as IL-1, IL-6, and TNFα, COX-2 and inducible iNOS
creased IgM-mediated responses directed to palmitic and (Brasier 2006). Upon activation, the inhibitory IκB proteins,
myristic acid and the NO-adducts show that O&NS have that hold NF-κB in an inactive state, are tagged from pro-
modified the structure of fatty acids and proteins (Maes et al. teosomal degradation following phosphorylation by IκB
2006b). Reduced levels of the antioxidants zinc, coenzyme kinases. In many inflammatory disorders, NF-κB in chron-
Q10, dehydroepiandrosterone sulfate and glutathione have ically upregulated. In ME/CFS, the increased production of
also been reported (Manuel-y-Keenoy et al. 2000, 2001; NF-κB is associated with fatigue and a subjective feeling of
Kennedy et al. 2003, 2005; Maes et al. 2005a, 2006a, infection (Maes et al. 2007a). Moreover, there were signif-
2009a; Maes 2011a). Low grade inflammation, activated icant and positive intercorrelations between the production
O&NS pathways and impaired oxidative defenses likely of NF-κB, COX-2, and inducible iNOS, suggesting that the
interact to increase the magnitude of abnormality in each increased production of COX-2 and iNOS is driven by the
leading to a vicious cycle (Maes and Twisk 2010). The transcription factor NF-κB. Thus, increased production of
symptoms of fatigue, malaise and pain, positively and sig- NF-κB is response to viral and bacterial infections may be
nificantly correlate with markers of lipid peroxidation and one key mechanism modulating the immuno-inflammatory
protein oxidation/nitrosylation (Kennedy et al. 2003, 2005; and O&NS pathways in ME/CFS.
Maes et al. 2006b; Maes and Twisk 2010; Maes 2011a). The 2-5A/RNaseL and PKR pathways are also upre-
Studies have demonstrated increased ventricular lactate in gulated in individuals with ME/CFS (Vojdani and Lapp
ME/CFS using 3 Tesla proton magnetic-resonance spectros- 1999). Importantly, the latter pathways have antiprolifer-
copy (Murrough et al. 2010; Mathew et al. 2009; Shungu et ative and antiviral effects and are induced by interferons
al. 2012). Ventricular CSF lactate was significantly in- in response to viral infections. Moreover, the enzyme
creased in ME/CFS as compared to normal volunteers that synthesizes 2-5A, i.e. 2′-5′-oligoadenylate synthe-
(Murrough et al. 2010). During exercise lactate is associated tase, is activated during bacterial infections. PKR is also
with the production of radical oxygen species and acidosis activated by bacterial LPS and PICs, including IL-1 and
(Groussard et al. 2000). Lactate is an antioxidant that scav- TNFα.
enges free radicals and attenuates lipid peroxidation, where-
as acidosis is a potent oxidative condition (Groussard et al. Bacterial translocation
2000).
The presence of O&NS is known to lead to mitochondrial The presence of intestinal permeability and resultant bacte-
damage in neurological diseases (Lin and Beal 2006; Trushina rial translocation into the lamina propina, the lymph nodes
and McMurray 2007) and is likely the source of mitochondrial and possibly the systemic circulation, is evidenced by the
dysfunction seen in people with ME/CFS (Maes and Twisk presence of IgM and IgA mediated immune responses
2010). Specific and highly sensitive alterations in the avail- against LPS of different gram negative bacteria in up to
ability of adenosine triphosphate (ATP), inadequate supplies 40–60 % of people with ME/CFS (Maes et al. 2012c). This
of ATP, and oxidative phosphorylation have been reported in may be due to the presence of systemic inflammation, which
neutrophil mitochondria of individuals with ME/CFS (Myhill can reduce the integrity of epithelial tight junctions. The IgA
et al. 2009; Vermeulen et al. 2010). Another study reported responses positively and significantly correlate with the
that all individuals with physio-somatic symptoms in depres- levels of PICs, such as IL-1β and TNFα, and neopterin
sion had a significantly lower ATP production in muscle (Maes et al. 2012c). LPS from translocated commensal
biopsies compared to controls (Gardner and Boles 2008). bacteria may provoke a peripheral immuno-inflammatory
response by binding to the Toll-like receptor-4 (TLR4)
Intracellular signaling networks complex, which in turn activates intracellular signaling
pathways, such as NF-κB, which subsequently will induce
A number of different signaling networks are upregulated in genes of PICs, iNOS and COX-2 (Maes et al. 2012c).
ME/CFS. Studies have demonstrated higher production of Increased LPS may also explain other immuno-
unstimulated, and TNFα- and PMA-stimulated NF-κB, inflammatory signs in ME/CFS, such as increased neopterin
cyclo-oxygenase-2 and inducible iNOS (Maes et al. 2007a, and elastase levels. Indeed, LPS may stimulate the produc-
b). NF-κβ is a major upstream molecule, which regulates tion of neopterin by monocytes/macrophages and elastase
immuno-inflammatory and O&NS responses. Upon by neutrophils and monocytes (Maes et al. 2012c).
Metab Brain Dis
Fig. 1 Effects of pathogens on the immune system leading to immuno- inflammatory cytokines (PICs), e.g. tumor necrosis factor-α (TNFα),
inflammatory cascades. Different pathogens in ME/CFS may activate interleukin-1 (IL-1) and IL-6, are increased in individuals with ME/
the immuno-inflammatory pathways, reactive oxygen and nitrogen CFS. This disease can feature elevations of Thelper (Th)1, Th17, Th2
species (ROS/RNS), oxidative and nitrosative stress (O&NS) path- and Tregulatory (Treg) cytokines. Increased O&NS, through the for-
ways, and nuclear factor κB (NF-κB) through the Toll-like receptor mation of neoepitopes, and enhanced Th17 and Th1 responses increase
(TLR) complex, and consequently inducible nitric oxide synthase the risk to develop autoimmune reactions
(iNOS) and cyclo-oxygenase-2 (COX-2). Consequently, pro-
Different infections can initiate or exacerbate a chronic in- cells (Fujinami et al. 2006). The latter cells are subsequently
flammatory condition and cause autoimmune responses, using lysed by cytotoxic CD4+T cells (Hahn et al. 1995).
processes described as molecular mimicry, bystander damage It is also possible that the initial infection may create autor-
and epitope spreading (Fujinami et al. 2006). Autoreactive T eactive T cells, but insufficient inflammation to induce overt
cells can be formed when the pathogen contains epitopes, which pathology (Hahn et al. 1995). Nevertheless, autoreactive T cells
are cross reactive to host molecules, leading to autoimmune can be activated by subsequent infections with a broad range of
targeting of both pathogen and self epitopes. Epitope spreading pathogens leading to overt inflammatory damage (Hahn et al.
and bystander damage are other sources of chronic immune 1995). This fertile field, induced by the interaction with the
activation, inflammation and autoimmune responses. Bystander primary infecting agent, can change with time (Fujinami et al.
cell death, which is caused by pathogen specific T cells, may 2006). The initial priming infection and subsequent challenges
increase the release of self-epitopes that induce de novo T cell that produce symptoms may be separated by many years and
responses. Pathogen-specific T cells may initiate bystander acti- sometimes more than one challenge is needed to induce symp-
vation by migrating to areas of infection, where they encounter toms. A fertile field can also be generated when an infection
pathogen-infected cells that present pathogen peptides in the with a pathogen, which shows molecular mimicry to self CNS
context of MHC human leukocyte antigen class I molecules to molecules, primes autoreactive T cells without initiating auto-
T cells. The CD8+ T cells recognize these infected cells and will immune inflammatory disease. Later challenges may then trig-
release cytotoxic granules which may result in the killing of the ger these autoimmune cells to cause overt symptoms (Fujinami
infected cell. Consequently, the dying cells, the CD8+ T cells et al. 2006).
and macrophages within the inflammatory focus, release PICs
and ROS/RNS, which may cause bystander damage of unin-
fected but neighbouring cells (Fujinami et al. 2006). This may From peripheral inflammation to neuro-inflammation
result in additional immunopathology (Duke 1989; Smyth and
Sedgwick 1998), because self proteins may be released and It is well known that peripheral inflammation may induce a
engulfed by dendritic cells, macrophages and antigen presenting behavioral complex characterized by fever, anorexia, weight
Metab Brain Dis
loss, and behavior inhibition, including reduced locomotor peripheral immuno-inflammatory signals activate the produc-
activity and listlessness (Goehler et al. 2005). Circulating tion of PICs in the amygadala, which acts to coordinate
PICs and other inflammatory and O&NS molecules may behavioral, CNS and autonomic responses (Engler et al.
affect brain function through several routes, mainly humoral 2011). These bottom-up pathways may explain that systemic
and neural pathways (Ferrari and Tarelli 2011). The blood inflammation is accompanied by neuro-inflammation and be-
brain barrier (BBB) controls the passage of inflammatory havioral changes. For example, systemic LPS administration
compounds from the plasma to the brain. There are several may elicit neuro-inflammation with increased brain TNFα,
ways by which these substances can cross the BBB (Perry et which remains elevated for months and is associated with
al. 2010). Firstly, these substances can enter the brain through behavior symptoms, including fatigue, malaise, and hyper-
the areas that lack a BBB, e.g. the circumventricular organs alagesia (Qin et al. 2007). Moreover, once the immuno-
(Whitton 2007). Secondly, cytokines may cross the BBB inflammatory signals have arrived in the CNS via the vagal
using specific transporters (Kim and de Vellis 2005). Thirdly, afferents, a counter-regulatory anti-inflammatory reflex re-
BBB permeability may be increased as a consequence of sponse is mounted that tends to downregulate the peripheral
immuno-inflammatory stimuli. For example, Th17 T cells immuno-inflammatory response (Perry et al. 2007; Tracey
can infiltrate the brain through the BBB via loosening of the 2002). These counter-regulatory responses will be discussed
endothelial tight junctions by increased secretion of IL-17 and in ‘Mechanistic explanations for the chronic and relapsing–
IL-22 (Jadidi-Niaragh and Mirshafiey 2011). Fourthly, periph- remitting nature of ME/CFS’.
eral stimuli may activate endothelial cells which in turn may
induce immune signals in the CNS (Ransohoff and Perry
2009). For example, Parkinson’s Disease is accompanied by Neurological dysfunctions in ME/CFS
breakdown of the BBB which allows noxious blood-borne
substances to penetrate in the CNS (Kortekaas et al. 2005). Single-photon emission computed tomography (SPECT)
Likewise, disruption of the BBB allows the extravasation of scans may reveal significantly lower cortical/cerebellar re-
PICs and immune cells, which can activate microglia and, gional cerebral blood flow frequently in the frontal, parietal,
therefore, induce neuroprogression (Leonard and Maes 2012). temporal, occipital, brain stem and throughout the cerebral
Neuroprogression is a progressive process, which is induced cortex (Schwartz et al. 1994; Biswal et al. 2011; Patrick et
by inflammatory and O&NS processes, and that is character- al. 2008). Positron emission tomography (PET) scans may
ized by neurodegeneration, reduced neurogenesis and neuro- reveal decreased density of 5-HT transporter in the brain
nal plasticity, and/or neuronal apoptosis (Berk et al. 2011). (Yamamoto et al. 2004). Proton magnetic resonance spec-
The second route of immune-to-brain communication is troscopy has revealed various abnormalities in metabolic
the neural pathway, which transmits peripheral inflammatory pathways in the brain (Chaudhuri et al. 2003; Chaudhuri
signals to the CNS via the autonomic nervous system. The and Behan 2004). Barnden et al. (2011) detected that white
most important afferent pathway that signals peripheral in- matter volume decreased with fatigue duration and they
flammation to the CNS is the vagus nerve. These neural found signs of impaired cerebrovascular autoregulation.
pathways are stimulated by inflammatory signals that increase The findings suggest that fatigue onset is accompanied by
the levels of brain cytokines (Perry et al. 2003, 2010; Tracey an insult that suppresses cognitive activity and modulates
2002). The role of the vagus nerve is crucial in inducing the autonomic system (Barnden et al. 2011). Elevated numb-
neuro-inflammation and activation of microglia (Goehler et ers of punctuate lesions have been detected in ME/CFS
al. 1997, 2005; Herkenham et al. 1998; Ericsson et al. 1997; without psychiatric comorbidity in the frontal lobes as com-
Benarroch 2011; Blatteis and Sehic 1997; Ek et al. 1998; pared to controls (Lange et al. 1999). 3 Tesla magnetic-
Saper 1995; Gallaher et al. 2012; Riazi et al. 2008; Engler et resonance imaging (3 T MRI) scans have revealed signifi-
al. 2011). Not only PICs, but also prostaglandins and comple- cant neuroanatomical changes, including reduced white
ment factors may activate the vagal nerve pathways, via matter volume (Puri et al. 2011). A 1.5 T MRI, on the other
chemoreceptors situated on glomus cells in the vagal para- hand, showed no abnormalities suggesting brain atrophy or
ganglia (Goehler et al. 1997). The latter serve as a monitoring white matter lesions in patients with ME/CFS (Perrin et al.
system that detects immuno-inflammatory stimuli in the 2010). Greco et al. (1997) found no specific white matter
lymph and spleen. Vagal afferents terminate in the dorsal abnormalities in ME/CFS. On SPECT imaging, ME/CFS
vagal complex of the caudal medulla, the area postrema, the shares some similarities with AIDS Dementia Complex:
nucleus of the solitary tract and the dorsal motor nucleus of the acute changes in radionuclide uptake in the younger popu-
vagus (Saper 1995). Ascending immuno-inflammatory sig- lation may be caused by inflammatory processes at the
nals activate microglia in the nucleus of the solitary tract and cellular or micro-vascular level (Schwartz et al. 1994). The
the doral motor nucleus of the vagus (Gallaher et al. 2012) and findings in ME/CFS are consistent with the hypothesis that
the hippocampus (Riazi et al. 2008). Subsequently, these this disease results from a viral infection of neurons, glia or
Metab Brain Dis
vasculature (Schwartz et al. 1994). Thus, viral infection can In ME/CFS there are significant correlations between typ-
provoke neurological dysfunction by interfering with intra- ical symptoms, such as fatigue, malaise, a subjective feeling of
cellular mechanisms or membrane transport systems or cere- infection, sleep disorders, autonomic symptoms and post exer-
bral hypoperfusion due to vasculitis. All in all, these brain tional malaise and activated O&NS (see above) and immuno-
studies could indicate metabolic dysfunctions, neuroinflam- inflammatory pathways, including increased neopterin and
mation or maybe neuroprogression in patients with ME/CFS. TNFα concentrations (Maes et al. 2012a). This indicates that
Given the different findings on activated immuno- these characteristic symptoms of ME/CFS are in part mediated
inflammatory pathways in ME/CFS, it comes as no surprise through effects of immuno-inflammatory and O&NS path-
therefore that autonomic (Newton et al. 2007; Winkler et al. ways. We have previously reviewed that PICs, e.g. IL-1 and
2004) and endocrine abnormalities (Van Den Eede et al. 2007; TNFα, and Th1-like cytokines, such as IFNγ, activated
Demitrack et al. 1991; Demitrack and Crofford 1998) accom- O&NS pathways and lowered antioxidant levels, including
pany ME/CFS. Recently we have reviewed the autonomic coenzyme Q10 and zinc, may elicit the characteristic symp-
dysfunctions and orthostatic abnormalities in ME/CFS (Maes toms of ME/CFS, such as fatigue, malaise, and neurocognitive
and Twisk 2009). Autonomic symptoms are associated with and autonomic symptoms (Maes and Twisk 2010; Maes et al.
cardiovascular abnormalities and may be explained by acti- 2012b; Anderson et al. 2012). For example, PICs are heavily
vated immuno-inflammatory, intracellular and O&NS path- implicated in the cause of fatigue in multiple sclerosis (MS)
ways, including increased TNFα, NO-related mechanisms (Heesen et al. 2006). Fatigue is also one of the most disabling
and NF-κB (Maes et al. 2011; Maes and Twisk 2009). symptoms in MS, with up to two thirds of patients describing
Hypothalamic-pituitary-adrenal (HPA) axis hypofunction has fatigue as their main complaint (Schwid et al. 2002). TNFα, as
been established in part of individuals with ME/CFS (Scott et a principal proinflammatory mediator, is associated with MS-
al. 1998). This entails decreased production of cortisol, related fatigue. This is in support of a pathogenic role of the
blunted diurnal variation of cortisol, and blunted responses MS-related inflammatory process in the development of fa-
to a variety of provocation tests, including psychological and tigue (Flachenecker et al. 2004). Also, the production of Th1
physical stressors (under review). Also these alterations in cytokines, e.g. IFNγ, in MS patients is associated with the
HPA-axis function may be explained by immuno- extent of fatigue (Pokryszko-Dragan et al. 2012). Other stud-
inflammatory and O&NS effects, including increased NO ies have reported that TNFα, IL-1β and IL-6 could be in-
production attenuating HPA-axis functions (under review). volved in the pathophysiology of mental fatigue through their
ability to attenuate the astroglial clearance of extracellular
glutamate (Rönnbäck and Elisabeth Hansson 2004). An ad-
Mechanistic explanations for the onset of ME/CFS verse effect caused by specific PICs, such as IL-1, on learning
symptoms including post exertional malaise and memory capacities, has been observed in several experi-
mental systems. IL-6 facilitates lipopolysaccharide-induced
This section will propose an immuno-inflammatory explana- disruption in working memory and expression of other PICs
tion for the hallmark symptoms of the disease, including (Sparkman et al. 2006). TFNα and IL-1β regulate sleep
fatigue, malaise, neurocognitive symptoms and a range of architecture together with IL-6 (Krueger and Majade 1987;
abnormal responses to exertion that may be delayed by 24 or Cavadini et al. 2007; Obal and Krueger 2003) and can lead to
even 48 h, which is often labeled as post exertional malaise sleep wake reversal and even complete loss of circadian
(VanNess et al. 2010; Van Oosterwijck et al. 2010; Carruthers control (Obal and Krueger 2003).
et al. 2011). The term refers to abnormal responses to even a As explained above, bacterial translocation through bind-
trivial increase in normal levels of physical or neurocognitive ing to the TLR4 complex induces NF-κB and immuno-
activity. Post activity relapse may therefore be a better term. inflammatory and O&NS pathways and thus may cause be-
This phenomenon is often reported by ME patients to be havioral and neuro-immune changes, including autoimmune
similar to the acute phase of influenza, with symptoms indi- responses directed against gangliosides (Maes et al. 2012c).
cating infection/inflammation, e.g. sore throat, lymph tender- Raised levels of IFNγ, IL-1, IL-2 and TNFα activate the
ness or swelling, malaise, hyperalgesia and brain fog (Twisk tryptophan-degrading enzyme, indoleamine 2,3-dioxygenase
and Maes 2009). Insignificant increases in physical activity or (IDO), which causes alterations in the tryptophan catabolite
minor cognitive tasks may exacerbate immune dysfunction, (TRYCAT) pathway (Anderson et al. 2012). Increased IDO
inflammation and O&NS thereby producing further signs of activation lowers plasma tryptophan and enhances the pro-
the disease (Twisk and Maes 2009). The effect may be duction of neurotoxic catabolites, e.g. quinolinic acid, a strong
delayed, but is predictable and accumulative, varying in dura- agonist of the glutamatergic N-methyl-D-aspartate (NMDA)
tion dependant on disease severity and accumulative activity receptor. Decreased tryptophan levels may cause physio-
levels (VanNess et al. 2010; Van Oosterwijck et al. 2010; somatic symptoms, such as fatigue, autonomic symptoms,
Carruthers et al. 2011). hyperalgesia and somatic presentations (Anderson et al.
Metab Brain Dis
2012). Disorders in the TRYCAT pathway are detected in during this “recharge” period, such as stress, will greatly
post-viral fatigue syndrome (Anderson et al. 2012). delay recharge and possibly create longer term disability by
In the presence of increased levels of IL-1 and TFNα, ATP further damaging the inner membrane. Chronically elevated
consumption is greatly accelerated, and levels of anaerobic cytokines act on the brain and hypothalamus to create a new
glycolysis and glucose consumption are increased (Berg et al. homeostatis of glucose metabolism, and the autonomic ner-
2003). The combined effects of these actions cause a greater vous system controlling heart rate, respiratory rate, blood
metabolic demand even in the resting state (Berg et al. 2003). pressure, vascular tone, gastric peristalsis, etc. (Del Rey et
TNFα increases ROS production in the mitochondrial elec- al. 2006). The autonomic symptoms observed in ME/CFS
tron transport chain. NO is known to inhibit mitochondrial could be obvious consequences of such a shift.
respiration, due to reversible binding to cytochrome C oxidase Increased TNFα and O&NS, iNOS, low ATP and high
thus competing with oxygen and inhibiting energy production TGFβ1 levels deplete glutathione (Castagna et al. 1995; Phelps
(Brown and Bal-Price 2003). Moreover, NO can be converted et al. 1995). A depletion in glutathione levels is often accom-
to other reactive species, including peroxynitrite, NO2, N2O3, panied by a partial blockade in the methylation cycle. More-
and S-nitrosothiols, that may inhibit mitochondrial respiration over, nitric and nitrous acid inhibit methionine synthase (Layser
and activate mitochondrial permeability transition, which 1978; Danishpajooh et al. 2001; Kerkeni et al. 2006; Culley et
eventually may trigger neuronal apoptosis or necrosis (Brown al. 2007). A number of studies mentioned earlier have demon-
and Bal-Price 2003). PICs can directly disrupt glucose ho- strated reduced glutathione in people with ME/CFS. A recent
meostasis and the availability of ATP. TNFα, IL-1β and IFNγ study confirmed these earlier findings and found that low
may significantly inhibit insulin secretion by beta cells in the glutathione levels and high lactate levels correlated with meas-
pancreas (Kiely et al. 2007). This increases cellular glucose urements of physical health and disability (Shungu et al. 2012).
consumption and lactate and glutamate levels (Kiely et al. Figure 2 summarizes how the different immuno-
2007). The resultant decrease in ATP may shift cellular me- inflammatory pathways may cause the typical symptoms of
tabolism towards a catabolic state, showing how mitochon- ME/CFS.
drial dysfunction can severely impair glucose metabolism.
PICs in general lead to the upregulation of nicotinamide
adenine dinucleotide phosphate (NADP) oxidase and resultant Mechanistic explanations for the chronic and relapsing–
increased production of ROS (Morgan et al. 2007). This remitting nature of ME/CFS
process may generate superoxide anion dependent damage
and formation of peroxinitrite which subsequently may con- ME/CFS is characterized by highly increased IgM-related
tribute to permanent damage to mitochondria (Mohankumar autoimmune response directed against disrupted lipid mem-
et al. 1991; Shintani et al. 1993; Linthorst et al. 1995). IL-1 not brane components, by-products of lipid peroxidation, an-
only causes hypoglycemia, but also resets glucose homeosta- chorage molecules, and NO-modified amino-acids as
sis in the brain; both phenomena together favor an increased compared with normal controls (Maes et al. 2006b). These
uptake of glucose by immune cells during inflammation (Del results show that self-epitopes, which are normally not
Rey et al. 2006). All in all, glucose usage can easily exceed detected by the immune system, have become immunogenic
supply and the increased demand for ATP cannot be met since following damage by O&NS (Maes et al. 2006b). A distinc-
mitochondrial functions are compromised by TNFα and NO. tive pattern of proteins modified by oxidative stress in
Cytokines can also lead to cerebral hypoperfusion via pro- people with ME/CFS was also reported in a recent study
longed exposure to IL-1β (Maher et al. 2003). The ensuing involving proteomic analysis (Schutzer et al. 2011).
lack of glucose would potentially explain the brain fog that so Such antibodies directed against lipids have also been
many people with this illness complain of. detected in remitting-relapsing MS (Geffard et al. 2002;
TNFα reduces mitochondrial respiration via effects at the Boullerne et al. 1996). However the latter study reported
level of complex III of the electron transport chain and by that there was no significant level of IgM against azelaic
ROS production (Samavati et al. 2008; Gudz et al. 1997). acid detected, whereas Maes et al. (2006b) found increased
The latter may induce opening of mitochondrial permeabil- IgM responses directed against azelaic acid in ME/CFS
ity transition pores, which may cause uncoupling of oxida- patients. The circulating IgM antibody titers in MS appear
tive phosphorylation (Chernyak 1997). The condition of to be related to the presence of inflammation, because they
increased ATP consumption and mitochondrial damage increase during relapses and decrease during remissions
means that exceeding a threshold of physical and mental (Bodet et al. 2004). This suggests that the level of immune
exertion will lead to all available ATP becoming exhausted activation, O&NS and magnitude of autoimmunity may in
and thus a crash may ensue. It takes damaged mitochondria fact be greater in ME/CFS than in MS.
time to replenish said ATP, leading to a profound post- Increased levels of peroxinitrite and nitrosothiols and low-
exertional disability. Anything that increases TNFα activity ered levels of reduced glutathione have been found in the CSF
Metab Brain Dis
ME / CFS
Mitochondria Physio-somatic ss
ATP Disabling fatigue
Glutathione PEM
Brain and
Neuro-immune Mental fatigue, neuro-
disorders cognitive ss
Fig. 2 Immuno-inflammatory pathways causing the typical symptoms and epitope spreading, and O&NS damage to self-epitopes. PICs and
of Myalgic Encephalomyelitis (ME) and chronic fatigue syndrome O&NS processes may activate the tryptophan catabolite (TRYCAT)
(CFS). Different pathogens activate nuclear factor κB (NF-κB) and pathway, cause mitochondrial dysfunctions including lowered ATP
consequently cyclooxygenase 2 (COX-2), inducible nitric oxide syn- levels and brain dysfunctions. Upon activation of NF-κB, tumor sup-
thase (iNOS), oxidative and nitrosative stress (O&NS) pathways and pressor p53 may be downregulated. Translational research has shown
pro-inflammatory cytokines (PICs). Autoimmune responses may ensue that these biological aberrations may cause the different symptoms (ss)
through different mechanisms, including bystander damage, mimicry of ME and CFS, including post-exertional malaise (PEM)
of patients with MS, suggesting a role for nitrosative stress in and a cascade of further inflammatory damage and dysfunc-
MS (Calabrese et al. 2003). This situation is also found in ME/ tion of mitochondria, constituting a flare up of active disease
CFS, as discussed earlier, and therefore following factors or relapse.
could contribute to relapses in MS and ME/CFS. Innate pat- We tentatively propose that (partial) remission is induced
tern recognition receptors recognize O&NS damaged self by the response of the CNS to immune activation. The CNS
epitopes, and the various components of the innate immune modulates immune responses through neuronal and hor-
system, including innate IgM antibodies, collaborate to medi- monal effects. Thus, the sympathetic and parasympathetic
ate their removal (Weismann and Binder 2012). In MS, IL-6 system and neuroendocrine axes attenuate immune
and TNFα levels are significantly increased during relapses as responses, whereas the peripheral nervous systems reinforce
compared to remission (Hautecoeur et al. 1997), as are IL-17 local immune responses (Sternberg 2006). In the first stage,
and IL-23 (Wang et al. 2011). Plasmacytoid dendritic cells, the peripheral immune and nervous systems work together
which are elevated in the CNS during relapses, may be to activate local immune responses and to eliminate invad-
explained by either virus infections or down regulatory pro- ing pathogens. In a later stage, however, counter-regulatory
cesses (Longhini et al. 2011). This dendritic subset is impor- neuronal and neuroendocrine mechanisms are mounted that
tant in stimulating or downregulating effector T cells in MS tend to terminate inflammation and thus restore host homeo-
(Geffard et al. 2002; Bayley-Bucktrout et al. 2008). By stim- stasis (Sternberg 2006). Activation of the vagus nerve indu-
ulating the maturation of dendritic cells, and upregulating the ces the release of transmitters that facilitate Th2 responses
membrane expression of CD80, CD86, CD83, and HLA-DR (Liang et al. 2011). Activation of the vagus nerve decreases
(Csillag et al. 2010), O&NS induces maturation of dendritic the expression of M1 monocytes, which produce TNFα, IL-
cells (Kantengwa et al. 2003). Moreover, NF-κB is activated 6, and IL-1β. Together, this indicates that activation of the
by threshold levels of oxidative stress (Khalaf et al. 2010) vagus neural circuit produces an anti-inflammatory cellular
leading to the activation of autoreactive T cells. Thus chronic milieu (Valdes-Ferrer et al. 2010).
inflammation, due to the causes mentioned earlier, leads to a The role of the peripheral nervous system in modulating
cycle where there is increasing damage to neoepitopes, ulti- the initial effects of immune activation and tissue inflam-
mately leading to a surge of PICs due to immune activation mation is complex and beyond the scope of this paper. The
Metab Brain Dis
antagonistic actions of the factors amplifying immuno- which may lead to a gradual worsening of symptoms. In-
inflammatory pathways and the CNS would potentially creasing levels of mitochondrial damage however act to
account for a relapsing-remitting pattern. The findings that “switch on” the transcription of p53. This can have the
the T cell pool of remitted MS patients is in a state of anergy effects of raising levels of Foxp3 and thus a reduction in
(Fransson et al. 2009) is of particular interest. Thus, active symptoms. P53 activation can also induce cell-cycle arrest
MS is associated with activated Th1/Th17 responses as and allow mitochondrial healing. A drop in free radical
discussed earlier, while it appears that people in remission production acts to suspend the activation of p53. This allows
have their immune system biased towards Th2 or Treg the levels of NF-κB and oxidative stress to downregulate
phenotypes. The immune abnormalities reported in people p53 and Foxp3, and remission is terminated. A number of
with ME/CFS vary between Th1 and maybe Th17, Th2 and such cycles are possible with a worsening of active disease
Th2/Treg responses. This is the pattern one would expect if over time. The presence of raised anticardiolipin antibodies
the relapsing-remitting nature of ME/CFS is produced by in up to 95 % of patients with ME/CFS (Klein and Berg
the interactions described above. A Th17 profile is induced 1995) is consistent with the presence of peroxidative mito-
by prolonged elevation of IL-6 and IL-23, which are in chondrial damage and loss of cytochrome C oxidase func-
themselves the product of elevated O&NS pathways and tion (Paradies et al. 1998).
chronic inflammation. IL-6 inhibits forkhead box P3 (scur-
fin, Foxp3) by activating the transcription of Signal Trans-
ducer and Activator of Transcription 3 (STAT-3) and hence Future research
attenuates the functions of Treg cells (Samanta et al. 2008;
Korn et al. 2009). Also, IL-23 inhibits Foxp3 and thus Future research should further examine the different inflam-
promotes Th17 differentiation (Zhu et al. 2010; Lal et al. matory and O&NS pathways, the transition to autoimmune
2011; Mus et al. 2010). responses and damage to mitochondria, the intracellular
Another plausible explanation for the chronic relapsing signaling networks involved, neuro-inflammation and brain
remitting nature of the disease revolves around tumor sup- metabolic changes in relation to the new diagnostic criteria
pressor p53. P53 is activated by factors that cause DNA (Maes et al. 2012a). We suggest that future research should
damage, including free radicals. P53 arrests the cell cycle, be based on cohort selection using PET scans to detect
activates DNA repair and initiates apoptosis. P53 is also activated microglia (Gerhard et al. 2006). We would suggest
crucial in matching increased mitochondrial function to objective testing for the existence of BBB permeability
increases in demand for ATP (Niu et al. 2005; Matoba et using dynamic Gadolinium-DTPA MRI scanning (Larsson
al. 2006). P53 regulates energy metabolism by balancing and Tofts 1992). The initial challenge would be to isolate
glycolysis and oxidative phosphorylation. In conditions of people with ME from those with CFS and CF (Maes et al.
oxidative stress, p53 activation results in decreased glycol- 2012a) via the use of PET, SPECT scans, 3 T MRI or
ysis. In such an environment, p53 expression acts to upre- Gadolinium-DTPA MRI.
gulate glutathione. The ultimate aim of both functions is the Mass spectroscopy and proteomic approaches for detect-
reduction of oxidative stress. p53 also regulates mitochon- ing and characterizing nitrosatively and oxidatively modi-
drial oxygen production via the synthesis of cytochrome C fied lipids, proteins and DNA produce specific sensitive and
oxidase. In normal circumstances activation of p53 leads to reproducible results (Aulak et al. 2001). Luminex assays for
an upregulation of mitochondrial respiration. NF-κB and the determination of cytokine levels in supernatants from
p53 have antagonistic strategies in the cell and therefore mitogen stimulated cultured peripheral blood mononuclear
cannot function simultaneously in the same cell (Ak and cells (PMBCs) are regarded as producing reliable and re-
Levine 2010). Thus, upon activation of one of these two producible results while minimizing intra and inter labora-
transcription factors the other is downregulated. Moreover, tory variation (Griffiths et al. 2002; Fulton et al. 1997; Fulya
STAT-3 activation leads to the downregulation of p53 ex- et al. 2006; de Jager et al. 2009). Real time reverse tran-
pression (Niu et al. 2005), while the latter is required for scriptase PCR provides a sensitive and quantitative measure
Foxp3 expression (Jung et al. 2010). of cytokine mRNA levels in PBMCs. STAT gene expres-
Therefore, it may be hypothesized that ME/CFS may be sion, p53 expression and Foxp3 transcription should be
accompanied by lowered p53 expression. Lower transcrip- measured.
tion of p53 causes a shift from ATP production by oxidative Mitochondrial dysfunction and ATP levels should be
phosphorylation to glycolysis. This can lead to a severe assessed using a combination of nuclear magnetic resonance
energy deficit and high levels of lactate production (Xiang spectroscopy, (31)P magnetic-resonance spectroscopy and
et al. 2010; Holley and St Clair 2009). Thus, increased blood tests measuring ATP synthesis in PMBCs (Dykens et
levels of NF-κB as observed in ME/CFS may cause a al. 2008; Chaumeil et al. 2009). Exercise stress testing has
gradual downregulation of p53 (Holley and St Clair 2009), the potential to do harm to people with ME/CFS and thus
Metab Brain Dis
the non-ischemic repeat handgrip test should be considered and other inflammatory and O&NS compounds are detected
as a simple non-invasive test for mitochondrial dysfunction by the vagus nerve and other bottom-up signals leading to
(Meulemans et al. 2007). The aerobic forearm test provides the activation of microglia and neuro-inflammation. The
the basis of a simple measure of mitochondrial function and latter and O&NS pathways may persist once the initial
could be adapted to perform measurements over a 3 day infection has been cleared. Elevated and persistent O&NS
period (Garrabou et al. 2006). lead to damage to lipids, proteins and DNA which then
Genetic polymorphisms in the DNA repair mecha- become recognized by the immune system as non self. This
nisms and O&NS pathways, single nucleotide polymor- leads to the production of autoreactive T cells which cause
phisms (SNPs) in the NF-κB and p53 pathways may further damage. Descent into overt autoimmunity may be
further reveal genetic vulnerabilities to develop ME/ precipitated by further infections via molecular mimicry,
CFS. Next generation sequencing offers a rapid and bystander activation and epitope spreading. This ongoing
sensitive method for detecting the wide range of SNPs insidious process increases the concentration of PICs via a
in the human genome and offers promise for investigat- number of mechanisms and activates NF-κB, COX-2 and
ing SNPs in patients with ME and CFS now that sci- iNOS. These processes may also involve enhanced STAT-3
entific diagnostic criteria have been established (Maes et leading to the depletion of Foxp3 and chronic activation of
al. 2012a; Goya et al. 2010; Karchin 2009). the immune system. The combination of existing abnormal-
ities leads to mitochondrial damage and ATP deficit, auto-
nomic dysregulation, and neuroendocrine and brain
Conclusion changes, including lowered cerebral blood flow and white
matter volume, and maybe inflammatory lesions in the
Figure 3 shows the neuro-immune pathways that may lead brain. Increased PICs, O&NS and mitochondrial damage
to ME/CFS. A prolonged severe but non persistent infection and ATP deficit are responsible for the hallmark symptoms
by a number of potential pathogens may result in chronic of the disease including post exertional malaise. The antag-
activation of immune responses. The latter involve the acti- onism between peripheral immune activation, chronic in-
vation of immuno-inflammatory and O&NS pathways and flammation and CNS responses is held to be responsible for
depletion of the body’s antioxidant defenses. Elevated PICs the relapsing-remitting pattern of the disease.
Fig. 3 The neuro-immune
pathways that are involved in
the pathophysiology of ME/
CFS and may cause the typical
symptoms of ME and CFS (see
Conclusion, and Figs. 1 and 2
for explanation)
Metab Brain Dis
Acknowledgments The authors would like to thank Victoria Storey Boullerne A, Petry KG, Geffard M (1996) Circulating antibodies
and Jane Clout for secretarial services. There was no specific financial directed against conjugated fatty acids in sera of patients with
support for this study. The authors declare that they do not have a multiple sclerosis. J Neuroimmunol 65:75–81
conflict of interest. Brasier AR (2006) The NF-kappaB regulatory network. Cardiovasc
Toxicol 6(2):111–130
Broderick G, Fuite J, Kreitz A, Vernon SD, Klimas N, Fletcher MA
(2010) A formal analysis of cytokine networks in chronic fatigue
References syndrome. Brain Behav Immun 24:1209–1217
Brown GC, Bal-Price A (2003) Inflammatory neurodegeneration me-
diated by nitric oxide, glutamate, and mitochondria. Mol Neuro-
Afzali B, Lombardi G, Lechler RI, Lord GM (2007) The role of T helper biol 27:325–355
17 (Th17) and regulatory T cells (Treg) in human organ transplan- Buchwald D, Wener MH, Pearlman T, Kith P (1997) Markers of
tation and autoimmune disease. Clin Exp Immunol 148(1):32–46 inflammation and immune activation in chronic fatigue and
Ak P, Levine AJ (2010) p53 and NF-κB: different strategies for chronic fatigue syndrome. J Rheumatol 24(2):372–376
responding to stress lead to a functional antagonism. FASEB J Calabrese V, Scapagnini G, Ravagna A et al (2003) Disruption of thiol
24(10):3643–3652 homeostasis and nitrosative stress in the cerebrospinal fluid of
Anderson G, Maes M, Berk M (2012) Biological underpinnings of the patients with active multiple sclerosis: evidence for a protective
commonalities in depression, somatization, and Chronic Fatigue role of acetylcarnitine. Neurochem Res 28:1321–1328
Syndrome. Med Hypotheses 78(6):752–756 Cannon JG, Angel JB, Abad LW et al (1997) Interleukin-1 beta,
Aulak KS, Miyagi M, Yan L et al (2001) Proteomic method identifies interleukin-1 receptor antagonist, and soluble interleukin-1 recep-
proteins nitrated in vivo during inflammatory challenge. Proc Natl tor type II secretion in chronic fatigue syndrome. J Clin Immunol
Acad Sci USA 98:12056–12061 17:253–261
Bankhead T, Chaconas G (2007) The role of VlsE antigenic variation Carruthers BM, van de Sande MI, De Meirleir KL et al (2011) Myalgic
in the Lyme disease spirochete: persistence through a mechanism encephalomyelitis: international consensus criteria. J Intern Med
that differs from other pathogens. Mol Microbiol 65:1547–1558 270:327–338
Barnden LR, Crouch B, Kwiatek R et al (2011) A brain MRI study of Castagna A, Le Grazie C, Accordini A, Giulidori P, Cavalli G, Bottiglieri
chronic fatigue syndrome: evidence of brainstem dysfunction and T, Lazzarin A (1995) Cerebrospinal fluid S-adenosylmethionine
altered homeostasis. NMR Biomed 24:1302–1312 (SAMe) and glutathione concentrations in HIV infection: effect of
Bassi N, Amital D, Amital H, Doria A, Shoenfeld Y, Shoenfeld Y parenteral treatment with SAMe. Neurology 45(9):1678–1683
(2008) Chronic fatigue syndrome: characteristics and possible Cavadini G, Petrzilka S, Kohler P et al (2007) TNF-alpha suppresses
causes for its pathogenesis. Isr Med Assoc J 10:79–82 the expression of clock genes by interfering with E-box-mediated
Bayley-Bucktrout SL, Caulkins SC, Goings G, Fischer JAA, Dzionek transcription. Proc Natl Acad Sci U S A 104:12843–12848
A, Miller SD (2008) Cutting edge: central nervous system plas- Chai LY, Netea MG, Vonk AG, Kullberg BJ (2009) Fungal strategies
macytoid dendritic cells regulate the severity of dendritic cells in for overcoming host innate immune response. Med Mycol
multiple sclerosis. J Immunol 180:6457–6461 47:227–236
Behnsen J, Hartmann A, Schmaler J, Gehrke A, Brakhage AA, Zipfel Chao CC, Janoff EN, Hu SX et al (1991) Altered cytokine release in
PF (2008) The opportunistic human pathogenic fungus Aspergil- PBMC cultures from patients with the chronic fatigue syndrome.
lus fumigatus evades the host complement system. Infect Immun Cytokine 3:292–298
76:820–827 Chaudhuri A, Behan PO (2004) In vivo magnetic resonance spectros-
Benarroch EE (2011) Clinical implications of neuroscience research. copy in chronic fatigue syndrome. Prostaglandins Leukot Essent
Neurology 77:1198–1204 Fatty Acids 71:181–183
Bennett AL, Chao CC, Hu S et al (1997) Elevation of bioactive trans- Chaudhuri A, Condon BR, Gow JW, Brennan D, Hadley DM
forming growth factor-beta in serum of patients with chronic (2003) Proton magnetic resonance spectroscopy of basal Gan-
fatigue syndrome. J Clin Immunol 17:160–166 glia in chronic fatigue syndrome. Brian Imaging Neuro Re-
Berg S, Sappington PL, Guzik LJ, Delude RL, Fink MP (2003) Proin- port 14:225–228
flammatory cytokines increase the rate of glycolysis and Chaumeil MM, Valette J, Guillermier M (2009) Multimodal neuro-
adenosine-5′-triphosphate turnover in cultured rat enterocytes. imaging provides a highly consistent picture of energy metabo-
Crit Care Med 31:1203–1212 lism, validating 31P MRS for measuring brain ATP synthesis.
Berk M, Kapczinski F, Andreazza AC, Dean OM, Giorlando F, Maes Proc Natl Acad Sci U S A 106:3988–3993
M, Yücel M, Gama CS, Dodd S, Dean B, Magalhães PV, Chernyak BV (1997) Redox regulation of the mitochondrial perme-
Amminger P, McGorry P, Malhi GS (2011) Pathways underlying ability transition pore. Biosci Rep 17(3):293–302
neuroprogression in bipolar disorder: focus on inflammation, Csillag A, Boldogh I, Pazmandi K et al (2010) Pollen-induced
oxidative stress and neurotrophic factors. Neurosci Biobehav oxidative stress influences both innate and adaptive immune
Rev 35(3):804–817 responses via altering dendritic cell functions. J Immunol
Biswal B, Kunwar P, Natelson BH (2011) Cerebral blood flow is 184:2377–2385
reduced in chronic fatigue syndrome as assessed by arterial spin Culley DJ, Raghavan SV, Waly M et al (2007) Nitrous oxide
labeling. J Neurol Sci 301:9–11 decreases cortical methionine synthase transiently but produ-
Blatteis CM, Sehic E (1997) Circulating pyrogen signaling of the ces lasting memory impairment in aged rats. Anesth Analg
brain. Ann NY Acad Sci 813:445–447 105:83–88
Bodet D, Glaize G, Dabadie MP, Geffard M (2004) Immunological Danishpajooh IO, Gudi T, Chen Y, Kharitonov VG, Sharma VS, Boss
follow-up for multiple slerosis. Immuno-Analyse & Biol Special- GR (2001) Nitric oxide inhibits methionine synthase activity in
ise 19:138–147 vivo and disrupts carbon flow through the folate pathway. J Biol
Bot A, Smith KA, von Herrath M (2004) Molecular and cellular Chem 276:27296–27303
control of T1/T2 immunity at the interface between antimi- De Becker P, Roeykens J, Reynders M, McGregor N, De Meirleir K
crobial defense and immune pathology. DNA Cell Biol 23 (2000) Exercise capacity in chronic fatigue syndrome. Arch Intern
(6):341–350 Med 160:3270–3277
Metab Brain Dis
de Jager W, Bourcier K, Rijkers GT, Prakken BJ, Seyfert-Margolis V Geffard M, Bodet D, Martinet Y, Dabadie M-P (2002) Detection of the
(2009) Prerequisites for cytokine measurements in clinical trials specific IgM and IgA circulating in sera of multiple sclerosis
with multiplex immunoassays. BMC Immunol 10:52 patients: interest and perspectives. Immuno-Analyse & Biol Spec
Del Rey A, Roggero E, Randolf A et al (2006) IL-1 resets glucose 17:302–310
homeostasis at central levels. PNAS 103:16039–16044 Gerhard A, Pavese N, Hotton G et al (2006) In vivo imaging of
Demitrack MA, Crofford LJ (1998) Evidence for and pathophysiologic microglial activation with [11C](R)-PK11195 PET in idiopathic
implications of hypothalamic-pituitary-adrenal axis dysregulation Parkinson's disease. Neurobiol Dis 21:404–412
in fibromyalgia and chronic fatigue syndrome. Ann N Y Acad Sci Gewurz BE, Vyas JM, Ploegh HL (2007) Herpesvirus evasion of T-cell
840:684–697 immunity. In: Arvin A, Campadelli-Fiume G, Mocarski E et al
Demitrack MA, Dale JK, Straus SE et al (1991) Evidence for impaired (eds) Human herpesviruses: biology, therapy, and immunopro-
activation of the hypothalamic-pituitary-adrenal axis in patients with phylaxis. Cambridge University Press, Cambridge, Chapter 2
chronic fatigue syndrome. J Clin Endocrinol Metab 73:1224–1234 Goehler LE, Relton JK, Dripps D et al (1997) Vagal paraganglia bind
Duke RC (1989) Self recognition by T cells. I. Bystander killing biotinylated interleukin-1 receptor antagonist: a possible mecha-
of target cells bearing syngeneic MHC antigens. J Exp Med nism for immune- to-brain communication. Brain Res Bull
170:59–71 43:357–364
Dykens JA, Will Y, Wiseman RW, Jeneson JAL (2008) Noninvasive Goehler LE, Gaykema RP, Opitz N, Reddaway R, Badr N, Lyte M
assessment of mitochondrial function using nuclear magnetic (2005) Activation in vagal afferents and central autonomic path-
resonance spectroscopy. In: Dykens JA, Will Y (eds) Drug- ways: early responses to intestinal infection with Campylobacter
induced mitochondrial dysfunction. Wiley, Hoboken, Chapter 4. jejuni. Brain Behav Immun 19:334–344
p 55 Goya R, Sun MGF, Morin RD et al (2010) SNVMix: predicting single
Ek M, Kurosawa M, Lundeberg T, Ericsson A (1998) Activation of nucleotide variants from next-generation sequencing of tumors.
vagal afferents after intravenous injection of interleukin-1b: role Bioinformatics 26:730–736
of endogenous prostaglandins. J Neurosci 18:9471–9479 Greco A, Tannock C, Brostoff J, Costa DC (1997) Brain MR in
Engler H, Doenlen R, Engler A et al (2011) Acute amygdaloid Chronic fatigue syndrome. Am J Neuroradiol 18:1265–1269
response to systemic inflammation. Brain Behav Immun Griffiths HR, Moller L, Bartosz G et al (2002) Biomarkers. Mol
25:1384–1392 Aspects Med 23:101–208
Ericsson A, Arias C, Sawchenko PE (1997) Evidence for an in- Groussard C, Morel I, Chevanne M, Monnier M, Cillard J, Delamarche
tramedullary prostaglandin-dependent mechanism in the activa- A (2000) Free radical scavenging and antioxidant effects of lac-
tion of stress-related neuroendocrine circuitry by intravenous tate ion: an in vitro study. J Appl Physiol 89(1):169–175
interleukin-1. J Neurosci 17:7166–7179 Gudz TI, Tserng KY, Hoppel CL (1997) Direct inhibition of mitochon-
Ferrari CC, Tarelli R (2011) Parkinson's disease and systemic inflam- drial respiratory chain complex III by cell-permeable ceramide. J
mation. Parkinsons Dis 2011:436813 Biol Chem 272:24154–24158
Flachenecker P, Bihler I, Weber F, Gottschalk M, Toyka KV, Rieckmann Hahn S, Gehri R, Erb P (1995) Mechanism and biological significance
P (2004) Cytokine mRNA expression in patients with multiple of CD4-mediated cytotoxicity. Immunol Rev 146:57–79
sclerosis and fatigue. Mult Scler J 10:165–169 Haldorsen K, Bjelland I, Bolstad AI, Jonsson R, Brun JG (2011) A
Fletcher MA, Zeng XR, Barnes Z, Levis S, Klimas NG (2009) five-year prospective study of fatigue in primary Sjögren's syn-
Plasma cytokines in women with chronic fatigue syndrome. J drome. Arthritis Res Ther 13(5):R167
Transl Med 7:96 Halliwell B, Gutteridge JMC (1999) Free radicals in biology and
Fletcher MA, Zeng XR, Maher K et al (2010) Biomarkers in chronic medicine. Oxford University Press, New York
fatigue syndrome: evaluation of natural killer cell function and Hautecoeur P, Forzy G, Gallois P, Demirbilek V, Feugas O (1997)
dipeptidyl peptidase IV/CD26. PLoS One 5(5):e10817 Variations of IL2, IL6, TNF alpha plasmatic levels in relapsing
Fransson ME, Liljenfeldt LSE, Fagius J, Tötterman TH, Loskog ASI remitting multiple sclerosis. Acta Neurol Belg 97:240–243
(2009) The T-cell pool is anergized in patients with multiple Heesen C, Nawrath L, Reich C, Bauer N, Schulz K-H, Gold SM (2006)
sclerosis in remission. Immunology 126:92–101 Fatigue in multiple sclerosis: an example of cytokine mediated
Fujinami RS, von Herrath MG, Christen U, Whitton JL (2006) Molec- sickness behaviour? J Neurol Neurosurg Psychiatry 77:34–39
ular mimicry, bystander activation, or viral persistence: infections Herkenham M, Lee HY, Baker RA (1998) Temporal and spatial pat-
and autoimmune disease. Clin Microbiol Rev 19:80–94 terns of c-fos mRNA induced by intravenous interleukin-1: a
Fukuda K, Straus SE, Hickie I, Sharpe M, Dobbins JG, Komaroff AL cascade of non-neuronal cellular activation at the blood brain
(1994) The chronic fatigue syndrome: a comprehensive approach barrier. J Comp Neurol 400:175–196
to its definition and study. Ann Intern Med 121:953–959 Herrod HG (1993) Management of the patient with IgG subclass defi-
Fulton RJ, McDade RL, Smith PL, Kienker LJ, Kettman JR Jr (1997) ciency and/or selective antibody deficiency. Ann Allergy 70:3–8
Advanced multiplexed analysis with the FlowMetrix system. Clin Hilgers A, Frank J (1994) Chronic fatigue syndrome. Immune dys-
Chem 43:1749–1756 function, role of pathogens and toxic agents and neurological and
Fulya I, Mehmet O, Handan A, Vedat B (2006) Cytokine measure- cardial changes. Wien Med Wochenschr 144:399–406
ments in lymphocyte culture supernatant of inactive lepromatous Holley AK, St Clair DK (2009) Watching the watcher: regulation of
leprosy patients. Ind J Med Microbiol 24:121–123 p53 by mitochondria. Future Oncol 5(1):117–130
Gallaher ZR, Ryu V, Herzog T, Ritter RC, Czaja K (2012) Changes in Ingersoll MA, Platt AM, Potteaux S, Randolph GJ (2011) Monocyte
microglial activation within the hindbrain, nodose ganglia, and trafficking in acute and chronic inflammation. Trends Immunol
the spinal cord following subdiaphragmatic vagotomy. Neurosci 32:470–477
Lett 513:31–36 Jadidi-Niaragh F, Mirshafiey A (2011) Th17 cell, the new player of
Gardner A, Boles RG (2008) Symptoms of solarization as a rapid neuroinflammatory process in multiple sclerosis. Scand J Immu-
screening tool for mitochondrial dysfunction in depression. Biop- nol 74:1–13
sychosoc Med 22(2):7 Jammes Y, Steinberg JG, Mambrini O, Bregeon F, Delliaux S (2005)
Garrabou G, Sanjurjo E, Miró O et al (2006) Noninvasive diagnosis of Chronic fatigue syndrome: assessment of increased oxidative
mitochondrial dysfunction in HAART-related hyperlactatemia. stress and altered muscle excitability in response to incremental
Clin Infect Dis 42:584–585 exercise. J Intern Med 257:299–310
Metab Brain Dis
Jung DJ, Jin DH, Hong SW, Kim JE, Shin JS, Kim D, Cho BJ, Hwang Leonard B, Maes M (2012) Mechanistic explanations how cell-
YI, Kang JS, Lee WJ (2010) Foxp3 expression in p53-dependent mediated immune activation, inflammation and oxidative and
DNA damage responses. J Biol Chem 285(11):7995–8002 nitrosative stress pathways and their sequels and concomitants
Kantengwa S, Jornot L, Devenoges C, Nicod LP (2003) Superoxide play a role in the pathophysiology of unipolar depression. Neuro-
anions induce the maturation of human dendritic cells. Am J sci Biobehav Rev 36:764–785
Respir Crit Care Med 167:431–437 Liang H, Xu L, Zhou C, Zhang Y, Xu M, Zhang C (2011) Vagal
Karchin R (2009) Next generation tools for the annotation of human activities are involved in antigen-specific immune inflammation
SNPs. Brief Bioinform 10:35–52 in the intestine. J Gastroenterol Hepatol 26:1065–1071
Kennedy G, Spence V, McLaren M, Hill S, Belch J (2003) Increased Lin MT, Beal MF (2006) Mitochondrial dysfunction and oxidative
plasma isoprostanes and other markers of oxidative stress in stress in neurodegenerative diseases. Nature 443:787–795
chronic fatigue syndrome. J Thromb Haemost 1 (Suppl): P0182 Linthorst AC, Flachskamm C, Müller-Preuss P, Holsboer F, Reul JM
Kennedy G, Spence VA, McLaren M, Hill A, Underwood C, Belch JJ (1995) Effect of bacterial endotoxin and interleukin-1 beta on
(2005) Oxidative stress levels are raised in chronic fatigue syn- hippocampal serotonergic neurotransmission, behavioral activity,
drome and are associated with clinical symptoms. Free Radic Biol and free corticosterone levels: an in vivo microdialysis study. J
Med 39:584–589 Neurosci 15:2920–2934
Kerkeni M, Addad F, Chauffert M et al (2006) Hyperhomocysteine- Longhini AL, von Glehn F, Brandão CO et al (2011) Plasmacy-
mia, endothelial nitric oxide synthase polymorphism, and risk of toid dendritic cells are increased in cerebrospinal fluid of
coronary artery disease. Clin Chem 52:53–58 untreated patients during multiple sclerosis relapse. J Neuro-
Khalaf H, Jass J, Olsson PE (2010) Differential cytokine regulation by inflammation 8:2
NF-kappaB and AP-1 in Jurkat T-cells. BMC Immunol 11:26 Maes M (2009) Inflammatory and oxidative and nitrosative stress
Kiely A, McClenaghan NH, Flatt PR, Newsholme P (2007) Pro- pathways underpinning chronic fatigue, solarization and psycho-
inflammatory cytokines increase glucose, alanine and triacylgly- somatic symptoms. Curr Opin Psychiatry 22:75–83
cerol utilization but inhibit insulin secretion in a clonal pancreatic Maes M (2011a) An intriguing and hitherto unexplained co-
ß-cell line. J Endocrinol 195:113–123 occurrence: depression and chronic fatigue syndrome are mani-
Kim SU, de Vellis J (2005) Microglia in health and disease. J Neurosci festations of shared inflammatory, oxidative and nitrosative
Res 81:302–313 (IO&NS) pathways. Prog Neuropsychopharmacol Biol Psychiatry
Klein R, Berg PA (1995) High incidence of antibodies to 5- 35:784–794
hydroxytryptamine, gangliosides and phospholipids in patients with Maes M (2011b) Nooit meer moe: CVS ontmaskerd. Uitgever: Zorro
chronic fatigue and fibromyalgia syndrome and their relatives: evi- Uitgeverij. Brugge. ISBN: 9461680015/9789461680013
dence for a clinical entity of both disorders. Eur J Med Res 1:21–26 Maes M, Twisk FN (2009) Why myalgic encephalomyelitis/chronic
Klimas NG, Salvato FR, Morgan R, Fletcher MA (1990) Immunologic fatigue syndrome (ME/CFS) may kill you: disorders in the in-
abnormalities in chronic fatigue syndrome. J Clin Microbiol flammatory and oxidative and nitrosative stress (IO&NS) path-
28:1403–1410 ways may explain cardiovascular disorders in ME/CFS. Neuro
Komaroff AL, Cho TA (2011) Role of infection and neurologic dys- Endocrinol Lett 30:677–693
function in chronic fatigue syndrome. Semin Neurol 31:325–337 Maes M, Twisk FN (2010) Chronic fatigue syndrome: Harvey and
Korn T, Bettelli E, Oukka M, Kuchroo VK (2009) IL-17 and Th17 Wessely's (bio)psychosocial model versus a bio(psychosocial)
Cells. Annu Rev Immunol 27:485–517 model based on inflammatory and oxidative and nitrosative stress
Kortekaas TR, Leenders KL, van Oostrom JCH et al (2005) Blood- pathways. BMC Med 8:35
brain barrier dysfunction in Parkinsonian midbrain in vivo. Ann Maes M, Mihaylova I, De Ruyter M (2005a) Decreased dehydroepian-
Neurology 57:176–179 drosterone sulfate but normal insulin-like growth factor in chronic
Krueger JM, Majade JA (1987) Sleep and the immune response. Ann fatigue syndrome (CFS): relevance for the inflammatory response
NY Acad Sci 496:510–516 in CFS. Neuro Endocrinol Lett 26(5):487–492
Kunert A, Losse J, Gruszin C et al (2007) Immune evasion of the Maes M, Mihaylova I, Leunis JC (2005b) In chronic fatigue syndrome,
human pathogen Pseudomonas aeruginosa: elongation factor Tuf the decreased levels of omega-3 poly-unsaturated fatty acids are
is a factor H and plasminogen binding protein. J Immunol related to lowered serum zinc and defects in T cell activation.
179:2979–2988 Neuro Endocrinol Lett 26:745–751
Lal G, Yin N, Xu J et al (2011) Distinct inflammatory signals have Maes M, Mihaylova I, De Ruyter M (2006a) Lower serum zinc in
physiologically divergent effects on epigenetic regulation of Foxp3 Chronic Fatigue Syndrome (CFS): relationships to immune dys-
expression and Treg function. Am J Transplant 11:203–214 functions and relevance for the oxidative stress status in CFS. J
Landay AL, Jessop C, Lennette ET, Levy JA (1991) Chronic fatigue Affect Disord 90:141–147
syndrome: clinical condition associated with immune activation. Maes M, Mihaylova I, Leunis JC (2006b) Chronic fatigue syndrome is
Lancet 338:707–712 accompanied by an IgM-related immune response directed against
Lane RJM, Barrett MC, Woodrow D et al (1998) Muscle fibre charac- neopitopes formed by oxidative or nitrosative damage to lipids
teristics and lactate responses to exercise in chronic fatigue syn- and proteins. Neuro Endocrinol Lett 27:615–621
drome. J Neurol Neurosurg Psychiatry 64:362–367 Maes M, Mihaylova I, Bosmans E (2007a) Not in the mind of neuras-
Lange G, DeLuca J, Maldjian JA, Lee H, Tiersky LA, Natelson BH thenic lazybones but in the cell nucleus: patients with chronic
(1999) Brain MRI abnormalities exist in a subset of patients with fatigue syndrome have increased production of nuclear factor
chronic fatigue syndrome. J Neurol Sci 171:3–7 kappa beta. Neuro Endocrinol Lett 28:456–462
Larsson HB, Tofts PS (1992) Measurement of blood-brain barrier Maes M, Mihaylova I, Kubera M, Bosmans E (2007b) Not in the mind
permeability using dynamic Gd-DTPA scanning–a comparison but in the cell: increased production of cyclo-oxygenase-2 and
of methods. Magn Reson Med 24:174–176 inducible NO synthase in chronic fatigue syndrome. Neuro Endo-
Layser RB (1978) Myeloneuropathy after prolonged exposure to ni- crinol Lett 28:463–469
trous oxide. Lancet 312:1227–1230 Maes M, Mihaylova I, Kubera M, Uytterhoeven M, Vrydags N, Bosmans
Le Grand D, Solsona M, Rosengarten R, Poumarat F (1996) Adaptive E (2009a) Coenzyme Q10 deficiency in myalgic encephalomyelitis/
surface antigen variation in Mycoplasma bovis to the host im- chronic fatigue syndrome (ME/CFS) is related to fatigue, autonomic
mune response. FEMS Microbiol Lett 144:267–275 and neurocognitive symptoms and is another risk factor explaining
Metab Brain Dis
the early mortality in ME/CFS due to cardiovascular disorder. Neuro Moss RB, Mercandetti A, Vojdani A (1999) TNF-alpha and chronic
Endocrinol Lett 30:462–469 fatigue syndrome. J Clin Immunol 9:314–316
Maes M, Mihaylova I, Kubera M, Uytterhoeven M, Vrydags N, Bosmans Mukherjee A, Morosky SA, Delorme-Axford E et al (2011) The
E (2009b) Increased 8- hydroxy-deoxyguanosine, a marker of oxi- Coxsackievirus B 3Cpro protease cleaves MAVS and TRIF to
dative damage to DNA, in major depression and myalgic encepha- attenuate host Type I interferon and apoptotic Signaling. PLoS
lomyelitis/chronic fatigue syndrome. Neuro Endocrinol Lett Pathog 7(3):e1001311
30:715–722 Murrough JW, Mao X, Collins KA et al (2010) Increased ventricular
Maes M, Ruckoanich P, Chang YS, Mahanonda N, Berk M (2011) lactate in chronic fatigue syndrome measured by 1H MRS imag-
Multiple aberrations in shared inflammatory and oxidative & ing at 3.0 T. II: comparison with major depressive disorder. NMR
nitrosative stress (IO&NS) pathways explain the co-association Biomed 23:643–650
of depression and cardiovascular disorder (CVD), and the in- Mus AM, Cornelissen F, Asmawidjaja PS et al (2010) Interleukin-
creased risk for CVD and due mortality in depressed patients. 23 promotes Th17 differentiation by inhibiting T-bet and
Prog Neuropsychopharmacol Biol Psychiatry 35:769–783 FoxP3 and is required for elevation of interleukin-22, but
Maes M, Twisk FNM, Johnson C (2012a) Myalgic Encephalomyelitis not interleukin-21, in autoimmune experimental arthritis. Ar-
(ME), Chronic Fatigue Syndrome (CFS), and Chronic Fatigue thritis Rheum 62:1043–1050
(CF) are distinguished accurately: results of supervised learning Myhill S, Booth NE, McLaren-Howard J (2009) Chronic fatigue syn-
techniques applied on clinical and inflammatory data. Psychiatr drome and mitochondrial dysfunction. Int J Clin Exp Med 2:1–16
Res Apr 20. doi:10.1016/j.psychres.2012.03.031 Naess H, Sundal E, Myhr KM, Nyland HI (2010) Postinfectious and
Maes M, Twisk FN, Kubera M, Ringel K (2012b) Evidence for chronic fatigue syndromes: clinical experience from a tertiary-
inflammation and activation of cell-mediated immunity in Myal- referral centre in Norway. In Vivo 24:185–188
gic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): In- Nakamura T, Schwander SK, Donnelly R, Ortega F, Togo F, Broderick
creased interleukin-1, tumor necrosis factor-4a, PMN-elastase, G, Yamamoto Y, Cherniack NS, Rapoport D, Natelson BH (2010)
lysozyme and neopterin. J Affect Disord 136:933–939 Cytokines across the night in chronic fatigue syndrome with and
Maes M, Twisk FN, Kubera M, Ringel K, Leunis JC, Geffard M without fibromyalgia. Clin Vaccine Immunol 17(4):582–587
(2012c) Increased IgA responses to the LPS of commensal bac- Newton JL, Okonkwo O, Sutcliffe K, Seth A, Shin J, Jones DE (2007)
teria is associated with inflammation and activation of cell- Symptoms of autonomic dysfunction in chronic fatigue syn-
mediated immunity in chronic fatigue syndrome. J Affect Disord drome. QJM 100:519–526
136:909–917 Nijs J, Meeus M, McGregor NR et al (2005) Chronic fatigue syn-
Maher CO, Anderson RE, Martin HS, McClelland RL, Meyer FB drome: exercise performance related to immune dysfunction. Med
(2003) Interleukin-1beta and adverse effects on cerebral blood Sci Sports Exerc 37:1647–1654
flow during long-term global hypoperfusion. J Neurosurg Nishikai M, Tomomatsu S, Hankins RW et al (2001) Autoantibodies to
99:907–912 a 68/48 kDa protein in chronic fatigue syndrome and primary
Manuel-y-Keenoy B, Moorkens G, Vertommen J, Noe M, Neve J, De fibromyalgia: a possible marker for hypersomnia and cognitive
Leeuw I (2000) Magnesium status and parameters of the oxidant- disorders. Rheumatology 40:806–810
antioxidant balance in patients with chronic fatigue: effects of Niu G, Wright KL, Ma Y et al (2005) Role of Stat3 in regulating p53
supplementation with magnesium. J Amer Coll Nutrition expression and function. Mol Cell Biol 25(17):7432–7440
19:374–382 Obal F Jr, Krueger JM (2003) Biochemical regulation of non-rapid-
Manuel-y-Keenoy B, Moorkens G, Vertommen J, De Leeuw I (2001) eye-movement sleep. Front Biosci 8:d520–550
Antioxidant status and lipoprotein peroxidation in chronic fatigue Ogawa M, Nishiura T, Yoshimura M et al (1998) Decreased nitric
syndrome. Life Sci 68:2037–2049 oxide-mediated natural killer cell activation in chronic fatigue
Martín P, Sánchez-Madrid F (2011) CD69: an unexpected regulator of syndrome. Eur J Clin Invest 28:937–943
TH17 cell-driven inflammatory responses. Sci Signal 4(165):pe14 Ortega-Hernandez OD, Shoenfeld Y (2009) Infection, vaccination, and
Mathew SJ, Mao X, Keegan KA et al (2009) Ventricular cerebrospinal autoantibodies in chronic fatigue syndrome, cause or coinci-
fluid lactate is increased in chronic fatigue syndrome compared dence? Ann NY Acad Sci 1173:600–609
with generalized anxiety disorder: an in vivo 3.0 T (1)H MRS Paradies G, Ruggiero FM, Petrosillo G, Quagliariello E (1998) Perox-
imaging study. NMR Biomed 22:251–258 idative damage to cardiac mitochondria: cytochrome oxidase and
Matoba S, Kang JG, Patino WD et al (2006) p53 regulates mitochon- cardiolipin alterations. FEBS Lett 424:155–158
drial respiration. Science 312:1650–1653 Patrick NJ, Roberts AD, Leavins N, Harrison MF, Croll JC, Sexsmith
Meeus M, Mistiaen W, Lambrecht L, Nijs J (2009) Immunological JR (2008) Prefrontal cortex oxygenation during incremental ex-
similarities between cancer and chronic fatigue syndrome: the ercise in chronic fatigue syndrome. Clin Physiol Funct Imaging
common link to fatigue? Anticancer Res 29:4717–4726 28:364–372
Meulemans A, Gerlo E, Seneca S et al (2007) The aerobic forearm Perrin R, Embleton K, Pentreath VW, Jackson A (2010) Longitudinal
exercise test, a non-invasive tool to screen for mitochondrial MRI shows no cerebral abnormality in chronic fatigue syndrome.
disorders. Acta Neurol Belg 107:78–83 Br J Radiol 83:419–423
Mihaylova I, DeRuyter M, Rummens JL, Bosmans E, Maes M (2007) Perry VH, Newman TA, Cunningham C (2003) The impact of systemic
Decreased expression of CD69 in chronic fatigue syndrome in infection on the progression of neurodegenerative disease. Nat
relation to inflammatory markers: evidence for a severe disorder Rev Neurosci 4:103–112
in the early activation of T lymphocytes and natural killer cells. Perry VH, Cunningham C, Holmes C (2007) Systemic infections and
Neuro Endocrinol Lett 28(4):477–483 inflammation affect chronic neurodegeneration. Nature Reviews
Mohankumar PS, Thyagarajan S, Quadri SK (1991) Interleukin-1 Immunol 7:161–167
stimulates the release of dopamine and dihydroxyphenylacetic Perry VH, Nicoll JAR, Holmes C (2010) Microglia in neurodegener-
acid from the hypothalamus in vivo. Life Sci 48:925–930 ative disease. Nat Rev Neurol 6:193–201
Morgan D, Oliveira-Emilio HR, Keane D et al (2007) Glucose, palmi- Phelps DT, Ferro TJ, Higgins PJ, Shankar R, Parker DM, Johnson A
tate and pro-inflammatory cytokines modulate production and (1995) TNF-alpha induces peroxynitrite-mediated depletion of
activity of a phagocyte-like NADPH oxidase in rat pancreatic lung endothelial glutathione via protein kinase C. Am J Physiol
islets and a clonal beta cell line. Diabetologia 50:359–369 269(4 Pt 1):L551–559
Metab Brain Dis
Pincus S (2005) Potential role of infections in chronic inflammatory cytokines in hippocampal neuronal cell layers. J Neurosci
diseases. ASM News 71:529–535 26:10709–10716
Pokryszko-Dragan A, Frydecka I, Kosmaczewska A, Ciszak L, Bilińnska Spence VA, Kennedy G, Belch JJ, Hill A, Khan F (2008) Low-grade
M, Gruszka E, Podemski R, Frydecka D (2012) Stimulated periph- inflammation and arterial wave reflection in patients with chronic
eral production of interferon-gamma is related to fatigue and depres- fatigue syndrome. Clin Sci (Lond) 114(8):561–566
sion in multiple sclerosis. Clin Neurol Neurosurg. doi:10.1016/j. Stephensen CB, Marquis GS, Douglas SD, Wilson CM (2005) Immune
clineuro.2012.02.048 activation and oxidative damage in HIV-positive and HIV-
Puri BK, Jakeman PM, Agour M et al (2011) Regional grey and white negative adolescents. J Acquir Immune Defic Syndr 38:180–190
matter volumetric changes in myalgic encephalomyelitis (chronic Sternberg EM (2006) Neural regulation of innate immunity: a coordi-
fatigue syndrome): a voxel-based morphometry 3-T MRI study. nated nonspecific host response to pathogens. Nat Rev Immunol
Br J Radiol [Pub. ahead of print] doi:10.1259/bjr/93889091 6:318–328
Qin L, Wu X, Block ML et al (2007) Systemic LPS causes chronic Strickland PS, Levine PH, Peterson DL, O'Brien K, Fears T (2001)
neuroinflammation and progressive neurodegeneration. Glia Neuromyasthenia and chronic fatigue syndrome (CFS) in North-
55:453–462 ern Nevada/California: a ten-year follow-up of an outbreak. J
Radolf JD (1994) Role of outer membrane architecture in immune Chron Fatigue Syndr 9:3–14
evasion by Treponema pallidum and Borrelia burgdorferi. Trends Tirelli U, Marotta G, Improta S, Pinto A (1994) Immunological abnor-
Microbiol 2:307–311 malities in patients with chronic fatigue syndrome. Scand J Immu-
Ransohoff RM, Perry VH (2009) Microglial physiology: unique stim- nol 40:601–608
uli, specialized responses. Ann Rev Immunol 27:119–145 Tracey KJ (2002) The inflammatory reflex. Nature 420:853–859
Riazi K, Galic MA, Kuzmiski JB, Ho W, Sharkey KA, Pittman QJ Trushina E, McMurray CT (2007) Oxidative stress and mitochon-
(2008) Microglial activation and TNFalpha production mediate drial dysfunction in neurodegenerative diseases. Neuroscience
altered CNS excitability following peripheral inflammation. Proc 145:1233–1248
Natl Acad Sci USA 105:17151–17156 Twisk FN, Maes M (2009) A review on cognitive behavorial therapy
Rönnbäck L, Elisabeth Hansson E (2004) On the potential role of (CBT) and graded exercise therapy (GET) in myalgic encephalo-
glutamate transport in mental fatigue. J Neuroinflamm 1:22 myelitis (ME)/chronic fatigue syndrome (CFS): CBT/GET is not
Samanta A, Li B, Song X et al (2008) TGF-beta and IL-6 signals only ineffective and not evidence-based, but also potentially
modulate chromatin binding and promoter occupancy by acety- harmful for many patients with ME/CFS. Neuro Endocrinol Lett
lated FOXP3. PNAS 105:14023–14027 30:284–299
Samavati L, Lee I, Mathes I, Lottspeich F, Hüttemann M (2008) Tumor Valdes-Ferrer S, Rosas-Ballina M, Olofsson P, Chavan S, Tracey K
necrosis factor alpha inhibits oxidative phosphorylation through (2010) Vagus nerve stimulation produces an anti-inflammatory
tyrosine phosphorylation at subunit I of cytochrome c oxidase. J monocyte phenotype in blood. J Immunol 184:138
Biol Chem 283:21134–21144 Van Den Eede F, Moorkens G et al (2007) Hypothalamic-pituitary-
Saper CB (1995) Central autonomic system. In: Paxinos G (ed) The rat adrenal axis function in chronic fatigue syndrome. Neuropsycho-
nervous system, 2nd edn. Academic, San Diego, pp 107–135 biol 55:112–120
Schutzer SE, Angel TE, Liu T et al (2011) Distinct Cerebrospinal fluid Van Oosterwijck J, Nijs J, Meeus M et al (2010) Pain inhibition
proteomes differentiate post-treatment Lyme disease from chronic and postexertional malaise in myalgic encephalomyelitis/
fatigue syndrome. PLoS One 6(2):e17287 chronic fatigue syndrome: an experimental study. J Intern
Schwartz RB, Komaroff AL, Garada BM et al (1994) SPECT imaging Med 268:265–278
of the brain: comparison of findings in patients with chronic VanNess JM, Stevens SR, Bateman L et al (2010) Postexertional
fatigue syndrome, AIDS dementia complex, and major unipolar Malaise in women with chronic fatigue syndrome. J Womens
depression. Am J Roentgenol 162:943–951 Health (Larchmt) 19:239–244
Schwid SR, Covington M, Segal BM, Goodman AD (2002) Fatigue in Vermeulen RCW, Kurk RM, Visser FC, Sluiter W, Scholte HR (2010)
multiple sclerosis: current understanding and future directions. J Patients with chronic fatigue syndrome performed worse than
Rehabil Res Dev 39:211–224 controls in a controlled repeated exercise study despite a normal
Scott LV, Medbak S, Dinan TG (1998) Blunted adrenocorticotropin oxidative phosphorylation capacity. J Transl Med 8:93
and cortisol responses to corticotropin-releasing hormone stimu- Vojdani A, Lapp CW (1999) Interferon-induced proteins are elevated
lation in chronic fatigue syndrome. Acta Psychiatr Scand 97 in blood samples of patients with chemically or virally induced
(6):450–457 chronic fatigue syndrome. Immunopharmacol Immunotoxicol
Shintani F, Kanba S, Nakaki T et al (1993) Interleukin-1 beta augments 21:175–202
release of norepinephrine, dopamine, and serotonin in the rat Wang HH, Dai YQ, Qiu W et al (2011) Interleukin-17-secreting T cells
anterior hypothalamus. J Neurosci 13:3574–3581 in neuromyelitis optica and multiple sclerosis during relapse. J
Shungu DC, Weiduschat N, Murrough JW et al (2012) Increased Clin Neurosci 18:1313–1317
ventricular lactate in chronic fatigue syndrome. III. Relationships Weismann D, Binder CJ (2012) The innate immune response to prod-
to cortical glutathione and clinical symptoms implicate oxidative ucts of phospholipid peroxidation. Biochim Biophys Acta.
stress in disorder pathophysiology. NMR Biomed. doi:10.1002/ doi:10.1016/j.bbamem.2012.01.018
nbm.2772 Wessels E, Duijsings D, Lanke KHW et al (2006) Effects of picorna-
Simmons WL, Dybvig K (2007) How some mycoplasmas evade host virus 3A proteins on protein transport and GBF1-dependent COP-
immune responses. Microbe 2:537–543 I recruitment. J Virol 80:11852–11860
Simopoulos A (2002) Omega-3 fatty acids in inflammation and auto- Whitton PS (2007) Inflammation as a causative factor in the aetiology
immune diseases. J Am Coll Nutr 21:495–505 of Parkinson's disease. Br J Pharmacol 150:963–976
Smyth MJ, Sedgwick JD (1998) Delayed kinetics of tumor necrosis Winkler AS, Blair D, Marsden JT, Peters TJ, Wessely S, Cleare AJ
factor-mediated bystander lysis by peptide-specific CD8+ cyto- (2004) Autonomic function and serum erythropoietin levels in
toxic T lymphocytes. Eur J Immunol 28:4162–4169 chronic fatigue syndrome. J Psychosom Res 56:179–183
Sparkman NL, Buchanan JB, Heyen JR, Chen J, Beverly JL, Johnson RW World Health Organization (1992) International statistical classifica-
(2006) Interleukin-6 facilitates lipopolysaccharide-induced disrup- tion of diseases and related health problems, Tenth Revision.
tion in working memory and expression of other proinflammatory Volume 1, Geneva, WHO. G93.3
Metab Brain Dis
Xiang Y, Xu G, Weigel-Van Aken KA (2010) Lactic Acid induces Zhang L, Gough J, Christmas D et al (2010) Microbial infections in
aberrant amyloid precursor protein processing by promoting its eight genomic subtypes of chronic fatigue syndrome/myalgic
interaction with endoplasmic reticulum chaperone proteins. PLoS encephalomyelitis. J Clin Pathol 63:156–164
One 3;5(11):e13820 Zhong G (2009) Killing me softly: chlamydial use of proteolysis for
Yamamoto S, Ouchi Y, Onoe H et al (2004) Reduction of serotonin evading host defenses. Trends Microbiol 17:467–474
transporters of patients with chronic fatigue syndrome. Neuro- Zhu J, Yamane H, Paul WE (2010) Differentiation of effector CD4 T
report 15:2571–2574 cell populations (*). Ann Rev Immunol 28:445–89