Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

J Neurol (2011) 258:533–548

DOI 10.1007/s00415-010-5836-5

REVIEW

The role of cytokines in Guillain–Barré syndrome


Ming-Ou Lu • Jie Zhu

Received: 7 June 2010 / Revised: 13 October 2010 / Accepted: 9 November 2010 / Published online: 23 November 2010
Ó Springer-Verlag 2010

Abstract Cytokines play an important role in the patho- 1.2–2.3 per 100,000 throughout the world [1]. It is a
genesis of autoimmune diseases including Guillain–Barré clinical syndrome whose pathological substrate may be
syndrome (GBS) and its animal model experimental auto- acute inflammatory demyelinating polyradiculoneuropathy
immune neuritis (EAN). In this article, we reviewed the (AIDP) or acute axonal motor (AMAN) or motor and
current knowledge of the role of cytokines such as TNF-a, sensory axonal neuropathy (AMSAN) [2], acute sensory
IFN-c, IL-1b, IL-6, IL-12, IL-18, IL-23, IL-17, IL-10, IL-4 neuronopathy, acute pandysautonomia and the Fisher syn-
and chemokines in GBS and EAN as unraveled by studies drome [3]. In two-thirds of GBS cases, the disease is
both in the clinic and the laboratory. However, these regarded as a post-infectious autoimmune disease that may
studies occasionally yield conflicting results, highlighting involve molecular mimicry triggered by many antecedent
the complex role that cytokines play in the disease process. pathogens [4], [5]. Both cellular and humoral immunity
Efforts to modulate cytokine function in GBS and other may participate. The different patterns of GBS are probably
autoimmune disease have shown efficiency indicating that due to the diverse interplay between antibodies and T cells
cytokines are important therapeutic targets. of differing specificities [6]. Experimental evidence
implicated that axonal subgroups of GBS and Fisher syn-
Keywords Cytokine  Guillain–Barré syndrome  drome are more related to auto-antibodies against gangli-
Experimental autoimmune neuritis  Autoimmunity  T cell oside, while AIDP more involves CD4? T cell induced
macrophage associated demyelination, although some
studies showed the importance of humoral immunity
Introduction especially in early stages of AIDP [1, 5, 7, 8]. Due to the
remarkable similarities in clinic and pathology, experi-
Guillain–Barré syndrome (GBS) is a common cause of mental autoimmune neuritis (EAN) is considered to be an
acute neuromuscular paralysis with an annual incidence of animal model of AIDP, the most common form of GBS [9].
Current therapies for GBS such as plasma exchange
(PE) and intravenous immunoglobulin (IVIg) mainly focus
M.-O. Lu  J. Zhu
on the humoral immunity. Although results of international
Department of Neurology, The First Hospital, Jilin University,
Xinmin Street No.17, Changchun, China randomized trials have shown equivalent efficacy of PE
and IVIg, but not corticosteroids, in hastening recovery
M.-O. Lu from GBS, IVIg is usually used because of its greater
Department of Neurology, The First Hospital, Dalian Medical
convenience and availability [1, 10, 11]. However, none of
University, Zhongshan Road 222, Dalian, China
the treatments significantly reduced mortality. Since
J. Zhu (&) *20% of patients die or have persistent disability despite
Division of Neurodegeneration (NOVUM, plan 5), immunotherapy, more studies are needed to identify better
Department of Neurobiology, Care Sciences and Society,
treatment regimens and new therapeutic strategies [8].
Karolinska Institute, Karolinska University Hospital
in Huddinge, 141 86 Stockholm, Sweden Cytokines are polypeptides that act as signal molecules
e-mail: jie.zhu@ki.se in a paracrine or autocrine fashion between the cells of the

123
534 J Neurol (2011) 258:533–548

immune system and also in other area including psycho- disease by playing an important anti-inflammatory role
neuroimmunological interactions. Cytokines play a key [17–19]. More recently, IL-17 producing Th17 cells has
role in initiating, propagating and regulating tissue-specific been found playing an important role in many autoimmune
autoimmune injury. The analysis of cytokine production is diseases including experimental autoimmune encephalo-
increasingly important in defining the mechanism of myelitis (EAE), an analogous disease of the central nervous
autoimmune responses and in evaluating specific therapies system (CNS), while thymic regulatory T (Treg) cells have
of autoimmune diseases. This review will focus on the role a suppressive role, although their roles need to be further
of cytokines in GBS and EAN in recent years, to better investigated in EAN and GBS [20, 21]. Actually, increased
understand the mechanism and treatment of GBS. levels of activated T cells and reduced levels of CD4/
CD25? cells (Treg cells) were found in peripheral blood of
acute stage of GBS, but marked improvement of Tregs in
Cytokine in GBS and EAN stable-stage of GBS after IVIg treatment [22, 23]. Single
cytokine in EAN and GBS would be discussed as below.
Cellar immunity is likely involved in the pathogenesis of
the GBS and some forms of EAN. The inflammatory cells IFN-c
infiltrating in the peripheral nervous system (PNS) of both
diseases are composed of lymphocytes and cells of the IFN-c is mainly produced by T cells and natural killer (NK)
macrophage lineage, exerting most of their effects through cells [24]. It is typically produced by Th1 cells and is the
immuno-regulatory cytokines [12] (Fig. 1). Pro-inflamma- principal marker of a Th1 response. The effects of IFN-c
tory cytokines such as IL-1b, IL-6, IL-12, IL-17, IL-18, are induced and regulated by IL-12, and also suppressed
IL-23, TNF-a, IFN-c and chemokines are thought to sig- markedly by apoptotic cells as evidenced in stimulated
nificantly contribute to disease development by recruiting human peripheral mononuclear cells [25]. Its functions
effecter cells to the PNS and by enabling in situ release of include activation of macrophages, differentiation of T
other products toxic for Schwann cells (SCs) and myelin cells to Th1 phenotype, induction of MHC I and MHC II
such as free radicals, oxygen intermediates and nitric oxide expression, B cell class switching, apoptosis of T cells and
(NO) [12–16]. Anti-inflammatory cytokines from Th2 and other cell types, enhancement of other cytokine production
Th3 cells such as IL-4, IL-10 and TGF-b may suppress the such as TNF-a, IL-1b, IL-6, and receptor activator of

Fig. 1 The role of cytokines in Cell migration


EAN and GBS

BNB breakdown

Increase BNB permeability


APC Th1
Adhesion molecules,
IL-1, 6,TNF-α, MCP-
Th2 1,MIP-1a, β, IP-10,
B cell RANTES,CCR2

IL-18
TGF-β (-)

IL-6,
TGF-β
TNF-α

Plasma cell
(-)

Th3

IL-12, sIL-6R

enhancement probably (-) inhibition

123
J Neurol (2011) 258:533–548 535

nuclear factor-kB (NF-kB) ligand, as well as up-regulation cells in GBS [50]. Thus, IFN-c might have a dual role in
of metalloproteinases, chemokines and adhesion mole- this disease and the protective role needs to be further
cules. IFN-c also up-regulates cytokine inducible NO investigated.
synthase (iNOS)-specific mRNA in SCs and precipitates
release of nitrite in a dose-dependent manner [26]. More- TNF-a
over, IFN-c promotes SCs to present exogenous P2 protein
to P2-specific neurogenic T cell line cells [27]. TNF-a is produced by many cells including macrophage, T
In EAN, the level of IFN-c producing cells in blood cells, SCs and so on [51]. Membrane-bound or soluble
lymph node and PNS tissue roughly parallel clinical TNF-a signals through TNF-a receptor 1 (TNFR1, p55)
symptoms. Systemic administration of recombinant rat and TNF-a receptor 2 (TNFR2, p75), mainly TNFR1, to
IFN-c (rrIFN-c) worsens EAN rats and IFN-c receptor elicit a wide variety of signals in many different cell types
deficient mice showed a suppression of EAN [28]. In GBS [52, 53].
patients, serum IFN-c was found elevated [29]. Although a In immune response, TNF-a induced matrix metallo-
significantly increased antigen specific peripheral blood proteinase (MMP-9) promotes macrophage recruitment
mononuclear cells (PBMC) proliferation was not shown, into injured peripheral nerve [54]. Induction of TNF-a by
PBMC secreting IFN-c spontaneously elevated in 25% of lipopolysaccharide (LPS) in SC is regulated by mitogen-
patients of GBS, which indicated a role of T cells in the activated protein kinase (MAPK) signals [55] and inhibi-
immunopathology of GBS [30]. Moreover, neutralizing tion of TNF-a-initiated p38MAPK pathway enhances
auto-antibodies to IFN-c are significantly correlated with axonal regeneration [56]. This indicates that MAPK might
down-regulation of IFN-c producing cells and with be an important pathway that leads TNF production and its
improved clinical disability in GBS patients [31]. action on the peripheral nerve. Moreover, it is also revealed
However, IFN-c has also been evidenced to have a that TNF-a inhabits SC proliferation and mediates SC
protective role in autoimmunity since some other autoim- death by up-regulating Src-suppressed protein kinase C
mune disease aggravated either in the anti-IFN-c treatment substrate and p75NTR expression separately [57, 58]. More
or gene knock out animal models such as EAE, collagen recently, TNFR1 is found as a positive T cell costimulatory
induced arthritis (CIA) and experimental autoimmune molecule that is important for T cell activation and effector
uveitis (EAU) [32–34]. Exogenous IFN-c prevents auto- cytokine production and the development of primary
immune diabetes both in BB rats and NOD mice [35–37]. immune responses in mice [59].
Also in patients with rheumatoid arthritis (RA), various TNF-a has a pivotal role in the pathogenesis of EAN and
studies described protective effects for the use of recom- GBS. In GBS, serum TNF-a, correlated to TNF-a two
binant IFN-c [38, 39], although one report established allele, was elevated and associated with the disease
beneficial effects for the antibody treatment [40]. severity [60, 61]. TNF-a converting enzyme (TACE),
The main protective roles of IFN-c in recent findings processing of membrane-bound inactive pro-TNF-a into
included as below: inhibit on neutrophil trafficking [41, the active soluble cytokine TNF-a, is up-regulated during
42]; convert CD4?CD25- cells into CD4? Treg cells EAN and expressed in nerves of GBS patients and thus
[43]; suppress the differentiation and effector function of may contribute to the pathogenesis of inflammatory
the pro-inflammatory Th17 cells [44, 45]; microglia acti- demyelination of the PNS [62]. Moreover, TNF-a and
vated by IFN-c or IL-4 can protect neutrons and support TNFR1 decreased while sTNFR1, an antagonist of TNF-a,
both neurogenesis and oligodendrogenesis [46–48]. In increased correlated to the neurological recovery in GBS
2008, Foulds et al. [49] reported IFN-c plays a critical role patients treated with IVIg [60, 63, 64]. This might be due to
for controlling all stages of Th1 differentiation and mem- the down-regulation of TNF-a production and sustained
ory formation. At first, IFN-c controls T cell expansion by buffering of TNF-a by sTNF-Rs, particularly sTNFR1, and
direct caspase induction and causing apoptosis. Later, IVIg could selectively induce this gene transcription and
IFN-c mediates the death of Th1 cells indirectly through secretion of anti-inflammatory cytokines as evidenced in
induction of numerous factors, such as NO, Indoleamine cultured human lymphocyte [65].
2,3-dixoygenase (IDO), and TNF-a receptors. Therefore, In EAN, TNFR1 deficiency reduces antigen-presenting
Kelchtermans [41] suggests that endogenous IFN-c can be capacity of SCs and ameliorates EAN in mice [66]. Pro-
considered as a process with bidirectional immune-regu- phylaxis with sTNFR1 treatment delayed the onset, which
lation and often resulting in moderation of pathology. was accompanied by inhibition of blood-nerve-barrier
To date, most researches indicate that IFN-c plays a (BNB) permeability and inflammation, however, after EAN
crucial inflammatory role in EAN and GBS while recent established, sTNFR1 cannot affect the course of the dis-
study reveals that IFN-c can convert peripheral ease, indicating that TNF plays a role in BNB permeability
CD4(?)CD25(-) T cells to CD4(?)CD25(?) regulatory T [67]. Through activation of caspase-3, TNF-a seemed

123
536 J Neurol (2011) 258:533–548

implicated in SC apoptosis in the natural course of EAN and TNFR2 increased after IVIg treatment in patients with
[55]. On the other hand, hypersensitivity of pain in rats AMAN [64]. Interaction of TNF with TNFR2 promotes
with EAN accompanied by pronounced autoimmune expansion and function of mouse CD4?CD25? Tregs
inflammation in peripheral nerve and SC-derived TNF-a is [76]. More recently, TNF is found as an activator of
a major mediator of this hyper-algesia [68, 69]. Indeed, CD4?FoxP3?TNFR2? Tregs, which help terminate
TNF-a contributing to neuropathic pain has been shown in immune response [77]. Moreover, TNF activates a NF-kB
many recent studies [70]. regulated cellular program in human CD45RA-Tregs that
Although release of sTNF-Rs appear to be protective modulates their suppressive function [78].
since they can buffer TNF-a biological activities [71], Taken together, it seems that TNF-a might also have a
TNF-a itself might have a beneficial role in EAN. The dual role in EAN and GBS. TNFR1 exerts most of the
inhibition of TNF-a leads to a decrease of T cell apoptosis inflammatory effects, while both TNFR1 and TNFR2 may
during high dose antigen therapy in AT-EAN rats, making have protective effects, though the mechanism is not quite
TNF-a the principal candidate for mediating some of the clear.
effects of antigen therapy [72]. Moreover, previous study
showed that TNFR1 deficient mice in EAN exhibited a IL-12 and IL-23
more severe clinical signs (Fig. 2), in parallel with
enhanced numbers of inflammatory infiltrating cells in IL-12, mainly produced by phagocytes and dentric cells
peripheral nerves and splenic P0-reactive T cell prolifera- (DC), is critical for the differentiation of Th1 cells. IL-12 is
tion, as well as increased MHCII and CCR3 expression on a disulfide-linked heterodimer p70 complex composed of
the macrophages in cauda equina [73]. However, this result p40 and p35 subunit. By promoting IFN-c production,
is contrary to our previous study, mentioned above, that proliferation, and proliferation and cytolytic activity of NK
TNFR1 deficiency ameliorates EAN in mice. The con- and T cells, IL-12 mediates cellular immunity [64, 79].
flicting results may be due to the different P0 protein More recently it is found that Il-12p40 homodimer suppress
peptide applied or differential timing, location and quantity Treg cells via NO [80].
of TNF-a expressed and released [66]. We suppose that the Additional treatment of EAN in mice and rats with
exacerbation of EAN in TNFR1 deficient mice might be by mouse recombinant IL-12 (mrIL-12) prolonged the course
altering antigen-presenting cell (APC) function, increasing of EAN characterized by earlier clinical signs and delayed
T cell receptor signaling, and decreasing apoptosis of recovery stage [81, 82]. IL-12p40 deficient mice showed a
potentially auto-reactive T cells. Further, this protective delayed onset, reduced incidence and severity of EAN.
role of TNF-a/TNFR1 was also demonstrated in our pre- Fewer IFN-c and TNF-a producing cells but more cells
vious data in kainic acid-induced hippocampal injury in secreting IL-4 were found in sciatic nerve from IL-12
mice [74]. Similarly, TNF-a antagonist exacerbate the deficient EAN mice.
disease in multiple sclerosis (MS), another autoimmune In human, although numbers of IL-12p70 secreting
demyelinating disease in CNS [75]. PBMCs were not affected over the course of GBS [83],
In addition, TNFR2 might also have a protective role as increased IL-12 and IL-12R on PBMCs were found in
demonstrated in another study from our group that TNF serum of AIDP patients and decreased IL-12R after the
IVIg treatment [64].
These results indicate IL-12 has a major role in the
initiation, enhancement and perpetuation of pathogenic
events in EAN and GBS by promoting a Th1 cell-mediated
immune response and suppressing the Th2 response [16].
IL-23, a newly identified heterodimeric pro-inflamma-
tory cytokine and a novel IL-12 family member comprising
the p40 subunit of IL-12 but a different p19 subunit, has
been reported to preferentially act on memory T cells and
play an important role during cellular immune responses.
Recent evidence suggested that IL-23 rather than IL-12 is
Fig. 2 Effect of TNFR1 deficiency on the outcome of EAN. The critically involved in the pathogenesis of various immune-
wild type mice (n = 22) and TNFR1-/- mice (n = 23) were mediated disorder including EAE [84, 85]. IL-23 induces a
immunized with P0 peptide 180–199 in Freund’s complete adjuvant population of IL-17-producing cells that is more critically
(FCA) twice, at day 0 and day 7, and monitored for the development
involved in EAE pathogenesis than Th1 cells [86].
of clinical EAN. The results are presented as mean values ± SD of
clinical scores P \ 0.05, P \ 0.01. Data are representative of four IL-23 may play an essential role during the early ef-
separate experiments fecter phase in immune-mediated demyelination of the

123
J Neurol (2011) 258:533–548 537

peripheral nerve [87]. IL-23p19 RNA increased prior to the IL-6


onset of first clinical symptoms and peaked expression
levels preceding maximum clinical disease during EAN. In Mononuclear phagocytes, T cells, vascular endothelial
GBS patients, IL-23p19 protein was detectable in cere- cells, SCs and other cells synthesize IL-6 in response IL-1
brospinal fluid (CSF) samples [87]. and/or, TNF-a IL-6 acts by binding to the IL-6 receptor
Recently, IL-27 and IL-35 belong to IL-12 family have (IL-6-R), which can also exist as a soluble protein (sIL-6R).
also been investigated. IL-27 not only induces Th1 dif- The pleiotropic effects of IL-6 include inhibiting apoptosis
ferentiation but also suppresses immune responses by of auto-reactive T cells [100], BNB disturbance [13], reg-
inhibition of Th17 cells development and induction of ulating macrophage infiltration [101], B cell stimulation
IL-10 production. Administration of IL-27 or augmentation [102]. Trans-signaling into T cells via the sIL-6R com-
of IL-27 signaling suppresses some autoimmune disease pletely abrogates the generation of Treg cells [103, 104].
such as RA, systemic lupus erythematosus (SLE) [88]. Like TNF-a and IL-1, IL-6 also contributes in neuropathic
IL-35, composed of the IL-12p35 and EBI3, has been pain [105].
newly identified as an anti-inflammatory cytokine [89]. Overproduction of IL-6 within the PNS or systemically
However, roles of these two cytokines need to be explored were found in EAN and IL-6 is up-regulated in serum and
in EAN and GBS. CSF of GBS patients [94, 102, 106]. Intra-neural injection
of recombinant rat IL-6 (rrIL-6) into the sciatic nerve
IL-1b induces high inflammation and severe demyelination in
EAN [107]. Clinical trials of tocilizumab, a humanized
IL-1b is a member of the IL-1 gene family and synthesized anti-IL-6RmAb that blocks IL-6 signaling, have demon-
by MNC, mainly macrophages. It is also produced by SCs strated dramatic therapeutic benefit in RA [108, 109]. The
in PNS [90]. Expression of IL-1b mRNA in lymph node IL-6/sIL-6R complex has been seen as a novel target for
MNC and sciatic nerve section was observed prior to therapeutic approaches in inflammatory, autoimmune dis-
clinical EAN, indicating IL-1b participate in initiating the eases as well as cancer [110]. However, IL-6 might also
autoimmune response of EAN [13]. IL-1b can induce both have an anti-inflammatory role in chronic EAN since nasal
apoptosis and proliferation in cultured SCs, showing IL-1b administration of rrIL-6 in the initial phase decreased the
is not only involved in destruction of nerve, but also in the severity and the duration of clinical symptoms, accompa-
regeneration [91]. Moreover, IL-1b might also contribute nied by reduced auto-antigen reactivity of T cells as well as
to the neuropathic pain since impairment of IL-1 signaling TNF-a production [18].
attenuates neuropathic pain, autotomy and spontaneous This potential neuro-protective action of IL-6 is also
ectopic neuronal activity, following nerve injury in mice shown in some other condition such as diabetes related
[92]. neuropathy [111]. Furthermore, IL-6 can enhance PMP22
In GBS, increased expression of IL-1b was immune- production in SC via a JAK2-dependent pathway by
localized on SC membranes in sural nerve biopsies [93]. probably activating STAT3 and thus may contribute to
IL-1b could be detected in CSF of GBS [94], however, myelination [112]. IL-6R might be importantly involved in
serum IL-1b principally produced by macrophage, has not only SC proliferation but also re-myelination of the rat
been observed in solely a minority of GBS patients [95]. sciatic nerve [113]. sIL-6R, besides acting as an agonist of
IL-6, has been found some other properties such as IL-6-
IL-18 antagonistic effects, [114].
Consequently, IL-6, besides its pro-inflammatory role in
IL-18, is a member of the IL-1 family of pro-inflammatory EAN and GBS, might have protective effects by reducing
cytokines [96]. IL-18 enhances the IL-12-driven Th1 TNF, IL-1 and remyelination and, etc.
immune responses, but it can also stimulate Th2 immune
responses in the absence of IL-12 [97]. IL-18 serum levels IL-17 and IL-21
have been found to be increased in GBS patients and IL-18
was up-regulated in the nerve roots of EAN rats [98]. EAN Currently, it appears that IL-17-producing T cells are
mice treated with anti-IL-18 monoclonal antibody exhib- responsible for many of the inflammatory and autoimmune
ited a reduced clinical severity of disease and impaired Th1 responses once attributed to Th1 cells [115]. IL-17A, a pro-
response in inflamed nerves [14]. However, our recent inflammatory cytokine, was shown to be the hallmark
study showed that IL-18 deficiency inhibit IFN-c, TNF-a cytokine of this new T helper subset termed ‘‘Th17’’ [116].
and IL-10 production but not the clinical symptoms in A combination of TGFb1 and IL-6, together with IL-23
EAN, suggesting IL-18 may act as a co-inducer of both leads to generation of this CD4? T cell subtype. IL-17
Th1 and Th2 cytokines in EAN and possibly in GBS [99]. might enhance TNF-a signaling by regulating TNF

123
538 J Neurol (2011) 258:533–548

receptor [117]. It also protects B cells from apoptosis, healthy subjects [135]. Moreover, number of cells secreting
thereby increasing the number of autoantibody-producing TGF-b is elevated in all phase of GBS, but was statistically
cells [118]. IL-17A deficiency or neutralization in vivo significant only in the acute phase [132].
attenuates EAE [119]. Th17-associated cytokines such as The temporal relationship between increased levels of
IL-21 has been on spot as well. It might increase the Tregs TGF-b and the end of the progressive phase indicates that
numbers and function, and also up-regulate the IL-10 TGF-b might have a role in terminating the pathological
expression. [120, 121]. immune response in GBS.
But so far, the function of Th17 and its related cytokines
such as IL-17, IL-21 needs to be explored in EAN and GBS. IL-10

TGF-b IL-10 is produced by monocytes, macrophages, B cells,


Th2 cells and various subsets of CD4? and CD8? T cells.
TGF-b family consists of TGF-b1, TGF-b2, TGF-b3, with Its dominant role is to be as an immunosuppressive cyto-
TGF-b1 being the predominant form expressed in the kine by inhibiting macrophage and DC function, including
immune system [122]. The potential principal cell sources cytokine production such as IL-1 and TNF-a, co-stimula-
of TGF-b include Treg cells, APCs, leukocytes and B cells tors and MHC expression, general APC function. IL-10 can
[123]. also inhibit T cell proliferation and promote humoral
The pivotal function of TGF-b is to maintain tolerance immune responses enhancing class II expression on B cells
via the regulation of lymphocyte proliferation, differenti- and Ig production including IgA [136]. Moreover, IL-10
ation, and survival. It also controls inflammatory response plays a crucial role for Treg cell development [136].
by regulating chemotaxis, activation and survival of NK Pathways leading to IL-10 production by different T cell
cells, DCs and macrophages, etc. [124]. Furthermore, TGF- populations have been recently revealed and it is becoming
b-driven APC exhibited the morphology, phenotypes and clear that some pro-inflammatory cytokines directly induce
functions of tolerogenic immature DC, and had lower IL-10, allowing immune responses to be inherently self-
capacity to stimulate T cells [125, 126]. regulating [137].
Recently TGF-b has been in spot because of its Administration of a low-dose of IL-10 permits better
emerging role in Treg and Th17 cells [20, 21]. TGF-b regeneration of damaged axons [138]. SCs produce IL-10
production by Treg cells has also been suggested as an that might counterbalance the pro-inflammatory cytokines
important (if controversial) mechanism of suppression in in reducing the neuropathic pain [139, 140]. Intrathecal
autoimmune response [21]. Actually, TGF-b1, by inducing IL-10 gene therapy attenuates paclitaxel-induced mechan-
Treg cells, has potential clinical application in treating SLE ical allodynia and pro-inflammatory cytokine expression in
[127]. Local but not systemic antibody blockade of TGF-b dorsal root ganglia in rats [140].
prevented Th17 cell differentiation and the onset of EAE In EAN, up-regulation of IL-10 mRNA expression was
[128]. However, it has been suggested that the differenti- seen in lymph node MNC and sciatic nerve on the onset of
ation of human Th17 cells is not associated with TGF-b, EAN, and maximal expression at onset of clinical recovery
even suppressed by TGF-b, thus differs fundamentally [13]. Treatment with IL-10 ameliorates the inflammatory
from that in mice. TGF-b enhanced expression of Th17 responses in EAN [19]. Amino acid copolymer-specific
transcription factor RORgammat but simultaneously IL-10-secreting Treg cells could ameliorate EAN [141].
inhibited its ability to induce IL-17 expression [129, 130]. In GBS, although serum IL-10 is un-elevated [29],
In peripheral tissues, similarly, TGF-b inhibits T cell IL-10-secreting BMNC increased during the acute phase,
proliferation, activation and effecter T cells differentiation which may account for the self-limited clinical course of
and maintains Treg cells, thereby might contributing to GBS [29, 83]. On the other hand, Myhr et al. [142] reported
prevent the autoimmune disease. In fact, TGF-b1 in plasma that GBS is related to IL-10 promoter polymorphisms and
were decreased in the first days of the disease and associ- IL-10 worsens the disease via enhancing the ganglioside
ated with a loss of immune-regulatory function [131], antibody production probably.
however, both TGF-b1 cytokine and mRNA level Altogether, the anti-inflammatory cytokine IL-10 has a
increased during the recovery of GBS [132]. This switch in role in terminating the EAN and GBS, however, might also
the cytokine profile may instead precede the reversal of worsen the disease by promoting the ganglioside antibody.
clinical symptoms, which was also as noted in EAN [133,
134]. Accordingly, the level of TGF-b1 in serum of GBS IL-4
peaked in the plateau phase, i.e. before onset of recovery
and declined during recovery phase but still higher than IL-4 is produced by CD4? Th2 cells and participates in the
patients with non-inflammatory neurological diseases and differentiation and growth of B cells. IL-4 promotes Th

123
J Neurol (2011) 258:533–548 539

cells towards a differentiation into Th2 phenotype through CX3CL1/CX3CR1 may play a role in the pain-like
intra-signaling factor STAT6. IL-4 inhibits the activation behavior [153].
of Th1 cells and this in turn, decreases the production of
IL-1 and TNF-a.
Increase of IL-4 in the recovery phase was shown by Therapies on cytokines in EAN and GBS (Table 1)
detection of cytokine mRNA expression in EAN [133].
Nasal administration of rrIL-4 ameliorates ongoing EAN Modulation of cytokine pathway
and inhibits demyelination [107]. Self-limitation of acute
EAN may owe to the rising levels of IL-4 and IL-10 [143]. IL-12 activated Th1 cells via the JAK2/STAT4 pathway,
Serum IL-4 were also elevated during acute and recovery whereas IL-4 activated Th2 cells via JAK1-3/STAT6 [154–
stage of GBS [29, 144]. 156]. Suppressors of cytokine signaling (SOC) act in a
Thus, the anti-inflammatory cytokine IL-4 may have a negative feedback loop to suppress further JAK/STAT
role in terminating EAN and GBS. signaling [157–159]. NF-kB and MAPK transcriptions are
also important in cytokine expression. The new anti-
Chemokines depression drugs treated the sickness behavior’s animal
models and the patients suffered from depression can
Chemokines are small cytokines that are involved in the inhibit inflammatory markers and SAPK/MAPK and/or
directed migrations (chemotaxis) and the activation of JAK/STAT signaling in vitro [160]. Many drugs targeting
cells, especially phagocytes and lymphocytes, and thereby NF-kB have been recently thought a promising molecular
play a central role in inflammatory responses [145–147]. therapy in autoimmune disease such as RA [161].
Chemokines such as MIP-1a, MCP-1, CCR2, RANTES Administered at the time of immunization and on day 14th
and IP-10 play an important role in the recruitment of post-immunization, immunosuppressive agent cyclophos-
MNC into the PNS of EAN rats and GBS patients [148– phamide (CY) prevents clinical EAN and also reduces
151]. Patients examined in the acute phase of GBS prior to cytokine and NF-kBp65 expression [162]. Inhibition of
treatment with IVIg had elevated CSF levels of MCP-1 and ERK MAPK suppresses IL-23- and IL-1-driven IL-17
IP-10 [152]. The expression of CX3CL1 (fractalkine) and production and attenuates autoimmune disease [163]. More
its receptor CX3CR1 were also higher in EAN than in recently, the signaling AKT axis becomes attractive for
control dorsal horns suggesting spinal microglia and therapeutic targeting in autoimmune disease [164].

Table 1 Possible therapies on EAN and GBS


Possible therapies intervention in GBS and EAN
Modulation of CK pathway JAK/STAT, NF-kB, MAPK, AKT axis
mAb and fusion protein sTNFR1, Anti-TNF-a mAb
IL-1bR antagonist
Anti-IL-6RmAb
Blocking IL-12/23
Blocking IFN-c
Neuroprotections Induce auto-reactive T cell apoptosis IFN-c, TNF-a, IL-6
Maintain Treg cell IFN-c, TGF-b, IL-10
Induce SC proliferation IFN-c, IL-1 b, IL-6
Reduce the neuropathic pain, enhance nerve repair IL-10
Counteract Th1 cells IL-10, IL-4
Increased GM1 specific T cell reactivity; down-regulation of macrophage function
Compounds applied in EAN Rolipram: down-regulate antigen-driven T cell proliferation, suppress TNF-a/b production
ABR-215062: regulation of Th1/Th2 cytokine balance, inhibited migration of inflammatory cells into the PNS
VPA: suppressed mRNA levels of IFN-c, TNF-a, IL-1b, IL-4, IL-6 and IL-17 in the lymph nodes
Minocycline: inhibit mRNA expressions of MMP-9, iNOS and proinflammatory cytokines IL-1b and TNF-a in
EAN sciatic nerves
Compound A: depressed Th1 and Th17 cytokines, but increased Th2 cytokine and Foxp3 expression in lymph
nodes of EAN rats
MS-275: suppressing inflammatory T cells, macrophages and cytokines

123
540 J Neurol (2011) 258:533–548

Table 2 Cytokine type, cellular origin and pathogenetic relevance, role as biomarker in EAN and GBS
Cytokine Origin Function Biomarker in EAN and GBS
Inflammation Anti-inflammation EAN GBS

IFN-r T, NK cells Mø activation T cell apoptosis IFN-c producing cells Serum IFN-c was
T differentiation to Th1 Inhibit neutrophil trafficking parallel clinical EAN elevated
MHC I/II induction Convert CD4?CD25- cells Systemic administration of Auto-Abs to IFN-c are
into CD4? Treg cells rrIFN-c worsens EAN correlated with down-
B cell class switching regulation of IFN-c
Suppress the differentiation IFN-cR-/- mice showed a
Enhance CK suppression of EAN producing cells and
production such as and effector function of with improved clinical
TNF-a, IL-1b, IL-6, Th17 cells disability
as well as Microglia activated by IFN-
Convert peripheral
metalloproteinases, c or IL-4 can protect CD4?CD25- T cells
chemokines and neutrons and support both to CD4?CD25? Treg
adhesion molecules neurogenesis and cells
Up-regulates CK oligodendrogenesis
iNOS-specific mRNA
in SCs
TNF-a Mø, T cells, SCs, etc. Promotes macrophage Might decrease APC Might have effect on BNB Serum TNF-a was
recruitment function TNFR1-/- reduces APC elevated
Inhabits SC Might down-regulate T cell function of SCs and TNF-a converting
proliferation and receptor signaling ameliorates EAN enzyme increased
mediates SC death Might increase apoptosis of Implicated in SC apoptosis sTNFR1 increased while
TNFR1, a positive T- auto-reactive T cells in EAN TNF-a and TNFR1
cell costimulatory Activator of SC-derived TNF-a, a major decreased following
molecule for T cell CD4?FoxP3?TNFR2? mediator of hyper-algesia treatment
activation Tregs, which help TNF and TNFR2
TNFR1-/- EAN mice
terminate immune showed more severe increased after IVIg
response clinical signs, with treatment
enhanced infiltrating TNF–TNFR2 promotes
inflammatory cells, T expansion and function
proliferation, and MHC of mouse Tregs
and CCR 3 expression on

IL-12 Mainly phagocytes Differentiation of Th1 mrIL-12 treatment Increased IL-12 and IL-
and DC cells prolonged EAN course 12R on PBMCs were
Promoting IFN-c IL-12p40-/- mice showed found in serum of
production, a delayed onset, reduced AIDP patients and
proliferation incidence and severity of decreased IL-12R after
EAN the IVIg treatment;
Proliferation and
cytolytic activity of Fewer IFN-c and TNF-a
NK and T cells producing cells, but more
IL-12p40 homodimer cells secreting IL-4 were
suppress Treg cells found in IL-12-/- EAN
via nitric oxide mice
IL-23 A novel IL-12 family Act on memory T cells IL-23p19 RNA increased IL-23p19 protein was
member and play an important prior to the onset of detectable in CSF
role during cellular clinical symptoms and
immune responses peaked preceding
May play an important maximum clinical disease
role during the early
effecter phase in
immune-mediated
demyelination of the
peripheral nerve
IL-1b MNC, mainly Mø, also Induce SC apoptosis Induce SC proliferation IL-1b mRNA in lymph node IL-1b could be detected
SC Contribute to the MNC and sciatic nerve in CSF
neuropathic pain section was observed prior Increased IL-1b was
to clinical EAN immunolocalized on
SC membranes in sural
nerve biopsies

123
J Neurol (2011) 258:533–548 541

Table 2 continued
Cytokine Origin Function Biomarker in EAN and GBS
Inflammation Anti-inflammation EAN GBS

IL-18 A member of the IL-1 Enhances the IL-12- IL-18 was up-regulated Serum IL-18 increased
family of pro- driven Th1 responses, Anti-IL-18 Ab reduced
inflammatory CK, as well as Th2 clinical severity and Th1
produced in many responses in the response
cells including Mø, absence of IL-12
endothelial cells, DC, IL-18-/- inhibit IFN-c,
etc. TNF-a and IL-10
production but not clinical
symptoms
IL-6 Mononuclear Inhibites apoptosis of Enhance PMP22 production Overproduction of IL-6 IL-6 is up-regulated in
phagocytes, T cells, auto-reactive T-cells in SC, thus may contribute within the PNS or serum and CSF of
vascular endothelial BNB disturbance to myelination systemically patients
cells, SCs and other IL-6R might be involved in Nasal administration of rrIL-
cells Regulates Mø
infiltration SC proliferation and re- 6 in initial EAN decreased
myelination of the rat severity and duration; but
B cell stimulation; sciatic nerve intra-neural injection of
contributes in rrIL-6 into the sciatic
neuropathic pain sIL-6R, also have IL-6-
antagonistic effects nerve induced high
IL-6/sIL-6R complex inflammation and
abrogates the demyelination
generation of Treg
cells
TGF-b Treg cells, APCs, Maintain tolerance by Decreased in the initial Decreased in the initial
leukocytes and B regulating differentiation, stage, TGF-b1 cytokine stage, TGF-b1
cells survival of lymphocyte, as and mRNA level increased cytokine and mRNA
well as NK cells, DCs, Mø during the recovery level increased during
and chemotaxis the recovery, peaked in
TGF-b-driven APC plateau phase
exhibited lower capacity to Number of cells
stimulate T cells secreting TGF-b is
Human Th17 cells are not elevated in all phase,
associated with TGF-b, but significant in acute
even suppressed by it stage
IL-10 Monocytes, Mø, B Inhibit Mø and DC function, Up-regulation of IL-10 Serum IL-10 is un-
cells, Th2 cells, including CK production mRNA expression was elevated, however, IL-
various subsets of such as IL-1 and TNF-a, seen on the onset and 10-secreting BMNC
CD4? and CD8? T co-stimulators and MHC maximal expression at increased during the
cells expression, general APC onset of clinical recovery acute phase
function Treatment with IL-10 IL-10 worsens the
Inhibit T cell proliferation ameliorates EAN disease via enhancing
and promote humoral Amino acid copolymer- the ganglioside Ab
immune responses specific IL-10-secreting production probably
enhancing class II Treg cells ameliorate EAN
expression on B cells and Ig
production including IgA
Treg cell development
Administration of a low-
dose of IL-10 permits
better regeneration of
axons
SCs produce IL-10 reducing
the neuropathic pain
Intrathecal IL-10 gene
therapy attenuates
paclitaxel-induced
mechanical allodynia and
pro-inflammatory CK
expression in dorsal root
ganglia in rats

123
542 J Neurol (2011) 258:533–548

Table 2 continued
Cytokine Origin Function Biomarker in EAN and GBS
Inflammation Anti-inflammation EAN GBS

IL-4 CD4? Th2 cells Promotes Th cells towards a mRNA expression of IL-4 is Serum IL-4 were
differentiation into Th2 shown in the recovery elevated during acute
Inhibits the activation of phage of EAN and recovery stage
Th1 and decreases the Nasal administration of rrIL-
production of IL-1 and 4 ameliorates EAN and
TNF-a inhibits demyelination
Mø, macrophage; -/-, knockout; CK cytokine

Neutralization of cytokines 3. Valproic acid (VPA): a short-chain branched fatty,


effectively suppress inflammation in EAN [177]. VPA
Blockade of cytokine by monoclonal antibody or fusion administration to EAN rats significantly suppressed
proteins have been investigated. Exogenous sTNFR1 mRNA levels of IFN-c, TNF-a, IL-1b, IL-4, IL-6 and
ameliorates murine EAN [52]. Anti-TNF-a mAb and IL-17 in the lymph nodes. In peripheral blood and
sTNFR, IL-1bR antagonist, anti-IL-6RmAb have effi- sciatic nerves of EAN rats, Foxp3(?) cells increased
ciency in treatment of autoimmune disease such as RA but IL-17(?) cells decreased during VPA treatment.
[165]. Drugs such as ABT-84, Apilimod, CNTO-1275 that 4. Minocycline: obviously inhibit mRNA expressions of
block IL12/IL23 are being tested clinically in disease such MMP-9, iNOS and proinflammatory cytokines IL-1b
as Crohn’s disease, psoriasis, RA, MS [166–168]. Although and TNF-a in EAN sciatic nerves. It is very clear that
absence of IFN-c causes worse EAE in some studies [169, Minocycline could effectively apply in the conditions
170], antibody to IFN-c not TNF-a showed significantly with the peripheral and spinal inflammation to improve
improvement in MS [171]. Skurkovich [172] indicated that outcome and attenuated mechanical allodynia in EAN
cytokines act in an interrelated immune cascade with IFN-c rats [178].
playing a central role, blocking IFN-c is the safest and most 5. Compound A: a plant-derived phenyl aziridine pre-
effective method of treatment of Th1-mediated autoim- cursor, depressed Th1 and Th17 cytokines, but
mune diseases, although blocking other cytokines in the increased Th2 cytokine and Foxp3 expression in
cascade may lead to some therapeutic effects as well. lymph nodes of EAN rats [179].
6. MS-275: a potent histone deacetylase inhibitor, could
Cytokines for neuroprotection and regeneration effectively suppress inflammation in EAN, through
suppressing inflammatory T cells, macrophages and
IFN-c, TNF-a, IL-6 induce auto-reactive T cell apoptosis; cytokines [180].
IFN-c, TGF-b, IL-10 maintain Treg cell; IL-1, IL-6, IFN-c
induce SC proliferation; IL-10 reduces the neuropathic
pain, enhance nerve repair, etc. More recently, patients
with increased anti-GM1 T cell reactivity were signifi- Conclusions
cantly less severe than those without such reactivity. This
raises the possibility that increased GM1 specific T cell Cytokines in EAN and GBS have been extensively studied
reactivity and its related cytokine might have an immune- these years and sometimes the conflicting results make it
regulatory or neuro-protective role [173]. more complicated (Table 2). Besides the complexity of the
cytokines themselves, this is partly due to the different
Reagents applied in EAN treatment methods used and aspects of cytokines observed in the
animal model and clinical studies. Agents that block the
1. Rolipram: markedly down-regulate antigen-driven T egress of T cells across the BNB and agents that antagonise
cell proliferation and suppress TNF-a/b production in T cell cytokine-induced priming of macrophages may prove
vitro and in vivo, which have led to its use in the efficacious [5]. However, in recent years, the traditional
treatment of EAE and EAN [174, 175]. inflammatory cytokines such as IFN-c, TNF-a, IL-6 and IL-1
2. ABR-215062: immune regulator derived from Lino- are also found to have a protective role in EAN and GBS,
mide, could suppress EAN by regulation of Th1/Th2 although the exact mechanism remain elusive. They might
cytokine balance and inhibited migration of inflam- exert their dual roles through different signal ways in vari-
matory cells into the PNS [176]. ous conditions. Therefore, therapies on cytokines especially

123
J Neurol (2011) 258:533–548 543

by regulating their pathways are important and promising. 16. Bao L, Lindgren JU, van der Meide P, Zhu S, Ljunggren HG,
Treg cells are becoming increasingly attentive due to their Zhu J (2002) The critical role of IL-12p40 in initiating,
enhancing, and perpetuating pathogenic events in murine
suppressive roles in EAN and GBS. Moreover, Th17 and experimental autoimmune neuritis. Brain Pathol 12:420–429
some cytokines such as IL-17, 21, 27, 35 are on spot pres- 17. Kiefer R, Funa K, Schweitzer T, Jung S, Bourde O, Toyka KV,
ently in autoimmune diseases, though their possible roles in Hartung HP (1996) Transforming growth factor-beta 1 in
GBS need to be further explored. experimental autoimmune neuritis. Cellular localization and
time course. Am J Pathol 148:211–223
18. Deretzi G, Pelidou S, Zou L, Quiding C, Mix E, Levi M,
Acknowledgments The studies of the authors referred to in this Wahren B, Zhu J (1999) Suppression of chronic experimental
review were supported by grants from the Swedish Research Council autoimmune neuritis by nasally administered recombinant rat
(K2007-62X-13133-09-3) and from ALF-project grant of Karolinska interleukin-6. Immunology 97:69–76
Institute University Hospital, Stockholm, Sweden and grant from 19. Bai XF, Zhu J, Zhang GX, Kaponides G, Hojeberg B, van der
Swedish Medical Association. Meide PH, Link H (1997) IL-10 suppresses experimental auto-
immune neuritis and down-regulates TH1-type immune
responses. Clin Immunol Immunopathol 83:117–126
20. Li MO, Wan YY, Flavell RA (2007) T cell-produced trans-
References forming growth factor-beta1 controls T cell tolerance and reg-
ulates Th1- and Th17-cell differentiation. Immunity 26:579–591
1. van Doorn PA, Ruts L, Jacobs BC (2008) Clinical features, 21. McGeachy MJ, Cua DJ (2007) T cells doing it for themselves:
pathogenesis, and treatment of Guillain–Barre syndrome. Lancet TGF-beta regulation of Th1 and Th17 cells. Immunity
Neurol 7:939–950 26:547–549
2. Griffin JW, Li CY, Ho TW, Xue P, Macko C, Gao CY, Yang C, 22. Harness J, McCombe PA (2008) Increased levels of activated
Tian M, Mishu B, Cornblath DR (1995) Guillain–Barre syn- T-cells and reduced levels of CD4/CD25? cells in peripheral
drome in northern China. The spectrum of neuropathological blood of Guillain–Barre syndrome patients compared to con-
changes in clinically defined cases. Brain 118(Pt 3):577–595 trols. J Clin Neurosci 15:1031–1035
3. Fisher M (1956) An unusual variant of acute idiopathic poly- 23. Chi LJ, Wang HB, Zhang Y, Wang WZ (2007) Abnormality of
neuritis (syndrome of ophthalmoplegia, ataxia and areflexia). circulating CD4(?)CD25(?) regulatory T cell in patients with
N Engl J Med 255:57–65 Guillain–Barre syndrome. J Neuroimmunol 192:206–214
4. Yuki N (2007) Ganglioside mimicry and peripheral nerve dis- 24. Schmidt B, Stoll G, van der Meide P, Jung S, Hartung HP (1992)
ease. Muscle Nerve 35:691–711 Transient cellular expression of gamma-interferon in myelin-
5. Vucic S, Kiernan MC, Cornblath DR (2009) Guillain–Barre induced and T-cell line-mediated experimental autoimmune
syndrome: an update. J Clin Neurosci 16:733–741 neuritis. Brain 115(Pt 6):1633–1646
6. Hughes RA, Hadden RD, Gregson NA, Smith KJ (1999) Patho- 25. Girkontaite I, Urbonaviciute V, Maseda D, Neubert K, Herr-
genesis of Guillain–Barre syndrome. J Neuroimmunol 100:74–97 mann M, Voll RE (2007) Apoptotic cells selectively suppress
7. Willison HJ, Yuki N (2002) Peripheral neuropathies and anti- the Th1 cytokine interferon gamma in stimulated human
glycolipid antibodies. Brain 125:2591–2625 peripheral blood mononuclear cells and shift the Th1/Th2 bal-
8. Hughes RA, Swan AV, Raphael JC, Annane D, van Koningsveld ance towards Th2. Autoimmunity 40:327–330
R, van Doorn PA (2007) Immunotherapy for Guillain–Barre 26. Gold R, Zielasek J, Kiefer R, Toyka KV, Hartung HP (1996)
syndrome: a systematic review. Brain 130:2245–2257 Secretion of nitrite by Schwann cells and its effect on T-cell
9. Hahn AF (1998) Guillain–Barre syndrome. Lancet 352:635–641 activation in vitro. Cell Immunol 168:69–77
10. Hughes RA, Swan AV, van Koningsveld R, van Doorn PA 27. Gold R, Toyka KV, Hartung HP (1995) Synergistic effect of
(2006) Corticosteroids for Guillain–Barre syndrome. Cochrane IFN-gamma and TNF-alpha on expression of immune molecules
Database Syst Rev: CD001446 and antigen presentation by Schwann cells. Cell Immunol
11. van der Meche FG, Schmitz PI (1992) A randomized trial 165:65–70
comparing intravenous immune globulin and plasma exchange 28. Zhu Y, Ljunggren HG, Mix E, Li HL, van der Meide P, Elhassan
in Guillain–Barre syndrome. Dutch Guillain–Barre Study AM, Winblad B, Zhu J (2001) Suppression of autoimmune
Group. N Engl J Med 326:1123–1129 neuritis in IFN-gamma receptor-deficient mice. Exp Neurol
12. Zhu J, Mix E, Link H (1998) Cytokine production and the 169:472–478
pathogenesis of experimental autoimmune neuritis and Guillain– 29. Hohnoki K, Inoue A, Koh CS (1998) Elevated serum levels of
Barre syndrome. J Neuroimmunol 84:40–52 IFN-gamma, IL-4 and TNF-alpha/unelevated serum levels of
13. Zhu J, Bai XF, Mix E, Link H (1997) Cytokine dichotomy in IL-10 in patients with demyelinating diseases during the acute
peripheral nervous system influences the outcome of experi- stage. J Neuroimmunol 87:27–32
mental allergic neuritis: dynamics of mRNA expression for IL-1 30. Csurhes PA, Sullivan AA, Green K, Greer JM, Pender MP,
beta, IL-6, IL-10, IL-12, TNF-alpha, TNF-beta, and cytolysin. McCombe PA (2005) Increased circulating T cell reactivity to
Clin Immunol Immunopathol 84:85–94 GM1 ganglioside in patients with Guillain–Barre syndrome.
14. Yu S, Chen Z, Mix E, Zhu SW, Winblad B, Ljunggren HG, Zhu J Clin Neurosci 12:409–415
J (2002) Neutralizing antibodies to IL-18 ameliorate experi- 31. Elkarim RA, Dahle C, Mustafa M, Press R, Zou LP, Ekerfelt C,
mental autoimmune neuritis by counter-regulation of autoreac- Ernerudh J, Link H, Bakhiet M (1998) Recovery from Guillain–
tive Th1 responses to peripheral myelin antigen. J Neuropathol Barre syndrome is associated with increased levels of neutral-
Exp Neurol 61:614–622 izing autoantibodies to interferon-gamma. Clin Immunol Im-
15. Nicoletti F, Creange A, Orlikowski D, Bolgert F, Mangano K, munopathol 88:241–248
Metz C, Di Marco R, Al Abed Y (2005) Macrophage migration 32. Chu CQ, Wittmer S, Dalton DK (2000) Failure to suppress the
inhibitory factor (MIF) seems crucially involved in Guillain– expansion of the activated CD4 T cell population in interferon
Barre syndrome and experimental allergic neuritis. J Neuroim- gamma-deficient mice leads to exacerbation of experimental
munol 168:168–174 autoimmune encephalomyelitis. J Exp Med 192:123–128

123
544 J Neurol (2011) 258:533–548

33. Jones LS, Rizzo LV, Agarwal RK, Tarrant TK, Chan CC, mediates the death of Th1 cells in a paracrine manner.
Wiggert B, Caspi RR (1997) IFN-gamma-deficient mice J Immunol 180:842–849
develop experimental autoimmune uveitis in the context of a 50. Huang S, Li L, Liang S, Wang W (2009) Conversion of
deviant effector response. J Immunol 158:5997–6005 peripheral CD4(?)CD25(-) T cells to CD4(?)CD25(?) regu-
34. Kelchtermans H, Struyf S, De Klerck B, Mitera T, Alen M, latory T cells by IFN-gamma in patients with Guillain–Barre
Geboes L, Van Balen M, Dillen C, Put W, Gysemans C, Billiau syndrome. J Neuroimmunol 217:80–84
A, Van Damme J, Matthys P (2007) Protective role of IFN- 51. Murwani R, Hodgkinson S, Armati P (1996) Tumor necrosis
gamma in collagen-induced arthritis conferred by inhibition of factor alpha and interleukin-6 mRNA expression in neonatal
mycobacteria-induced granulocyte chemotactic protein-2 pro- Lewis rat Schwann cells and a neonatal rat Schwann cell line
duction. J Leukoc Biol 81:1044–1053 following interferon gamma stimulation. J Neuroimmunol
35. Nicoletti F, Zaccone P, Di Marco R, Magro G, Grasso S, Stivala 71:65–71
F, Calori G, Mughini L, Meroni PL, Garotta G (1998) Para- 52. Bao L, Lindgren JU, Zhu Y, Ljunggren HG, Zhu J (2003)
doxical antidiabetogenic effect of gamma-interferon in DP-BB Exogenous soluble tumor necrosis factor receptor type I ame-
rats. Diabetes 47:32–38 liorates murine experimental autoimmune neuritis. Neurobiol
36. Sobel DO, Newsome J (1997) Gamma interferon prevents dia- Dis 12:73–81
betes in the BB rat. Clin Diagn Lab Immunol 4:764–768 53. Smith CA, Farrah T, Goodwin RG (1994) The TNF receptor
37. Sobel DO, Han J, Williams J, Yoon JW, Jun HS, Ahvazi B superfamily of cellular and viral proteins: activation, costimu-
(2002) Gamma interferon paradoxically inhibits the develop- lation, and death. Cell 76:959–962
ment of diabetes in the NOD mouse. J Autoimmun 19:129–137 54. Shubayev VI, Angert M, Dolkas J, Campana WM, Palenscar K,
38. (1992) Double blind controlled phase III multicenter clinical Myers RR (2006) TNFalpha-induced MMP-9 promotes macro-
trial with interferon gamma in rheumatoid arthritis. German phage recruitment into injured peripheral nerve. Mol Cell
Lymphokine Study Group. Rheumatol Int 12:175–185 Neurosci 31:407–415
39. Machold KP, Neumann K, Smolen JS (1992) Recombinant 55. Cheng C, Qin Y, Shao X, Wang H, Gao Y, Cheng M, Shen A
human interferon gamma in the treatment of rheumatoid (2007) Induction of TNF-alpha by LPS in Schwann cell is
arthritis: double blind placebo controlled study. Ann Rheum Dis regulated by MAPK activation signals. Cell Mol Neurobiol
51:1039–1043 27:909–921
40. Sigidin YA, Loukina GV, Skurkovich B, Skurkovich S (2001) 56. Myers RR, Sekiguchi Y, Kikuchi S, Scott B, Medicherla S,
Randomized, double-blind trial of anti-interferon-gamma anti- Protter A, Campana WM (2003) Inhibition of p38 MAP kinase
bodies in rheumatoid arthritis. Scand J Rheumatol 30:203–207 activity enhances axonal regeneration. Exp Neurol 184:606–614
41. Kelchtermans H, Billiau A, Matthys P (2008) How interferon- 57. Boyle K, Azari MF, Cheema SS, Petratos S (2005) TNFalpha
gamma keeps autoimmune diseases in check. Trends Immunol mediates Schwann cell death by upregulating p75NTR expres-
29:479–486 sion without sustained activation of NFkappaB. Neurobiol Dis
42. Su SB, Grajewski RS, Luger D, Agarwal RK, Silver PB, Tang J, 20:412–427
Tuo J, Chan CC, Caspi RR (2007) Altered chemokine profile 58. Tao T, Ji Y, Cheng C, Yang H, Liu H, Sun L, Qin Y, Yang J,
associated with exacerbated autoimmune pathology under con- Wang H, Shen A (2009) Tumor necrosis factor-alpha inhibits
ditions of genetic interferon-gamma deficiency. Invest Oph- Schwann cell proliferation by up-regulating Src-suppressed
thalmol Vis Sci 48:4616–4625 protein kinase C substrate expression. J Neurochem 111:647–
43. Wang Z, Hong J, Sun W, Xu G, Li N, Chen X, Liu A, Xu L, Sun 655
B, Zhang JZ (2006) Role of IFN-gamma in induction of Foxp3 59. Evangelidou M, Tseveleki V, Vamvakas SS, Probert L (2010)
and conversion of CD4? CD25- T cells to CD4? Tregs. J Clin TNFRI is a positive T-cell costimulatory molecule important for
Invest 116:2434–2441 the timing of cytokine responses. Immunol Cell Biol 88:586–
44. Luger D, Silver PB, Tang J, Cua D, Chen Z, Iwakura Y, 595
Bowman EP, Sgambellone NM, Chan CC, Caspi RR (2008) 60. Radhakrishnan VV, Sumi MG, Reuben S, Mathai A, Nair MD
Either a Th17 or a Th1 effector response can drive autoimmu- (2004) Serum tumour necrosis factor-alpha and soluble tumour
nity: conditions of disease induction affect dominant effector necrosis factor receptors levels in patients with Guillain–Barre
category. J Exp Med 205:799–810 syndrome. Acta Neurol Scand 109:71–74
45. Kelchtermans H, Schurgers E, Geboes L, Mitera T, Van Damme 61. Zhang J, Dong H, Li B, Li CY, Guo L (2007) Association of
J, Van Snick J, Uyttenhove C, Matthys P (2009) Effector tumor necrosis factor polymorphisms with Guillain–Barre syn-
mechanisms of interleukin-17 in collagen-induced arthritis in drome. Eur Neurol 58:21–25
the absence of interferon-gamma and counteraction by inter- 62. Kurz M, Pischel H, Hartung HP, Kieseier BC (2005) Tumor
feron-gamma. Arthritis Res Ther 11:R122 necrosis factor-alpha-converting enzyme is expressed in the
46. Shaked I, Tchoresh D, Gersner R, Meiri G, Mordechai S, Xiao inflamed peripheral nervous system. J Peripher Nerv Syst
X, Hart RP, Schwartz M (2005) Protective autoimmunity: 10:311–318
interferon-gamma enables microglia to remove glutamate 63. Sharief MK, Ingram DA, Swash M, Thompson EJ (1999) I.v.
without evoking inflammatory mediators. J Neurochem 92:997– immunoglobulin reduces circulating proinflammatory cytokines
1009 in Guillain–Barre syndrome. Neurology 52:1833–1838
47. Schwartz M, Butovsky O, Kipnis J (2006) Does inflammation in 64. Deng H, Yang X, Jin T, Wu J, Hu LS, Chang M, Sun XJ, Adem
an autoimmune disease differ from inflammation in neurode- A, Winblad B, Zhu J (2008) The role of IL-12 and TNF-alpha in
generative diseases? Possible implications for therapy. J Neuro- AIDP and AMAN. Eur J Neurol 15:1100–1105
immune Pharmacol 1:4–10 65. Ruiz de Souza V, Carreno MP, Kaveri SV, Ledur A, Sadeghi H,
48. Shaked I, Porat Z, Gersner R, Kipnis J, Schwartz M (2004) Early Cavaillon JM, Kazatchkine MD, Haeffner-Cavaillon N (1995)
activation of microglia as antigen-presenting cells correlates Selective induction of interleukin-1 receptor antagonist and
with T cell-mediated protection and repair of the injured central interleukin-8 in human monocytes by normal polyspecific IgG
nervous system. J Neuroimmunol 146:84–93 (intravenous immunoglobulin). Eur J Immunol 25:1267–1273
49. Foulds KE, Rotte MJ, Paley MA, Singh B, Douek DC, Hill BJ, 66. Mao XJ, Zhang XM, Zhang HL, Quezada HC, Mix E, Yang X,
O’Shea JJ, Watford WT, Seder RA, Wu CY (2008) IFN-gamma Winblad B, Adem A, Zhu J (2010) TNF-alpha receptor 1

123
J Neurol (2011) 258:533–548 545

deficiency reduces antigen-presenting capacity of Schwann cells recombinant mouse interleukin-12 increases inflammation and
and ameliorates experimental autoimmune neuritis in mice. demyelination in chronic experimental autoimmune neuritis in
Neurosci Lett 470:19–23 Lewis rats. Scand J Immunol 51:29–35
67. Taylor JM, Pollard JD (2007) Soluble TNFR1 inhibits the 83. Press R, Ozenci V, Kouwenhoven M, Link H (2002) Non-T(H)1
development of experimental autoimmune neuritis by modulat- cytokines are augmented systematically early in Guillain–Barre
ing blood-nerve-barrier permeability and inflammation. J Neu- syndrome. Neurology 58:476–478
roimmunol 183:118–124 84. Cua DJ, Sherlock J, Chen Y, Murphy CA, Joyce B, Seymour
68. Campana WM (2007) Schwann cells: activated peripheral glia B, Lucian L, To W, Kwan S, Churakova T, Zurawski S,
and their role in neuropathic pain. Brain Behav Immun Wiekowski M, Lira SA, Gorman D, Kastelein RA, Sedgwick
21:522–527 JD (2003) Interleukin-23 rather than interleukin-12 is the
69. Moalem-Taylor G, Allbutt HN, Iordanova MD, Tracey DJ critical cytokine for autoimmune inflammation of the brain.
(2007) Pain hypersensitivity in rats with experimental autoim- Nature 421:744–748
mune neuritis, an animal model of human inflammatory demy- 85. Goriely S, Neurath MF, Goldman M (2008) How microorgan-
elinating neuropathy. Brain Behav Immun 21:699–710 isms tip the balance between interleukin-12 family members.
70. Campana WM, Li X, Shubayev VI, Angert M, Cai K, Myers RR Nat Rev Immunol 8:81–86
(2006) Erythropoietin reduces Schwann cell TNF-alpha, Wal- 86. Touil T, Fitzgerald D, Zhang GX, Rostami AM, Gran B (2006)
lerian degeneration and pain-related behaviors after peripheral Pathophysiology of interleukin-23 in experimental autoimmune
nerve injury. Eur J Neurosci 23:617–626 encephalomyelitis. Drug News Perspect 19:77–83
71. Creange A, Belec L, Clair B, Raphael JC, Gherardi RK (1996) 87. Hu W, Dehmel T, Pirhonen J, Hartung HP, Kieseier BC (2006)
Circulating tumor necrosis factor (TNF)-alpha and soluble TNF- Interleukin 23 in acute inflammatory demyelination of the
alpha receptors in patients with Guillain–Barre syndrome. peripheral nerve. Arch Neurol 63:858–864
J Neuroimmunol 68:95–99 88. Yoshida H, Nakaya M, Miyazaki Y (2009) Interleukin 27: a
72. Weishaupt A, Gold R, Hartung T, Gaupp S, Wendel A, Bruck W, double-edged sword for offense and defense. J Leukoc Biol
Toyka KV (2000) Role of TNF-alpha in high-dose antigen 86:1295–1303
therapy in experimental autoimmune neuritis: inhibition of TNF- 89. Gee K, Guzzo C, Che Mat NF, Ma W, Kumar A (2009) The
alpha by neutralizing antibodies reduces T-cell apoptosis and IL-12 family of cytokines in infection, inflammation and auto-
prevents liver necrosis. J Neuropathol Exp Neurol 59:368–376 immune disorders. Inflamm Allergy Drug Targets 8:40–52
73. Lu MO, Duan RS, Quezada HC, Chen ZG, Mix E, Jin T, Yang 90. Armati PJ, Pollard JD (1996) Immunology of the Schwann cell.
X, Ljunggren HG, Zhu J (2007) Aggravation of experimental Baillieres Clin Neurol 5:47–64
autoimmune neuritis in TNF-alpha receptor 1 deficient mice. 91. Conti G, De Pol A, Scarpini E, Vaccina F, De Riz M, Baron P,
J Neuroimmunol 186:19–26 Tiriticco M, Scarlato G (2002) Interleukin-1 beta and interferon-
74. Lu MO, Zhang XM, Mix E, Quezada HC, Jin T, Zhu J, Adem A gamma induce proliferation and apoptosis in cultured Schwann
(2008) TNF-alpha receptor 1 deficiency enhances kainic acid- cells. J Neuroimmunol 124:29–35
induced hippocampal injury in mice. J Neurosci Res 86:1608– 92. Wolf G, Gabay E, Tal M, Yirmiya R, Shavit Y (2006) Genetic
1614 impairment of interleukin-1 signaling attenuates neuropathic
75. van Oosten BW, Barkhof F, Truyen L, Boringa JB, Bertelsmann pain, autotomy, and spontaneous ectopic neuronal activity, fol-
FW, von Blomberg BM, Woody JN, Hartung HP, Polman CH lowing nerve injury in mice. Pain 120:315–324
(1996) Increased MRI activity and immune activation in two 93. Hayashi R, Xiao W, Kawamoto M, Yuge O, Bennett GJ (2008)
multiple sclerosis patients treated with the monoclonal anti- Systemic glucocorticoid therapy reduces pain and the number of
tumor necrosis factor antibody cA2. Neurology 47:1531–1534 endoneurial tumor necrosis factor-alpha (TNFalpha)-positive
76. Chen X, Baumel M, Mannel DN, Howard OM, Oppenheim JJ mast cells in rats with a painful peripheral neuropathy. J Phar-
(2007) Interaction of TNF with TNF receptor type 2 promotes macol Sci 106:559–565
expansion and function of mouse CD4?CD25? T regulatory 94. Sivieri S, Ferrarini AM, Lolli F, Mata S, Pinto F, Tavolato B,
cells. J Immunol 179:154–161 Gallo P (1997) Cytokine pattern in the cerebrospinal fluid from
77. Chen X, Oppenheim JJ (2010) TNF-alpha: an activator of patients with GBS and CIDP. J Neurol Sci 147:93–95
CD4?FoxP3?TNFR2? regulatory T cells. Curr Dir Autoim- 95. Hartung HP, Reiners K, Schmidt B, Stoll G, Toyka KV (1991)
mun 11:119–134 Serum interleukin-2 concentrations in Guillain–Barre syndrome
78. Nagar M, Jacob-Hirsch J, Vernitsky H, Berkun Y, Ben-Horin S, and chronic idiopathic demyelinating polyradiculoneuropathy:
Amariglio N, Bank I, Kloog Y, Rechavi G, Goldstein I (2010) comparison with other neurological diseases of presumed im-
TNF activates a NF-kappaB-regulated cellular program in munopathogenesis. Ann Neurol 30:48–53
human CD45RA-regulatory T cells that modulates their sup- 96. Felderhoff-Mueser U, Schmidt OI, Oberholzer A, Buhrer C,
pressive function. J Immunol 184:3570–3581 Stahel PF (2005) IL-18: a key player in neuroinflammation and
79. Del Vecchio M, Bajetta E, Canova S, Lotze MT, Wesa A, Par- neurodegeneration? Trends Neurosci 28:487–493
miani G, Anichini A (2007) Interleukin-12: biological properties 97. Nakanishi K, Yoshimoto T, Tsutsui H, Okamura H (2001)
and clinical application. Clin Cancer Res 13:4677–4685 Interleukin-18 regulates both Th1 and Th2 responses. Annu Rev
80. Brahmachari S, Pahan K (2009) Suppression of regulatory T Immunol 19:423–474
cells by IL-12p40 homodimer via nitric oxide. J Immunol 98. Jander S, Stoll G (2001) Interleukin-18 is induced in acute
183:2045–2058 inflammatory demyelinating polyneuropathy. J Neuroimmunol
81. Calida DM, Kremlev SG, Fujioka T, Hilliard B, Ventura E, 114:253–258
Constantinescu CS, Lavi E, Rostami A (2000) Experimental 99. Duan RS, Zhang XM, Mix E, Quezada HC, Adem A, Zhu J
allergic neuritis in the SJL/J mouse: induction of severe and (2007) IL-18 deficiency inhibits both Th1 and Th2 cytokine
reproducible disease with bovine peripheral nerve myelin and production but not the clinical symptoms in experimental
pertussis toxin with or without interleukin-12. J Neuroimmunol autoimmune neuritis. J Neuroimmunol 183:162–167
107:1–7 100. Kallen KJ (2002) The role of transsignalling via the agonistic
82. Pelidou SH, Zou LP, Deretzi G, Nennesmo I, Wei L, Mix E, soluble IL-6 receptor in human diseases. Biochim Biophys Acta
Van Der Meide PH, Zhu J (2000) Intranasal administration of 1592:323–343

123
546 J Neurol (2011) 258:533–548

101. Tofaris GK, Patterson PH, Jessen KR, Mirsky R (2002) Shi FD (2008) IL-21 modulates CD4?CD25? regulatory T-cell
Denervated Schwann cells attract macrophages by secretion of homeostasis in experimental autoimmune encephalomyelitis.
leukemia inhibitory factor (LIF) and monocyte chemoattractant Scand J Immunol 67:37–46
protein-1 in a process regulated by interleukin-6 and LIF. 121. Spolski R, Kim HP, Zhu W, Levy DE, Leonard WJ (2009) IL-21
J Neurosci 22:6696–6703 mediates suppressive effects via its induction of IL-10.
102. Zhu J, Link H, Weerth S, Linington C, Mix E, Qiao J (1994) The J Immunol 182:2859–2867
B cell repertoire in experimental allergic neuritis involves 122. Li MO, Flavell RA (2008) TGF-beta: a master of all T cell
multiple myelin proteins and GM1. J Neurol Sci 125:132–137 trades. Cell 134:392–404
103. Dominitzki S, Fantini MC, Neufert C, Nikolaev A, Galle PR, 123. Horwitz DA (2006) Transforming growth factor-beta: taking
Scheller J, Monteleone G, Rose-John S, Neurath MF, Becker C control of T cells’ life and death. Immunity 25:399–401
(2007) Cutting edge: trans-signaling via the soluble IL-6R 124. Li MO, Wan YY, Sanjabi S, Robertson AK, Flavell RA (2006)
abrogates the induction of FoxP3 in naive CD4?CD25 T cells. Transforming growth factor-beta regulation of immune respon-
J Immunol 179:2041–2045 ses. Annu Rev Immunol 24:99–146
104. Oukka M (2007) Interplay between pathogenic Th17 and regu- 125. Xiao BG, Zhu WH, Lu CZ (2007) The presence of GM-CSF and
latory T cells. Ann Rheum Dis 66(Suppl 3):iii87–iii90 IL-4 interferes with effect of TGF-beta1 on antigen presenting
105. Uceyler N, Sommer C (2008) Cytokine regulation in animal cells in patients with multiple sclerosis and in rats with exper-
models of neuropathic pain and in human diseases. Neurosci imental autoimmune encephalomyelitis. Cell Immunol
Lett 437:194–198 249:30–36
106. Weller M, Stevens A, Sommer N, Melms A, Dichgans J, Wie- 126. Laouar Y, Town T, Jeng D, Tran E, Wan Y, Kuchroo VK,
tholter H (1991) Comparative analysis of cytokine patterns in Flavell RA (2008) TGF-beta signaling in dendritic cells is a
immunological, infectious, and oncological neurological disor- prerequisite for the control of autoimmune encephalomyelitis.
ders. J Neurol Sci 104:215–221 Proc Natl Acad Sci USA 105:10865–10870
107. Deretzi G, Pelidou SH, Zou LP, Quiding C, Zhu J (1999) Local 127. Lu LY, Chu JJ, Lu PJ, Sung PK, Hsu CM, Tseng JC (2008)
effects of recombinant rat interleukin-6 on the peripheral ner- Expression of intracellular transforming growth factor-beta1 in
vous system. Immunology 97:582–587 CD4?CD25? cells in patients with systemic lupus erythema-
108. Choy DS, Black W, April R, Goldfinger G (2008) The Black– tosus. J Microbiol Immunol Infect 41:165–173
Choy sign. Photomed Laser Surg 26:273 128. Veldhoen M, Hocking RJ, Flavell RA, Stockinger B (2006)
109. Nishimoto N, Kishimoto T (2008) Humanized antihuman IL-6 Signals mediated by transforming growth factor-beta initiate
receptor antibody, tocilizumab. Handb Exp Pharmacol autoimmune encephalomyelitis, but chronic inflammation is
181:151–160 needed to sustain disease. Nat Immunol 7:1151–1156
110. Rose-John S, Waetzig GH, Scheller J, Grotzinger J, Seegert D 129. Manel N, Unutmaz D, Littman DR (2008) The differentiation of
(2007) The IL-6/sIL-6R complex as a novel target for thera- human T(H)-17 cells requires transforming growth factor-beta
peutic approaches. Expert Opin Ther Targets 11:613–624 and induction of the nuclear receptor RORgammat. Nat Immu-
111. Andriambeloson E, Baillet C, Vitte PA, Garotta G, Dreano M, nol 9:641–649
Callizot N (2006) Interleukin-6 attenuates the development of 130. Yang L, Anderson DE, Baecher-Allan C, Hastings WD, Bettelli
experimental diabetes-related neuropathy. Neuropathology E, Oukka M, Kuchroo VK, Hafler DA (2008) IL-21 and TGF-
26:32–42 beta are required for differentiation of human T(H)17 cells.
112. Ito T, Ikeda K, Tomita K, Yokoyama S (2010) Interleukin-6 Nature 454:350–352
upregulates the expression of PMP22 in cultured rat Schwann 131. Creange A, Belec L, Clair B, Degos JD, Raphael JC, Gherardi
cells via a JAK2-dependent pathway. Neurosci Lett 472:104–108 RK (1998) Circulating transforming growth factor beta 1 (TGF-
113. Lara-Ramirez R, Segura-Anaya E, Martinez-Gomez A, Dent beta1) in Guillain–Barre syndrome: decreased concentrations in
MA (2008) Expression of interleukin-6 receptor alpha in normal the early course and increase with motor function. J Neurol
and injured rat sciatic nerve. Neuroscience 152:601–608 Neurosurg Psychiatry 64:162–165
114. Knupfer H, Preiss R (2008) sIL-6R: more than an agonist? 132. Ossege LM, Sindern E, Voss B, Malin JP (2000) Expression of
Immunol Cell Biol 86:87–91 TNFalpha and TGFbeta1 in Guillain–Barre syndrome: correla-
115. Kunz M, Ibrahim SM (2009) Cytokines and cytokine profiles in tion of a low TNFalpha-/TGFbeta1-mRNA ratio with good
human autoimmune diseases and animal models of autoimmu- recovery and signs for immunoregulation within the cerebro-
nity. Mediators Inflamm 2009:979258 spinal fluid compartment. Eur J Neurol 7:17–25
116. Yu JJ, Gaffen SL (2008) Interleukin-17: a novel inflammatory 133. Zhu J, Mix E, Olsson T, Link H (1994) Cellular mRNA
cytokine that bridges innate and adaptive immunity. Front expression of interferon-gamma, IL-4 and transforming growth
Biosci 13:170–177 factor-beta (TGF-beta) by rat mononuclear cells stimulated with
117. Zrioual S, Ecochard R, Tournadre A, Lenief V, Cazalis MA, peripheral nerve myelin antigens in experimental allergic neu-
Miossec P (2009) Genome-wide comparison between IL-17A- ritis. Clin Exp Immunol 98:306–312
and IL-17F-induced effects in human rheumatoid arthritis 134. Dahle C, Kvarnstrom M, Ekerfelt C, Samuelsson M, Ernerudh J
synoviocytes. J Immunol 182:3112–3120 (2003) Elevated number of cells secreting transforming growth
118. Doreau A, Belot A, Bastid J, Riche B, Trescol-Biemont MC, factor beta in Guillain–Barre syndrome. APMIS 111:1095–1104
Ranchin B, Fabien N, Cochat P, Pouteil-Noble C, Trolliet P, 135. Sindern E, Schweppe K, Ossege LM, Malin JP (1996) Potential
Durieu I, Tebib J, Kassai B, Ansieau S, Puisieux A, Eliaou JF, role of transforming growth factor-beta 1 in terminating the
Bonnefoy-Berard N (2009) Interleukin 17 acts in synergy with B immune response in patients with Guillain–Barre syndrome.
cell-activating factor to influence B cell biology and the path- J Neurol 243:264–268
ophysiology of systemic lupus erythematosus. Nat Immunol 136. O’Garra A, Barrat FJ, Castro AG, Vicari A, Hawrylowicz C
10:778–785 (2008) Strategies for use of IL-10 or its antagonists in human
119. Segal BM (2010) Th17 cells in autoimmune demyelinating disease. Immunol Rev 223:114–131
disease. Semin Immunopathol 32:71–77 137. Saraiva M, Christensen JR, Veldhoen M, Murphy TL, Murphy
120. Piao WH, Jee YH, Liu RL, Coons SW, Kala M, Collins M, KM, O’Garra A (2009) Interleukin-10 production by Th1 cells
Young DA, Campagnolo DI, Vollmer TL, Bai XF, La Cava A, requires interleukin-12-induced STAT4 transcription factor and

123
J Neurol (2011) 258:533–548 547

ERK MAP kinase activation by high antigen dose. Immunity STAT4 and STAT6 on the induction of experimental autoim-
31:209–219 mune encephalomyelitis. J Clin Invest 108:739–747
138. Atkins S, Loescher AR, Boissonade FM, Smith KG, Occleston 155. Wurster AL, Tanaka T, Grusby MJ (2000) The biology of Stat4
N, O’Kane S, Ferguson MW, Robinson PP (2007) Interleukin- and Stat6. Oncogene 19:2577–2584
10 reduces scarring and enhances regeneration at a site of sciatic 156. Kaplan MH, Sun YL, Hoey T, Grusby MJ (1996) Impaired
nerve repair. J Peripher Nerv Syst 12:269–276 IL-12 responses and enhanced development of Th2 cells in
139. Milligan ED, Sloane EM, Watkins LR (2008) Glia in patho- Stat4-deficient mice. Nature 382:174–177
logical pain: a role for fractalkine. J Neuroimmunol 157. O’Sullivan LA, Liongue C, Lewis RS, Stephenson SE, Ward AC
198:113–120 (2007) Cytokine receptor signaling through the Jak-Stat-Socs
140. Ledeboer A, Jekich BM, Sloane EM, Mahoney JH, Langer SJ, pathway in disease. Mol Immunol 44:2497–2506
Milligan ED, Martin D, Maier SF, Johnson KW, Leinwand LA, 158. Fujimoto M, Tsutsui H, Xinshou O, Tokumoto M, Watanabe D,
Chavez RA, Watkins LR (2007) Intrathecal interleukin-10 gene Shima Y, Yoshimoto T, Hirakata H, Kawase I, Nakanishi K,
therapy attenuates paclitaxel-induced mechanical allodynia and Kishimoto T, Naka T (2004) Inadequate induction of suppressor
proinflammatory cytokine expression in dorsal root ganglia in of cytokine signaling-1 causes systemic autoimmune diseases.
rats. Brain Behav Immun 21:686–698 Int Immunol 16:303–314
141. Stern JN, Keskin DB, Zhang H, Lv H, Kato Z, Strominger JL 159. Egan PJ, Lawlor KE, Alexander WS, Wicks IP (2003) Sup-
(2008) Amino acid copolymer-specific IL-10-secreting regula- pressor of cytokine signaling-1 regulates acute inflammatory
tory T cells that ameliorate autoimmune diseases in mice. Proc arthritis and T cell activation. J Clin Invest 111:915–924
Natl Acad Sci USA 105:5172–5176 160. Malemud CJ, Miller AH (2008) Pro-inflammatory cytokine-
142. Myhr KM, Vagnes KS, Maroy TH, Aarseth JH, Nyland HI, induced SAPK/MAPK and JAK/STAT in rheumatoid arthritis
Vedeler CA (2003) Interleukin-10 promoter polymorphisms in and the new anti-depression drugs. Expert Opin Ther Targets
patients with Guillain–Barre syndrome. J Neuroimmunol 12:171–183
139:81–83 161. Roman-Blas JA, Jimenez SA (2008) Targeting NF-kappaB: a
143. Yun W, Hua-bing W, Wei-zhi W (2007) A study of associated promising molecular therapy in inflammatory arthritis. Int Rev
cell-mediated immune mechanisms in experimental autoim- Immunol 27:351–374
mune neuritis rats. J Neuroimmunol 185:87–94 162. Mangano K, Dati G, Quattrocchi C, Proietti L, Mazzarino C, Di
144. Dahle C, Ekerfelt C, Vrethem M, Samuelsson M, Ernerudh J Marco R, Bendtzen K, Greco B, Zaratin P, Nicoletti F (2008)
(1997) T helper type 2 like cytokine responses to peptides from Preventive and curative effects of cyclophosphamide in an
P0 and P2 myelin proteins during the recovery phase of Guil- animal model of Guillain–Barre syndrome. J Neuroimmunol
lain–Barre syndrome. J Neurol Sci 153:54–60 196:107–115
145. Oppenheim JJ, Zachariae CO, Mukaida N, Matsushima K 163. Brereton CF, Sutton CE, Lalor SJ, Lavelle EC, Mills KH (2009)
(1991) Properties of the novel proinflammatory supergene ‘‘in- Inhibition of ERK MAPK suppresses IL-23- and IL-1-driven IL-
tercrine’’ cytokine family. Annu Rev Immunol 9:617–648 17 production and attenuates autoimmune disease. J Immunol
146. Ransohoff RM, Glabinski A, Tani M (1996) Chemokines in 183:1715–1723
immune-mediated inflammation of the central nervous system. 164. Wu T, Mohan C (2009) The AKT axis as a therapeutic target in
Cytokine Growth Factor Rev 7:35–46 autoimmune diseases. Endocr Metab Immune Disord Drug
147. Luster AD (1998) Chemokines–chemotactic cytokines that Targets 9:145–150
mediate inflammation. N Engl J Med 338:436–445 165. Peifer C, Wagner G, Laufer S (2006) New approaches to the
148. Zou LP, Pelidou SH, Abbas N, Deretzi G, Mix E, Schaltzbeerg treatment of inflammatory disorders small molecule inhibitors of
M, Winblad B, Zhu J (1999) Dynamics of production of MIP- p38 MAP kinase. Curr Top Med Chem 6:113–149
1alpha, MCP-1 and MIP-2 and potential role of neutralization of 166. Ding C, Xu J, Li J (2008) ABT-874, a fully human monoclonal
these chemokines in the regulation of immune responses during anti-IL-12/IL-23 antibody for the potential treatment of auto-
experimental autoimmune neuritis in Lewis rats. J Neuroimmu- immune diseases. Curr Opin Investig Drugs 9:515–522
nol 98:168–175 167. Billich A (2007) Drug evaluation: apilimod, an oral IL-12/IL-23
149. Orlikowski D, Chazaud B, Plonquet A, Poron F, Sharshar T, inhibitor for the treatment of autoimmune diseases and common
Maison P, Raphael JC, Gherardi RK, Creange A (2003) variable immunodeficiency. IDrugs 10:53–59
Monocyte chemoattractant protein 1 and chemokine receptor 168. Wittig BM (2007) Drug evaluation: CNTO-1275, a mAb against
CCR2 productions in Guillain–Barre syndrome and experi- IL-12/IL-23p40 for the potential treatment of inflammatory
mental autoimmune neuritis. J Neuroimmunol 134:118–127 diseases. Curr Opin Investig Drugs 8:947–954
150. Kieseier BC, Tani M, Mahad D, Oka N, Ho T, Woodroofe N, 169. Badovinac VP, Tvinnereim AR, Harty JT (2000) Regulation of
Griffin JW, Toyka KV, Ransohoff RM, Hartung HP (2002) antigen-specific CD8? T cell homeostasis by perforin and
Chemokines and chemokine receptors in inflammatory demye- interferon-gamma. Science 290:1354–1358
linating neuropathies: a central role for IP-10. Brain 125:823– 170. Hassan AT, Dai Z, Konieczny BT, Ring GH, Baddoura FK,
834 Abou-Dahab LH, El-Sayed AA, Lakkis FG (1999) Regulation of
151. Fujioka T, Kolson DL, Rostami AM (1999) Chemokines and alloantigen-mediated T-cell proliferation by endogenous inter-
peripheral nerve demyelination. J Neurovirol 5:27–31 feron-gamma: implications for long-term allograft acceptance.
152. Press R, Pashenkov M, Jin JP, Link H (2003) Aberrated levels of Transplantation 68:124–129
cerebrospinal fluid chemokines in Guillain–Barre syndrome and 171. Skurkovich S, Boiko A, Beliaeva I, Buglak A, Alekseeva T,
chronic inflammatory demyelinating polyradiculoneuropathy. Smirnova N, Kulakova O, Tchechonin V, Gurova O, Deomina
J Clin Immunol 23:259–267 T, Favorova OO, Skurkovic B, Gusev E (2001) Randomized
153. Luongo L, Sajic M, Grist J, Clark AK, Maione S, Malcangio M study of antibodies to IFN-gamma and TNF-alpha in secondary
(2008) Spinal changes associated with mechanical hypersensi- progressive multiple sclerosis. Mult Scler 7:277–284
tivity in a model of Guillain–Barre syndrome. Neurosci Lett 172. Skurkovich B, Skurkovich S (2007) Autoimmune diseases are
437:98–102 connected with disturbances in cytokine synthesis, and therapy
154. Chitnis T, Najafian N, Benou C, Salama AD, Grusby MJ, Say- with IFN-gamma blockers is their main pathogenetic treatment.
egh MH, Khoury SJ (2001) Effect of targeted disruption of Ann N Y Acad Sci 1109:167–177

123
548 J Neurol (2011) 258:533–548

173. Csurhes PA, Sullivan AA, Green K, Pender MP, McCombe PA inflammatory cells into the peripheral nerve tissue. Neurophar-
(2005) T cell reactivity to P0, P2, PMP-22, and myelin basic macology 42:731–739
protein in patients with Guillain–Barre syndrome and chronic 177. Zhang Z, Zhang ZY, Fauser U, Schluesener HJ (2008) Valproic
inflammatory demyelinating polyradiculoneuropathy. J Neurol acid attenuates inflammation in experimental autoimmune neu-
Neurosurg Psychiatry 76:1431–1439 ritis. Cell Mol Life Sci 65:4055–4065
174. Abbas N, Zou LP, Pelidou SH, Winblad B, Zhu J (2000) Pro- 178. Zhang ZY, Zhang Z, Fauser U, Schluesener HJ (2009) Improved
tective effect of Rolipram in experimental autoimmune neuritis: outcome of EAN, an animal model of GBS, through ameliora-
protection is associated with down-regulation of IFN-gamma tion of peripheral and central inflammation by minocycline.
and inflammatory chemokines as well as up-regulation of IL-4 J Cell Mol Med 13:341–351
in peripheral nervous system. Autoimmunity 32:93–99 179. Zhang Z, Zhang ZY, Schluesener HJ (2009) Compound A, a
175. Zou LP, Deretzi G, Pelidou SH, Levi M, Wahren B, Quiding C, plant origin ligand of glucocorticoid receptors, increases regu-
van der Meide P, Zhu J (2000) Rolipram suppresses experi- latory T cells and M2 macrophages to attenuate experimental
mental autoimmune neuritis and prevents relapses in Lewis rats. autoimmune neuritis with reduced side effects. J Immunol
Neuropharmacology 39:324–333 183:3081–3091
176. Zou LP, Abbas N, Volkmann I, Nennesmo I, Levi M, Wahren B, 180. Zhang ZY, Zhang Z, Schluesener HJ (2010) MS-275, an histone
Winblad B, Hedlund G, Zhu J (2002) Suppression of experi- deacetylase inhibitor, reduces the inflammatory reaction in rat
mental autoimmune neuritis by ABR-215062 is associated experimental autoimmune neuritis. Neuroscience 169(1):370–
with altered Th1/Th2 balance and inhibited migration of 377

123

You might also like