YEXNR-11682; No. of pages: 10; 4C: Experimental Neurology xxx (2014) xxx–xxx

Contents lists available at ScienceDirect

Experimental Neurology

1

Review

2

Th17 cells in central nervous system autoimmunity

3Q1

Christopher Sie a, Thomas Korn a,b,⁎, Meike Mitsdoerffer a,b,⁎

4 5

a

6

a r t i c l e

7 8 9 10 11

Article history: Received 12 January 2014 Revised 12 March 2014 Accepted 19 March 2014 Available online xxxx

12 13 14 15 25

Keywords: Multiple sclerosis Experimental autoimmune encephalomyelitis Th17 cells

a b s t r a c t

R O

i n f o

D

P

Multiple sclerosis (MS) is the most important autoimmune disease of the central nervous system (CNS). Its animal model experimental autoimmune encephalomyelitis (EAE) has been instrumental in defining the features of the novel T helper cell subset Th17. Conversely, the broad characterization of Th17 immune responses has substantially advanced our understanding of organ-specific autoimmunity and inspired almost a decade of immunological research. Here, we review the current knowledge on Th17 cells and their contribution to the immunopathology in EAE and MS, covering recent proceedings in the induction, modulation and effector mechanisms of this versatile T lymphocyte subset. In particular, we discuss the emerging role of mucosal immunity in the regulation of Th17 cells and CNS autoimmunity, the accumulating evidence for extensive plasticity in the Th17 subset, and their molecular mode of action in promoting this debilitating disease. © 2014 Published by Elsevier Inc.

29 27 26

. . . . . . . . . . . .

R

44

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

E

. . . . . . . . . . . .

R

Introduction . . . . . . . . . . . . . . . . . . . Induction of Th17 cells . . . . . . . . . . . . . . Role of mucosal tissues in the induction of Th17 cells Trafficking of Th17 to CNS tissues . . . . . . . . . Plasticity of Th17 cells . . . . . . . . . . . . . . Th17 effector functions . . . . . . . . . . . . . . Regulation of Th17 responses . . . . . . . . . . . Th17 cells in multiple sclerosis . . . . . . . . . . . Concluding remarks . . . . . . . . . . . . . . . Conflict of interest . . . . . . . . . . . . . . . . Acknowledgments . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . .

C

Contents

N C O

32 33 34 35 36 37 38 39 40 41 42 43

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

Introduction

46

Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system (CNS). Decades of research have established the hallmark that CNS-infiltrating T lymphocytes are among the major mediators of this debilitating disease in many experimental settings (Fletcher et al., 2010) and recent genome-wide association studies (GWAS) strongly support the notion that pathways in T cell activation

49 50 51

U

45

47 48

16 17 18 19 20 21 22 23 24

T

28 31 30

O

Klinikum rechts der Isar, Technische Universität München, Department of Neurology, Ismaninger Str. 22, 81675 Munich, Germany Munich Cluster for Systems Neurology (SyNergy), Munich, Germany

E

b

F

journal homepage: www.elsevier.com/locate/yexnr

⁎ Corresponding authors at: Klinikum rechts der Isar, Department of Neurology, Ismaninger Str. 22, 81675 Munich, Germany. Fax: +49 89 4140 4675. E-mail addresses: [email protected] (T. Korn), [email protected] (M. Mitsdoerffer).

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

. . . . . . . . . . . .

0 0 0 0 0 0 0 0 0 0 0 0

and differentiation are relevant to the human pathology (Sawcer et al., 2011). A large body of evidence for the role of T cells in CNS autoimmunity comes from studies in rodent models of MS, with experimental autoimmune encephalomyelitis (EAE) being the most prominent approach for more than 70 years (Gold et al., 2006; Rangachari and Kuchroo, 2013; Zamvil and Steinman, 1990). In its classical induction regime, susceptible mouse strains are immunized by subcutaneous injection of CNS antigen emulsified in Complete Freund's Adjuvant (CFA). T lymphocytes primed in the peripheral compartment eventually infiltrate into the CNS. Importantly, EAE can also be induced passively by transferring autoaggressive CD4+ T lymphocytes into naïve recipients (Ben-Nun et al., 1981; Schluesener and Wekerle, 1985), highlighting the significance of T cells as mediators of the disease.

http://dx.doi.org/10.1016/j.expneurol.2014.03.009 0014-4886/© 2014 Published by Elsevier Inc.

Please cite this article as: Sie, C., et al., Th17 cells in central nervous system autoimmunity, Exp. Neurol. (2014), http://dx.doi.org/10.1016/ j.expneurol.2014.03.009

52 53 54 55 56 57 58 59 60 61 62 63 64 65

91 92 93 94 95 96 97 98 99 100 101 102 103 104 105 106 107

113 114 115 116 117 118 119 120 121 122 123 124 125 126 127 128 129

C

89 90

E

87 88

R

85 86

R

83 84

O

81 82

C

79 80

N

77 78

U

75 76

Role of mucosal tissues in the induction of Th17 cells

146

F

The cytokine milieu provided by antigen-presenting and bystander cells during T cell priming is dominant over other signals including antigen dose and costimulatory molecules in the commitment of naïve T cells to distinct T helper cell subsets. Emulating this microenvironment by stimulating T cells in vitro in the presence of exogenous cytokines has been instrumental in defining the molecular cues for T cell differentiation and in identifying core features of many T cell subsets. While the cytokine IL-23 was initially considered a differentiation factor for Th17 cells (Langrish et al., 2005), its capacity to induce IL-17 in naïve T cells was virtually absent and indeed, the IL-23R is not expressed on naïve T lymphocytes (Parham et al., 2002). By serendipity, it was found that the combination of TGFβ1 and IL-6 could drive the generation of Th17 cells and their expression of IL23R (Bettelli et al., 2006; Mangan et al., 2006; Veldhoen et al., 2006), a process that was amplified by the auto- and paracrine effect of IL-21, produced by Th17 cells themselves (Korn et al., 2007; Nurieva et al., 2007; Zhou et al., 2007). Th17 cells express IL-23R and IL-23 is paramount to the pathological function of CD4+ T helper cells which has been shown in many paradigms of T cell mediated inflammatory diseases (Croxford et al.,

73 74

O

111 112

72

R O

Induction of Th17 cells

70 71

130 131

In the past few years, mucosal tissues and their local immune compartments, i.e. mucosa-associated lymphoid tissues (MALT), have received a special attention with regard to their influence on T cell immunity. Mucosal surfaces of the gut and lung are in constant interaction with a plethora of microbiota, pathogens and environmental factors. Therefore, they are of dual interest for MS and hence EAE pathology: first, they provide a means to explain the enigmatic environmental component in MS etiology and relapse triggering. Second, if there is a phase in which T cell responses are naturally modulated in these compartments, this opens a therapeutic window for pharmaceutical intervention by oral or aerosol medication. The lung mucosa is continuously exposed to environmental influences and has only recently been implicated in CNS autoimmunity. Odoardi et al. (2012) used an adoptive transfer model of EAE in rats to track myelin-specific, GFP+ T cells on their way to the central nervous system (Odoardi et al., 2012). Surprisingly, these encephalitogenic T cells did not only briefly pass through the lung's capillary network upon intravenous transfer but penetrated deep into the parenchyma, specifically homing to bronchus-associated lymphoid tissues (BALT) as well as bronchial structures. Here, they resided for several hours, actively scanning the mucosal surface of the bronchiolar lumen, before continuing into the periphery and infiltrating the CNS. Importantly, in addition to these observations from passive EAE induction regimes with T cells pre-activated ex vivo, bronchiole- and BALT-associated T cells were also found in actively immunized animals that had previously received resting GFP+ T cells. While the cultured memory T lymphocytes used in this model are known to have a mixed phenotype of IL-17/IFN-γ double producers (Bartholomäus et al., 2009), their intriguing accumulation and behavior in pulmonary structures could allude to a general scheme in activated T cells. However, whether this holds true for Th17 cells in mice and, more importantly, men remains to be discovered. For the gastrointestinal tract, a number of studies have implicated microbiota and nutritional factors in the modulation of Th17 responses and CNS autoimmunity (Berer and Krishnamoorthy, 2012). Th17 cells are abundant in the gut-associated lymphoid tissue (GALT) of mice colonized under normal, specific-pathogen free (SPF) conditions (Ivanov et al., 2006). However, mice treated with antibiotics (Ivanov et al., 2008) or hosted in completely pathogen-free environments failed to generate Th17 cells in the lamina propria, a feature that could be restored by re-colonizing the mice with specific commensal species, notably segmented filamentous bacteria (SFB) among others (GaboriauRouthiau et al., 2009; Ivanov et al., 2008, 2009). A possible mediator of this effect was commensal-derived adenosine triphosphate that induced the production of the Th17 differentiating cytokines IL-6 and IL-23 in cells of the lamina propria (Atarashi et al., 2008). Given their strong influence on local Th17 cells in the GALT, the impact of intestinal microbiota on Th17-driven diseases, notably EAE, was

P

110

68 69

2012; McGeachy et al., 2009). It remains to be determined whether other subsets of T helper cells are also IL-23R positive. However, in contrast to Th17 cells, Th1 cells induced with IL-12 do not express IL-23R. An alternative way to induce Th17 cells in vitro was reported to occur in the presence of IL-1β (and independently of TGF-β). In fact, IL-1β together with IL-6 and IL-23 were described to induce a “pathogenic” subset of Th17 cells that in addition to ROR-γt also expressed T-bet and lacks IL-10 production (Ghoreschi et al., 2010). These findings have spurred the concept of different “flavors” of Th17 cells (Lee et al., 2012). It is well possible that different Th17 phenotypes are defined by the biological niche in which they are raised. It was found that CD103− dendritic cells (DCs) could prime Th17 cells independently of IL-6 in the spleen while IL-6 was critical for the induction of Th17 cells in skin and mucosa, where regulating CD103+ DCs that produced large amounts of TGFβ and retinoic acid dictated the requirement for IL-6 in Th17 priming (Hu et al., 2011).

T

108 109

A key concept in our understanding of T cell biology is that the outcome of an unfolding adaptive immune response is largely defined by the phenotype of its constituting T helper (Th) cells. This in turn is determined by the microenvironmental signals integrated in naïve T cells during their initial encounter with a cognate antigen in a sequence of events subsumed as T cell priming. In the course of this process, T helper cells were originally believed to commit to one of two possible and mutually exclusive states of differentiation, coined Th1 and Th2 in a seminal paper by Mosmann et al. (1986). These two subsets were distinguished by means of their signature cytokine secretion, i.e. IFN-γ and IL-4 for Th1 and Th2 cells, respectively. Given the inflammatory nature of Th1 cytokines and their presence in MS lesions (Traugott and Lebon, 1988a, 1988b), Th1 cells were considered to solely drive the disease pathology in EAE. Initially, this concept was supported by the fact that gene targeted deletion of the p40 subunit of IL-12, a major Th1 differentiation factor, prevented the onset of EAE (Segal et al., 1998). However, other studies revealed that mice with genetic ablation of the Th1 signature cytokine IFN-γ (Ferber et al., 1996) or its receptor (Willenborg et al., 1996) as well as the IL12 subunit p35 (Becher et al., 2002; Gran et al., 2002) or its receptor IL-12Rβ2 (Zhang et al., 2003) were not only still susceptible to EAE but in some cases even presented with exacerbated rather than alleviated disease course. Therefore, the original Th1 paradigm could not exclusively account for the T cell driven EAE pathology observed in mice. The key to reconciling the concept of distinct T helper subsets with experimental evidence came with the notion that mice devoid of IL-23, a cytokine that shares the p40 subunit with IL-12 but constitutes a distinct molecule using p19 rather than p35 (Oppmann et al., 2000), were completely resistant to EAE (Cua et al., 2003). Prominently expanding the long standing dichotomy of Th1 and Th2 cells, it was then realized that IL-23 is implicated in the maintenance of a novel T cell subset, identified by its signature cytokine IL-17 as Th17 cells (Harrington et al., 2005; Langrish et al., 2005). Following this important discovery, it was soon appreciated that many organ-specific autoimmune diseases were not, as previously perceived, solely driven by Th1 cells but could also exhibit a relevant, if not decisive contribution of Th17 cells (Dardalhon et al., 2008). Here, we review the current knowledge on Th17 cells with a special focus on their role in CNS autoimmunity. While accumulating evidence suggests that T cell phenotypes are not limited to set profiles but can also exhibit a considerable degree of plasticity (Hirahara et al., 2013), the given Th nomenclature remains to provide a valuable conceptual framework that will be used throughout this review, bearing in mind that the described subsets likely represent only limiting cases of a highly dynamic system.

D

66 67

C. Sie et al. / Experimental Neurology xxx (2014) xxx–xxx

E

2

Please cite this article as: Sie, C., et al., Th17 cells in central nervous system autoimmunity, Exp. Neurol. (2014), http://dx.doi.org/10.1016/ j.expneurol.2014.03.009

132 133 134 135 136 137 138 139 140 141 142 143 144 145

147 148 149 150 151 152 153 154 155 156 157 158 159 160 161 162 163 164 165 166 167 168 169 170 171 172 173 174 175 176 177 178 179 180 181 182 183 184 185 186 187 188 189 190 191 192 193

C. Sie et al. / Experimental Neurology xxx (2014) xxx–xxx

F

O

R O

213 214

P

211 212

Central nervous system Secondary lymphatic organs

D

209 210

E

207 208

Th17

APC

T

205 206

a

Lamina propria

Th17

APC

b

C

203 204

Lamina propria

c

portal vein

Lamina propria

E

201 202

R

200

of the gut and even more so the complete absence of microbiota in germ-free mice entails a severe disturbance of the whole immune system, producing extreme effects in EAE models that are not reflective of the human situation. From a mechanistic point of view, one of the important control elements for integrating these environmental signals directly within T cells is the aryl-hydrocarbon receptor (AhR). It is expressed in both Th17 and regulatory T cells, and different AhR-ligands can either enhance (Quintana et al., 2008; Veldhoen et al., 2008) or suppress Th17 induction (Quintana et al., 2008), thereby modulating the disease intensity of EAE. While environmental toxins can be a source of AhR ligands, they can also be derived from natural tryptophan metabolites of intestinal microbiota (Perdew and Babbs, 1991). Importantly, these bacteria-derived ligands as well as other immunogenic components can also enter the circulation (Henao-Mejia et al., 2012; Wikoff et al., 2009), potentially offering a way to reach and influence Th17 cells in extra-intestinal immune compartments (see also Fig. 1). The most recent evidence for the impact of mucosal immunity and nutrition comes from two studies that suggested dietary salt as a major factor in both Th17 induction and EAE pathology (Kleinewietfeld et al., 2013; Wu et al., 2013). These groups discovered that the presence

blood vessel

lymph vessel

R

198 199

N C O

196 197

addressed by several groups. In active immunization regimes, initial reports suggested no change in EAE severity in germ-free mice (Lampropoulou et al., 2008) whereas a substantial alleviation of EAE symptoms was reported by another group (Lee et al., 2011). In the latter study, EAE susceptibility could be restored by re-colonization with SFB. Conversely, other distinct commensal species (Ochoa-Repáraz et al., 2010) as well as certain probiotics (Lavasani et al., 2010) were also found to be protective in actively induced EAE. Recently, Berer and colleagues have highlighted the relevance of gut microbiota in spontaneous models of EAE (Berer et al., 2011). The disease incidence in their MOG-specific TCR transgenic mice varied substantially between different housing facilities, prompting a study to compare entirely germ-free with SPF-housed mice. While the latter had an 80% EAE incidence, those mice without intestinal microbiota were virtually resistant to disease development. Importantly, this protection could be reversed by re-colonization with gut bacteria from SPF mice even after 12 weeks of germ-free housing, whereas inoculation with SFB alone was not sufficient. The sum of these data provides strong circumstantial evidence for a potential modulation of the propensity to develop CNS autoimmunity by intestinal microbiota. However, despite diligent controls in these studies, it cannot be ruled out that a mono-colonization

APC

NaCl

Bacteria

Polycyclics

U

194 195

3

Nutrition Intestinal lumen

Modulation within the mucosa

Intestinal lumen

Modulation by auxiliary cells

Intestinal lumen

Systemic modulation

Fig. 1. Scenarios for Th17 cell modulation by the intestinal mucosa. Microbiota of the gut (lower panel) do not usually enter extra-intestinal, secondary lymphatic organs (SLO, medium panel) or even the CNS (top panel) to readily interfere with Th17 cells. Likewise, dietary salt does not dramatically raise sodium blood levels. Hence, the reported influence on encephalitogenic lymphocytes likely is exerted either directly within the mucosa associated lymphoid tissue or through intermediate effects. (a) T cells are modulated by various mechanisms within the lamina propria, thereby potentially committing to the Th17 phenotype. Nutritional or microbiota derived polycyclic aromatic hydrocarbons could directly interfere with the AhR in T cells. Similarly, other small molecules of either source could also exert an influence. Dietary salt would be sensed through SGK1 in T cells and thus favor a Th17 response. Furthermore, the local innate immune system (e.g. intraepithelial lymphocytes) can modulate T cells after interacting with the luminal bacteria. Influence on APCs by most of these mechanisms is also possible (not depicted). Subsequently, T cells could either infiltrate the CNS directly or enter the circulation with a pre-commitment to the Th17 phenotype to be unleashed in a later priming event. (b) Dendritic cells can engulf bacterial components from the intestinal lumen and then migrate into the draining lymph nodes. Although it is a well-established concept that these gut-derived DCs instruct T cells to home back to the dendritic cells' origin, it is conceivable that under certain conditions the T cells may be diverted to enter the CNS. (c) Nutrition or microbiota derived molecules could be carried into extra-intestinal lymphatic organs by convection in lymph or blood vessels, eventually influencing T cells in SLOs. Before this, an effect on lymphocytes within the liver after passing through the portal vein is also possible (not depicted).

Please cite this article as: Sie, C., et al., Th17 cells in central nervous system autoimmunity, Exp. Neurol. (2014), http://dx.doi.org/10.1016/ j.expneurol.2014.03.009

215 216 217 218 219 220 221 222 223 224 225 226 227 228 229 230 231 232 233 234 235

288

Plasticity of Th17 cells

289 290

Th1 and Th2 cells were described as inherently stable T cell subsets (Murphy et al., 1996). Together with the finding that Th1 and Th2 differentiation programs were mutually exclusive and were governed by some “master” transcription factors, this prompted the idea that T helper cell subsets could be conceptualized as “lineages”. Although the signature cytokines of Th1 and Th2 cells cross-inhibit the development of Th17 cells, and the transcriptional program of Th17 cells is absolutely dependent on the transcription factor ROR-γt, investigators were reluctant to call Th17 cells a “lineage” because these cells appeared to come

261 262

266 267 268 269 270 271 272 273 274 275 276 277 278 279 280 281 282 283 284 285

291 292 293 294 295 296 297

C

259 260

E

257 258

R

255 256

R

253 254

O

251 252

C

249 250

N

247 248

U

245 246

Th17 effector functions

332

Inflammation, demyelination and axonal damage constitute the pathological hallmarks of CNS autoimmunity. Breakdown of the blood brain barrier (BBB) with subsequent infiltration of mononuclear cells is considered an essential prerequisite for the development of autoimmune inflammatory lesions in the CNS (Kermode et al., 1990). The mechanisms of this barrier dysfunction are incompletely understood but assumed to involve a detrimental combination of distinct cytokines, chemokines and metalloproteinases produced by infiltrating as well as resident cells (Minagar and Alexander, 2003). The mode of action for Th17 cell involvement in this destructive cascade has only partially been unraveled so far. Here, we will review the role archetypal members of the Th17 cytokine profile play in CNS autoimmunity, covering the signature cytokine IL-17, the recently highlighted granulocyte macrophage colony factor (GM-CSF), and other molecules like IL-9, IL-21, and IL-22 (Korn et al., 2009; Ouyang et al., 2008). Before discussing their individual contribution, we will briefly address the composition, cellular sources and receptor distribution for these cytokines. The IL-17 family consists of six members in total, designated IL-17AF. Both, IL-17 (IL-17A) and IL-17F are produced by Th17 cells and can either form homodimers or IL-17A/IL-17F heterodimers (Korn et al., 2009). Of these, IL-17 homodimers exhibit the highest inflammatory potency (Liang et al., 2007). Apart from certain CD4+ T cells, other immune cell types such as CD8+ T cells, γδ T cells, NK cells, lymphoid tissue inducer-like cells and neutrophils produce IL-17 (Korn et al., 2009). IL-17 binds to IL-17R (IL-17RA) and IL-17RC which are both expressed on a variety of cells including astrocytes and microglia (Das Sarma et al., 2009; Kawanokuchi et al., 2008). IL-17R has a much higher affinity for IL-17 than for IL-17F (Ely et al., 2009; Liu et al., 2013).

333

F

286 287

While the entry of generic lymphocytes into the CNS is a well characterized process (Engelhardt and Ransohoff, 2012), the distinct molecular cues for Th17 cell trafficking to the CNS have not been addressed extensively. One report suggested that Th17 cells – by expressing CCR6 – used the epithelium of the choroid plexus (which expresses the CCR6 ligand CCL20) to access the subarachnoid space and operate as “pioneering” cells to initiate inflammation (Reboldi et al., 2009). We and others recently showed that Th1 and Th17 cells, albeit both capable of inducing EAE in mice, entailed diseases with different pathological and clinical features in dependence of their integrin equipment (Glatigny et al., 2011; Rothhammer et al., 2011). Specifically, transferred MOG-specific Th17 cells brought about an atypical EAE with predominating ataxia rather than paralysis, reminiscent of supraspinal lesions that were indeed preferentially triggered by the transferred Th17 cells (Jäger et al., 2009; Kroenke et al., 2008; Lees et al., 2008; Stromnes et al., 2008). This pattern of CNS infiltration by Th17 cells was mediated by the integrin LFA-1, whereas Th1 cells targeted the spinal cord in a VLA-4 dependent manner (Glatigny et al., 2011; Rothhammer et al., 2011). Therefore, in the absence of VLA-4, Th17 cells could still enter the CNS. Interestingly, natalizumab that blocks VLA-4 failed to be efficient in neuromyelitis optica patients who are believed to exhibit a Th17 cell driven disease pathology (Kleiter et al., 2012; Varrin-Doyer et al., 2012).

243 244

O

265

242

R O

Trafficking of Th17 to CNS tissues

240 241

298 299

P

264

238 239

in different flavors (see above) and seemed to be exquisitely “plastic” (see also Fig. 2). Fate tracking reporter systems have been developed to monitor Th17 cells in vivo. During EAE, about one fourth of all CD4+ T cells in the spinal cord that had previously produced IL-17, now produced IFN-γ instead. Conversely, the vast majority of all IFN-γ producers, i.e. about 70%, had previously produced IL-17 while only 30% were bona fide Th1 cells with no preceding IL-17 production (Hirota et al., 2011). Thus, Th17 cells “convert” into IFN-γ producers. The molecular underpinnings of this process have been in part investigated. It appears that IL-23 is important for the “re-programming” of Th17 cells into T cells that produce IFN-γ in vivo, designated as Th17/1 cells. In these cells, T-bet was shown to down-modulate the expression of ROR-γt by inhibiting its Runx-1 mediated transactivation. This might result in the loss of IL-17 production in previous Th17 cells (Lazarevic et al., 2011). A more recent report suggested that IL-23 might drive the expression of IFN-γ in Th17 cells independently of T-bet (Duhen et al., 2013). In humans, Th17/1 cells have also been isolated from inflamed tissue of IBD patients as well as from MS lesions (Kebir et al., 2009). A series of surface markers have been suggested for these cells including a combination of CCR6 and CD161 (Annunziato et al., 2007; Cosmi et al., 2008) as well as a combination of CCR6 and CXCR3 (Acosta-Rodriguez et al., 2007). As compared with bona fide Th1 cells, specific molecules like the transcription factor AhR are private to ex-Th17 cells (and are not expressed in bona fide Th1 cells). IL-23R, IL-1R and CCR6 that are also comprised in the Th17 transcriptional program, persist in reprogrammed Th17 cells. Thus, although both subsets produce IFN-γ, Th1 cells and ex-Th17 cells might fundamentally differ in their homing behavior, their response to the innate cytokine milieu in inflamed tissues as well as their mode of cell death and way of regulation by regulatory T cells (Tregs, see below). In addition, Th17 cells also respond to IL-12 (Lee et al., 2009), which might lead to yet another re-programming outcome in vivo.

T

263

of elevated sodium chloride concentration enhanced the generation of Th17 cells in vitro and increased their expression of ROR-γt, IL17 and IL-23R in both murine (Wu et al., 2013) and human T cells (Kleinewietfeld et al., 2013). Intriguingly, both groups report that mice fed with a high-salt diet and subjected to actively induced EAE, showed an exacerbated disease course with increased frequencies of Th17 cells in the CNS. This effect was found to be in part dependent on the salt-sensing kinase SGK1 (serum/glucocorticoid regulated kinase 1), in that mice lacking this enzyme specifically in T cells exhibited a less dramatic EAE increase under high-salt diet (Wu et al., 2013). These reports are of particular interest in light of an increasing salt intake in western countries (Brown et al., 2009) and the partially overlapping rise in MS incidence (Koch-Henriksen and Sørensen, 2010). However, a high-salt diet does not increase salt concentration in most extraintestinal immune compartments and thus a definite mode of action for the effect in mice is still to be revealed. Therefore, despite the tempting correlation, extensive studies in men are necessary before implying a genuine causality between dietary salt and autoimmune diseases like MS. For all described mucosal effects on EAE, it will remain a crucial challenge for future research to investigate whether the described CNS pathology is conveyed by the exact same cells found to be induced or modulated in mucosal tissues or is instead facilitated by auxiliary cells or systemic effects, impacting on distant effector cells resident in lymphoid tissues (Fig. 1). In any case, the recent discoveries in this area of mucosal immunity have spurred microbial and nutritional studies in men that will potentially help to advance our understanding of the human disease.

D

236 237

C. Sie et al. / Experimental Neurology xxx (2014) xxx–xxx

E

4

Please cite this article as: Sie, C., et al., Th17 cells in central nervous system autoimmunity, Exp. Neurol. (2014), http://dx.doi.org/10.1016/ j.expneurol.2014.03.009

300 301 302 303 304 305 306 307 308 309 310 311 312 313 314 315 316 317 318 319 320 321 322 323 324 325 326 327 328 329 330 331

334 335 336 337 338 339 340 341 342 343 344 345 346 347 348 349 350 351 352 353 354 355 356 357 358 359 360

C. Sie et al. / Experimental Neurology xxx (2014) xxx–xxx

5

Th17 precursor

IL-17

GM-CSF

IL-10

IFN-γ

? Th17

IL-23

ROR-γt c-Maf AhR

ROR-γt (T-bet)

Tissue destruction

F

Tissue remodeling

Th17/1

373 374 375 376 377 378 379 380 381 382 383 384 385 386 387 388 389 390 391 392 393 394 395 396 397 398 399 400 401 402 403 404

D

P

direct disruption of the BBB by IL-17 and IL-22, mediated by modulation of endothelial tight junctions (Kebir et al., 2007). In an in vitro model of the BBB, these features enabled Th17 cells to cross a microvascular endothelial monolayer more efficiently than Th1 cells or uncommitted T lymphocytes. Furthermore, IL-17 signaling provoked excess oxidative stress in these endothelial cells which in turn activated their contractile machinery, leading to a downregulation of the tight junction molecule occludin and eventually to an impaired barrier function (Huppert et al., 2010). There is also evidence for a direct effect of IL-17 on CNS-resident cells. When an important component of the IL-17R signal transduction pathway (Gaffen, 2009), the NF-κB activator 1 (Act1), was specifically deleted in cells of neuroectodermal origin (i.e. neurons, astrocytes, and oligodendrocytes), this resulted in reduced EAE severity (Kang et al., 2010). In contrast, ablation in endothelial cells, macrophages or microglia had no effect. In the follow-up study, this effect could be narrowed down to NG2+ glial cells which represent oligodendrocyte progenitors (Kang et al., 2013). Furthermore, IL-17 inhibited the maturation and reduced the survival of oligodendrocyte lineage cells in vitro. In another study, IL-17 alone did not affect survival of oligodendrocytes, but provoked exacerbation of TNF-α induced oligodendrocyte loss and inhibited differentiation of oligodendrocyte progenitor cells (Paintlia et al., 2011). Finally, there is also evidence that direct interactions of Th17 cells with neurons in demyelinating lesions induce severe neuronal dysfunction (Siffrin et al., 2010), whereas the exact mechanism remains enigmatic. In summary, there is evidence for a wide range of proinflammatory and neurotoxic effects exerted by IL-17 in CNS autoimmunity. However, several studies have shown that IL-17 signaling is dispensable for the induction of EAE in that deficiency or neutralization of IL-17 at best entailed attenuation but never complete resistance to the disease (Chen et al., 2006; Haak et al., 2008; Komiyama et al., 2006; Kroenke et al., 2008; Tigno-Aranjuez et al., 2009; Uyttenhove and Van Snick, 2006). Several factors could account for this phenomenon: First, in the absence of IL-17 producing Th17 cells, other T cell subsets such as Th1 cells might compensate via IL-17 independent effector mechanisms. Second, other Th17 derived proinflammatory cytokines such as GMCSF, IL-9, IL-21 or IL-22 could drive the pathology in the absence of IL-17. Indeed, it was known even before the rise of the Th17 concept that genetic deletion of GM-CSF in all murine cells resulted in complete resistance to EAE (McQualter et al., 2001). More recent studies refined this observation and attributed the lack of EAE pathology specifically to the absence of GM-CSF producing CD4+ T lymphocytes, most notably

E

T

C

371 372

E

369 370

R

367 368

R

365 366

N C O

363 364

GM-CSF is part of the CSF family of hematopoietic growth factors. Although this cytokine is produced by a multitude of different cells, T cells represent the major source of GM-CSF under inflammatory conditions. It signals through a heterodimeric receptor consisting of a unique α-chain and a common β-chain. GM-CSF acts on various cell types controlling activation and survival of macrophages, neutrophils and eosinophils. It is also involved in the maturation of DCs as well as the differentiation of NKT cells and alveolar macrophages (Hamilton, 2008). IL-9 is mainly produced by T cells and has initially been described as an autocrine growth factor which is also active in mast cells. The IL-9R is composed of two subunits: the private α-chain (IL-9Rα) and the common γ-chain (γc) shared by other cytokines including IL-2 and IL-21. The receptor is expressed on various hematopoietic cells such as T cells, B cells and mast cells, while non-hematopoietic targets probably include epithelial and smooth muscle cells (Goswami and Kaplan, 2011). IL-21 belongs to the IL-2 family of cytokines and is expressed in various CD4+ T cell subsets as well as NKT cells. Apart from its abundant expression in Th17 cells, IL-21 is also produced by Th2 cells and follicular T helper cells (Tfh), a specialized population of follicular CD4+ T cells involved in B cell help. IL-21R is composed of the γc and a private IL-21 receptor chain (IL21R). The IL21R is expressed on a plethora of hematopoietic cell types, including T cells, B cells, NK cells, DCs, and macrophages, indicative of the highly pleiotropic effects exerted by IL-21. Indeed, it does not only modulate various aspects of T cell biology but is simultaneously involved in the expansion of B cells and their class switch recombination of immunoglobulin isotypes (Ouyang et al., 2008). IL-22 is a member of the IL-10 family of cytokines and IL-22R and IL10R2 were identified as the heterodimeric receptor complex mediating IL-22 signaling. T cells are its main cellular source. Whereas IL-10R is ubiquitously expressed, IL-22R expression is restricted to tissueresident cells (Ouyang et al., 2008). How do these individual cytokines contribute to the proinflammatory features of Th17 cells observed in CNS autoimmunity? In a cascade of downstream effects, IL-17 induces a distinct set of cytokines and chemokines that eventually promote neuroinflammation: For instance, IL-17 can stimulate epithelial cells to produce CXCL1 and CXCL2 (Kolls and Lindén, 2004), two chemokines that have been detected within EAE and MS lesions (Das Sarma et al., 2009; Filipovic et al., 2003). These so-called ELR+ chemokines attract neutrophils that drive inflammation and activation of the BBB (Carlson et al., 2008). Consequently, large numbers of leukocytes can be recruited to the perivascular white matter. In addition to these indirect effects, there is also evidence for

U

361 362

R O

O

Fig. 2. Plasticity of Th17 cells. Th17 cells are increasingly perceived to exhibit a particular tissue- and context-dependent plasticity. The endpoints of this dynamic system appear to be a phenotype rather involved in tissue remodeling (left) and a highly destructive phenotype involved in efficient pathogen clearance but also autoimmunity (right). The hallmark transcription factor of both extremes is ROR-γt while additional environmental and transcriptional cues further define the outcome of the Th17 response. Under the influence of IL-23, Th17 precursor cells or previously IL-17 producing T helper cells would acquire a destructive phenotype including the secretion of IFN-γ, GM-CSF, and other cytokines. While this might be in part mediated by T-bet signaling, it is not absolutely required for this phenotype. On the opposite, as yet only partially characterized signals (e.g. TGF-β1 and IL6 in the absence of IL-23 in vitro but not necessarily in vivo) drive a Th17 response that, by trend, carries out regulatory and remodeling functions via IL-10 and IL-17, respectively. In part, this could be directed by signaling via AhR although the effect is dependent on the specific AhR-ligand. Another potential transcription factor involved in this phenotype is the proto-oncogene c-Maf.

Please cite this article as: Sie, C., et al., Th17 cells in central nervous system autoimmunity, Exp. Neurol. (2014), http://dx.doi.org/10.1016/ j.expneurol.2014.03.009

405 406 407 408 409 410 411 412 413 414 415 416 417 418 419 420 421 422 423 424 425 426 427 428 429 430 431 432 433 434 435 436 437 438 439 440 441 442 443 444 445 446 447 448

474 475 476 477 478 479 480 481 482 483 484 485 486 487 488

494 495 496 497 498 499 500 501 502 503 504 505 506 507 508 509 510 511 512

C

472 473

E

470 471

R

468 469

R

466 467

O

464 465

C

462 463

N

460 461

U

458 459

Th17 cells in multiple sclerosis

528

Interestingly, IL-17 mRNA and protein levels have been found to be elevated in circulating leukocytes and cerebrospinal mononuclear cells of MS patients, in particular during relapses (Brucklacher-Waldert et al., 2009; Durelli et al., 2009; Matusevicius et al., 1999; VakninDembinsky et al., 2006). In line with these findings, microarray approaches revealed increased transcripts encoding IL-17 and RORC (gene name for ROR-γt) in MS plaques compared to normal brain tissue (Lock et al., 2002; Montes et al., 2009). Furthermore, immunohistochemical studies detected IL-17 positive CD4+ and CD8+ T cells in active MS lesions (Tzartos et al., 2008). Interestingly, increased IL-17 production of mononuclear cells in response to myelin basic protein correlated with disease activity as determined by magnetic resonance imaging (Hedegaard et al., 2008). Several studies have addressed the question whether therapeutic agents utilized in the therapy of MS patients have the potential to influence the differentiation or activity of Th17 cells. It has been shown that IFN-β, one of the first-line disease modifying agents in MS, inhibited Th17 differentiation in vitro (Durelli et al., 2009; Ramgolam et al., 2009). These effects of IFN-β are possibly mediated by direct and indirect mechanisms. IFN-β shifted cytokine production of APCs such as DCs from rather pro-inflammatory cytokines such as IL-1β or IL-23 to anti-inflammatory cytokines such as IL-27 (Ramgolam et al., 2009; Sweeney et al., 2011) arguing in favor of an indirect effect of IFN-β on Th17 cell development. In contrast, IFN-β also seemed to directly inhibit differentiation of naïve T cells into Th17 cells (Ramgolam et al., 2009). Interestingly, poor response to IFN-β coincided with higher IL-17F serum levels compared to IFN-β responders (Axtell et al., 2010). It was suggested that Th17 mediated disease was paralleled by an endogenous upregulation of IFN-β expression and it was postulated that exogenous administration of IFN-β would therefore not be beneficial any more in these patients. It was even speculated that excess IFN-β could exert detrimental effects under these conditions (Axtell et al., 2011). However, another study revealed no significant difference in IL-17F serum levels between responders and non-responders to IFN-β (Bushnell et al., 2012), thus refuting the idea to use serum IL-17F levels as a biomarker to stratify patients for the allotment of IFN-β therapy. Other agents used in the treatment of MS patients such as fingolimod (FTY-720) or dimethyl fumarate have also been implicated with a reduction of the Th17 response (Mehling et al., 2010; Peng et al., 2012). So far, it is not clear whether a more specific blockade of the Th17 pathway has beneficial effects in MS patients. Treatment with an antibody directed against IL-12p40 and therefore neutralizing both IL-12 and IL-23 did not result in a significant reduction of disease activity (Segal et al., 2008). However, this study had several drawbacks. First, it is not clear whether the antibody reached the CNS in sufficient quantities. Second, it is not known what effect neutralization of both IL-12 and IL-23 has on already established Th1 or Th17 cells. In this context,

529 530

F

As described above, Th17 cells can facilitate a strong inflammatory response. These effects not only are required in host defense but also need to be properly controlled in order to avoid excessive immunopathology. Also, under physiological conditions, erroneous activation of Th17 precursor cells has to be prevented. Foxp3+ Tregs contribute critically to the preservation of immune homeostasis. However, it is not clear whether they utilize universal mechanisms to mediate their suppressive activity or whether their approach is individually tailored for different types of effector responses. Several functional studies of transcription factors have demonstrated that Treg responses mirror the effector T cell response on a molecular level. Rudensky and colleagues showed that Treg-specific ablation of Stat3, a key transcription factor for Th17 cell differentiation, resulted in an uncontrolled Th17 cell response (Chaudhry et al., 2009). This was putatively mediated via the production of IL-6 and TGF-β by Stat-3 deficient Treg cells. Furthermore, Stat-3 deficiency in Tregs resulted in lower levels of CCR6, a chemokine receptor involved in migration to sites of Th17 inflammation. In a subsequent study, the same group reported that Tregs needed to sense IL-10 in order to control Th17 mediated inflammation. Their data suggested a positive feedback loop in which signaling via the IL-10R on Tregs led to the Stat3-dependent

456 457

O

492 493

455

R O

Regulation of Th17 responses

453 454

513 514

P

491

451 452

expression of further IL-10, collectively containing Th17 responses (Chaudhry et al., 2011). IL-10 is known to serve as a potent negative regulator of inflammation (Jankovic et al., 2010) and several studies have suggested that IL-10 inhibits differentiation of Th17 cells in a direct and indirect manner (Gu et al., 2008; Li and Flavell, 2008; McGeachy et al., 2007). Some of these data indicated that the IL-10 loop of effector T cell control was particularly important for Th17 responses and less relevant for Th1 or Th2 responses. Another option to preserve immune homeostasis is the elimination of activated T cells by cell intrinsic mechanisms, for example by activation-induced cell death (AICD). In this process, TCR signaling leads to apoptosis, mainly mediated by Fas. Several studies have shown that Th17 cells undergo Fas-mediated AICD, even though they were less susceptible than Th1 cells (Fang et al., 2010; Zhang et al., 2008).

T

489 490

Th17 cells (Codarri et al., 2011; El-behi et al., 2011). In particular, adoptively transferred, autoaggressive T cells that were unable to produce GM-CSF failed to induce disease and abrogation of GM-CSF by monoclonal antibody treatment reduced EAE severity. Of note, Th17 cells produced GM-CSF in an IL-23 dependent manner (Codarri et al., 2011; El-behi et al., 2011) and GM-CSF in turn triggered IL-23 production by APCs, thus establishing a positive feedback loop (El-behi et al., 2011; Sonderegger et al., 2008a). Mechanistically, the pathogenic targets of GM-CSF were likely cells of myeloid origin that were recruited to the CNS (Codarri et al., 2011) and activated (El-behi et al., 2011) under the influence of this cytokine. However, details on the exact mode of action remain to be elucidated and human trials will have to show whether antibody treatment against GM-CSF is beneficial in autoimmune diseases. In contrast to GM-CSF, the role of IL-9 in CNS autoimmunity is still largely controversial. One study reported that neutralization of IL-9 or deficiency in IL-9 receptor expression resulted in delayed onset of EAE (Nowak et al., 2009) whereas another study observed an earlier onset of disease combined with an even more severe disease course in IL-9 receptor deficient mice (Elyaman et al., 2009). Studies on the role of IL-21 and IL-22 have clearly shown that these two cytokines were dispensable for the induction of EAE (Coquet et al., 2008; Kreymborg et al., 2007; Sonderegger et al., 2008b). A final effector mechanism to be discussed here is the intriguing interaction of Th17 cells with B cells. Meticulous histological analysis of mice subjected to adoptive transfer of encephalitogenic Th17 cells revealed that the development of EAE in this model was accompanied by the formation of ectopic lymphoid follicles in the CNS (Jäger et al., 2009). These structures contained clusters of B cells, in part well organized by reticulin fibers, indicative of a specific accumulation. The presence of ectopic follicles was partially mediated by IL-17 and the cell surface molecule Podoplanin that was expressed on Th17 cells but not on other Th subsets (Peters et al., 2011). These observations are in line with reports, that Th17 cell derived IL-17 promoted the generation of germinal centers (Hsu et al., 2008; Mitsdoerffer et al., 2010). As IL-21 is known to be of particular importance in B cell biology it was tempting to speculate that IL-21 is also involved in the process of ectopic follicle formation. However, disease development and follicle formation was independent of IL-21 signaling in host cells (Peters et al., 2011). The discovered processes are of notable interest since ectopic lymphoid follicles can also be found in the CNS of MS patients with a chronic progressive disease course (Magliozzi et al., 2007; Serafini et al., 2004).

D

449 450

C. Sie et al. / Experimental Neurology xxx (2014) xxx–xxx

E

6

Please cite this article as: Sie, C., et al., Th17 cells in central nervous system autoimmunity, Exp. Neurol. (2014), http://dx.doi.org/10.1016/ j.expneurol.2014.03.009

515 516 517 518 519 520 521 522 523 524 525 526 527

531 532 533 534 535 536 537 538 539 540 541 542 543 544 545 546 547 548 549 550 551 552 553 554 555 556 557 558 559 560 561 562 563 564 565 566 567 568 569 570 571 572 573 574 575 576

C. Sie et al. / Experimental Neurology xxx (2014) xxx–xxx

598 599 600 601 602 603 604 605 606 607 608 609 610 611 612 613

618 619 620 621 622 623 624 625 626 627 628 629 630 631 632 633 634 635 636 637 638 639

C

596 597

E

594 595

R

643

References

648

F

592 593

R

590 591

N C O

588 589

U

586 587

This work was supported by a Ph.D. fellowship to C.S. from Boehringer Ingelheim Fonds, grants by the German Research Foundation (DFG) to T.K. (Heisenberg, TR128, SFB1054) and M.M. (SyNergy) as well as intramural grants to M.M. from the Technische Universität München (KKF).

Acosta-Rodriguez, E.V., Rivino, L., Geginat, J., Jarrossay, D., Gattorno, M., Lanzavecchia, A., Sallusto, F., Napolitani, G., 2007. Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nat. Immunol. 8, 639–646. Annunziato, F., Cosmi, L., Santarlasci, V., Maggi, L., Liotta, F., Mazzinghi, B., Parente, E., Filì, L., Ferri, S., Frosali, F., Giudici, F., Romagnani, P., Parronchi, P., Tonelli, F., Maggi, E., Romagnani, S., 2007. Phenotypic and functional features of human Th17 cells. J. Exp. Med. 204, 1849–1861. Atarashi, K., Nishimura, J., Shima, T., Umesaki, Y., Yamamoto, M., Onoue, M., Yagita, H., Ishii, N., Evans, R., Honda, K., Takeda, K., 2008. ATP drives lamina propria TH17 cell differentiation. Nature 455, 808–812. Axtell, R.C., de Jong, B.A., Boniface, K., van der Voort, L.F., Bhat, R., De Sarno, P., Naves, R., Han, M., Zhong, F., Castellanos, J.G., Mair, R., Christakos, A., Kolkowitz, I., Katz, L., Killestein, J., Polman, C.H., de Waal Malefyt, R., Steinman, L., Raman, C., 2010. T helper type 1 and 17 cells determine efficacy of interferon-beta in multiple sclerosis and experimental encephalomyelitis. Nat. Med. 16, 406–412. Axtell, R.C., Raman, C., Steinman, L., 2011. Interferon-β exacerbates Th17-mediated inflammatory disease. Trends Immunol. 32, 272–277. Bartholomäus, I., Kawakami, N., Odoardi, F., Schläger, C., Miljkovic, D., Ellwart, J.W., Klinkert, W.E.F., Flügel-Koch, C., Issekutz, T.B., Wekerle, H., Flügel, A., 2009. Effector T cell interactions with meningeal vascular structures in nascent autoimmune CNS lesions. Nature 462, 94–98. Becher, B., Durell, B.G., Noelle, R.J., 2002. Experimental autoimmune encephalitis and inflammation in the absence of interleukin-12. J. Clin. Invest. 110, 493–497. Ben-Nun, A., Wekerle, H., Cohen, I.R., 1981. The rapid isolation of clonable antigen-specific T lymphocyte lines capable of mediating autoimmune encephalomyelitis. Eur. J. Immunol. 11, 195–199. Berer, K., Krishnamoorthy, G., 2012. Commensal gut flora and brain autoimmunity: a love or hate affair? Acta Neuropathol. 123, 639–651. Berer, K., Mues, M., Koutrolos, M., Rasbi, Z. Al, Boziki, M., Johner, C., Wekerle, H., Krishnamoorthy, G., 2011. Commensal microbiota and myelin autoantigen cooperate to trigger autoimmune demyelination. Nature 479, 538–541. Bettelli, E., Carrier, Y., Gao, W., Korn, T., Strom, T.B., Oukka, M., Weiner, H.L., Kuchroo, V.K., 2006. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441, 235–238. Brown, I.J., Tzoulaki, I., Candeias, V., Elliott, P., 2009. Salt intakes around the world: implications for public health. Int. J. Epidemiol. 38, 791–813. Brucklacher-Waldert, V., Stuerner, K., Kolster, M., Wolthausen, J., Tolosa, E., 2009. Phenotypical and functional characterization of T helper 17 cells in multiple sclerosis. Brain 132, 3329–3341. Bushnell, S.E., Zhao, Z., Stebbins, C.C., Cadavid, D., Buko, A.M., Whalley, E.T., Davis, J.A., Versage, E.M., Richert, J.R., Axtell, R.C., Steinman, L., Medori, R., 2012. Serum IL-17F does not predict poor response to IM IFNβ-1a in relapsing–remitting MS. Neurology 79, 531–537. Carlson, T., Kroenke, M., Rao, P., Lane, T.E., Segal, B., 2008. The Th17-ELR + CXC chemokine pathway is essential for the development of central nervous system autoimmune disease. J. Exp. Med. 205, 811–823. Chaudhry, A., Rudra, D., Treuting, P., Samstein, R.M., Liang, Y., Kas, A., Rudensky, A.Y., 2009. CD4 + regulatory T cells control TH17 responses in a Stat3-dependent manner. Science 326, 986–991. Chaudhry, A., Samstein, R.M., Treuting, P., Liang, Y., Pils, M.C., Heinrich, J.-M., Jack, R.S., Wunderlich, F.T., Brüning, J.C., Müller, W., Rudensky, A.Y., 2011. Interleukin-10 signaling in regulatory T cells is required for suppression of Th17 cell-mediated inflammation. Immunity 34, 566–578. Chen, Y., Langrish, C.L., McKenzie, B., Joyce-Shaikh, B., Stumhofer, J.S., McClanahan, T., Blumenschein, W., Churakovsa, T., Low, J., Presta, L., Hunter, C.A., Kastelein, R.A., Cua, D.J., 2006. Anti-IL-23 therapy inhibits multiple inflammatory pathways and ameliorates autoimmune encephalomyelitis. J. Clin. Invest. 116, 1317–1326. Codarri, L., Gyülvészi, G., Tosevski, V., Hesske, L., Fontana, A., Magnenat, L., Suter, T., Becher, B., 2011. RORγt drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat. Immunol. 12, 560–567. Coquet, J.M., Chakravarti, S., Smyth, M.J., Godfrey, D.I., 2008. Cutting edge: IL-21 is not essential for Th17 differentiation or experimental autoimmune encephalomyelitis. J. Immunol. 180, 7097–7101. Cosmi, L., De Palma, R., Santarlasci, V., Maggi, L., Capone, M., Frosali, F., Rodolico, G., Querci, V., Abbate, G., Angeli, R., Berrino, L., Fambrini, M., Caproni, M., Tonelli, F., Lazzeri, E., Parronchi, P., Liotta, F., Maggi, E., Romagnani, S., Annunziato, F., 2008. Human

O

Inflammatory demyelination in multiple sclerosis is most likely the result of an antigen specific adaptive immune response. The priming of lymphocytes and the commitment of T cells to a specific cytokine phenotype happens in the peripheral immune system. Th17 cells appear to play a paramount role in autoimmune tissue inflammation due to their exclusive repertoire of effector functions that are mediated by secretion of IL-17, GM-CSF and other molecules. Whether mucosa associated lymphoid tissue has a prominent role in the differentiation of Th17 precursor cells is an intriguing hypothesis that needs to be further validated. Because of the important role of IL-17 in barrier function at epithelial surfaces, the cellular sources of IL-17 that include Th17 lymphocytes but also cells of the innate immune system (like ILC3 cells) are equipped with a molecular machinery to sense and integrate environmental cues and translate them into appropriate immune responses. The latest insights into these mechanisms might help to provide us with a cell biological framework that translates environmental triggers into disease susceptibility. Besides the ongoing hunt for relevant autoantigens in MS, this new concept could be a rich source for further discoveries that might eventually be exploited for therapeutic approaches. Until then, a greater understanding of Th17 cell biology as well as the exploration of specific biomarkers that are dependent on or coincide with Th17 responses could provide invaluable tools to stratify patients for already existing therapies.

584 585

642

R O

617

583

641

Acknowledgments

P

Concluding remarks

581 582

The authors declare no conflict of interest.

D

616

579 580

640

Conflict of interest

T

614 615

after some promising preliminary data, it will be interesting to see whether the monoclonal antibody secukinumab, directed against IL-17A, will show beneficial effects in MS patients in the ongoing larger phase IIb study and eventually in a phase III trial. From the immunologic analysis of various recent MS therapy trials emerges the concept that distinct treatment regimens – either by design or incidentally – affect various T cell subsets in a differential manner. The consequential question is whether this knowledge can be used to stratify patients for distinct treatments more efficiently. So far, the disease course of MS patients has mainly been used to discern subtypes of MS in relapsing–remitting, secondary progressive, and primary progressive. However, it is known that patients exhibit significant differences with regard to lesion location, histological features, or various “biomarkers” in the CSF. It will be a major challenge to integrate this additional information and perhaps correlate it with a distinct immunologic reaction type on an individual basis. For example, in Asian patients, two different subtypes of MS can be distinguished: optico-spinal MS (OS-MS) and conventional MS. Whereas lesions in OS-MS patients are primarily localized in the optic nerve and spinal cord, patients with conventional MS exhibit lesions scattered throughout the CNS including the brain. Interestingly, OS-MS patients showed significantly higher levels of IL-17 and IL-8, the functional homolog of mouse CXCL1, in the CSF than conventional MS patients. It was particularly intriguing that IL-8 levels were positively correlated with clinical parameters, measured by the expanded disability status scale (EDSS), as well as the length of spinal cord lesions detected by MRI. Histological analysis revealed massive infiltration of neutrophils in necrotic spinal cord lesions in autopsies of 3 out of 6 OS-MS patients (Ishizu et al., 2005). Notably, patients suffering from neuromyelitis optica, another demyelinating disease of the CNS predominantly affecting the optic nerves and spinal cord, showed increased frequencies of Th17 cells (Varrin-Doyer et al., 2012; Wang et al., 2011). In summary, there was evidence for a “Th17 mediated disease” in OS-MS patients whereas patients affected by conventional MS exhibited a more Th1 like reactivity in their immunologic evaluation. Even though European MS patients do not show such a clear distinction, it is possible that there are patients with a disease type that is, by trend, predominantly mediated by Th1 effects and others that exhibit more prominent Th17 features.

E

577 578

7

Please cite this article as: Sie, C., et al., Th17 cells in central nervous system autoimmunity, Exp. Neurol. (2014), http://dx.doi.org/10.1016/ j.expneurol.2014.03.009

644 Q2 645 646 647 Q3

649 650 651 652 653 654 655 656 657 658 659 660 661 662 663 664 665 666 667 668 669 670 671 672 673 674 675 676 677 678 679 680 681 682 683 684 685 686 687 688 689 690 691 692 693 694 695 696 697 698 699 700 701 702 703 704 705 706 707 708 709 710 711 712 713 714 715

D

P

R O

O

F

Hsu, H.-C., Yang, P., Wang, J., Wu, Q., Myers, R., Chen, J., Yi, J., Guentert, T., Tousson, A., Stanus, A.L., Le, T.L., Lorenz, R.G., Xu, H., Kolls, J.K., Carter, R.H., Chaplin, D.D., Williams, R.W., Mountz, J.D., 2008. Interleukin 17-producing T helper cells and interleukin 17 orchestrate autoreactive germinal center development in autoimmune BXD2 mice. Nat. Immunol. 9, 166–175. Hu, W., Troutman, T.D., Edukulla, R., Pasare, C., 2011. Priming microenvironments dictate cytokine requirements for T helper 17 cell lineage commitment. Immunity 35, 1010–1022. Huppert, J., Closhen, D., Croxford, A., White, R., Kulig, P., Pietrowski, E., Bechmann, I., Becher, B., Luhmann, H.J., Waisman, A., Kuhlmann, C.R.W., 2010. Cellular mechanisms of IL-17-induced blood–brain barrier disruption. FASEB J. 24, 1023–1034. Ishizu, T., Osoegawa, M., Mei, F.-J., Kikuchi, H., Tanaka, M., Takakura, Y., Minohara, M., Murai, H., Mihara, F., Taniwaki, T., Kira, J., 2005. Intrathecal activation of the IL-17/ IL-8 axis in opticospinal multiple sclerosis. Brain 128, 988–1002. Ivanov, I.I., McKenzie, B.S., Zhou, L., Tadokoro, C.E., Lepelley, A., Lafaille, J.J., Cua, D.J., Littman, D.R., 2006. The orphan nuclear receptor RORγt directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell 126, 1121–1133. Ivanov, I.I., Frutos, R.deL., Manel, N., Yoshinaga, K., Rifkin, D.B., Sartor, R.B., Finlay, B.B., Littman, D.R., 2008. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 4, 337–349. Ivanov, I.I., Atarashi, K., Manel, N., Brodie, E.L., Shima, T., Karaoz, U., Wei, D., Goldfarb, K.C., Santee, C.A., Lynch, S.V., Tanoue, T., Imaoka, A., Itoh, K., Takeda, K., Umesaki, Y., Honda, K., Littman, D.R., 2009. Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell 139, 485–498. Jäger, A., Dardalhon, V., Sobel, R.A., Bettelli, E., Kuchroo, V.K., 2009. Th1, Th17, and Th9 effector cells induce experimental autoimmune encephalomyelitis with different pathological phenotypes. J. Immunol. 183, 7169–7177. Jankovic, D., Kugler, D.G., Sher, A., 2010. IL-10 production by CD4+ effector T cells: a mechanism for self-regulation. Mucosal Immunol. 3, 239–246. Kang, Z., Altuntas, C.Z., Gulen, M.F., Liu, C., Giltiay, N., Qin, H., Liu, L., Qian, W., Ransohoff, R. M., Bergmann, C., Stohlman, S., Tuohy, V.K., Li, X., 2010. Astrocyte-restricted ablation of interleukin-17-induced Act1-mediated signaling ameliorates autoimmune encephalomyelitis. Immunity 32, 414–425. Kang, Z., Wang, C., Zepp, J., Wu, L., Sun, K., Zhao, J., Chandrasekharan, U., DiCorleto, P.E., Trapp, B.D., Ransohoff, R.M., Li, X., 2013. Act1 mediates IL-17-induced EAE pathogenesis selectively in NG2+ glial cells. Nat. Neurosci. 16, 1401–1408. Kawanokuchi, J., Shimizu, K., Nitta, A., Yamada, K., Mizuno, T., Takeuchi, H., Suzumura, A., 2008. Production and functions of IL-17 in microglia. J. Neuroimmunol. 194, 54–61. Kebir, H., Kreymborg, K., Ifergan, I., Dodelet-Devillers, A., Cayrol, R., Bernard, M., Giuliani, F., Arbour, N., Becher, B., Prat, A., 2007. Human TH17 lymphocytes promote blood– brain barrier disruption and central nervous system inflammation. Nat. Med. 13, 1173–1175. Kebir, H., Ifergan, I., Alvarez, J.I., Bernard, M., Poirier, J., Arbour, N., Duquette, P., Prat, A., 2009. Preferential recruitment of interferon-γ-expressing TH17 cells in multiple sclerosis. Ann. Neurol. 66, 390–402. Kermode, A.G., Thompson, A.J., Tofts, P., MacManus, D.G., Kendall, B.E., Kingsley, D.P., Moseley, I.F., Rudge, P., McDonald, W.I., 1990. Breakdown of the blood–brain barrier precedes symptoms and other MRI signs of new lesions in multiple sclerosis. Pathogenetic and clinical implications. Brain 113 (Pt 5), 1477–1489. Kleinewietfeld, M., Manzel, A., Titze, J., Kvakan, H., Yosef, N., Linker, R.A., Muller, D.N., Hafler, D.A., 2013. Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells. Nature 496, 518–522. Kleiter, I., Hellwig, K., Berthele, A., Al, E., 2012. Failure of natalizumab to prevent relapses in neuromyelitis optica. Arch. Neurol. 69, 239–245. Koch-Henriksen, N., Sørensen, P.S., 2010. The changing demographic pattern of multiple sclerosis epidemiology. Lancet Neurol. 9, 520–532. Kolls, J.K., Lindén, A., 2004. Interleukin-17 family members and inflammation. Immunity 21, 467–476. Komiyama, Y., Nakae, S., Matsuki, T., Nambu, A., Ishigame, H., Kakuta, S., Sudo, K., Iwakura, Y., 2006. IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis. J. Immunol. 177, 566–573. Korn, T., Bettelli, E., Gao, W., Awasthi, A., Jager, A., Strom, T.B., Oukka, M., Kuchroo, V.K., 2007. IL-21 initiates an alternative pathway to induce proinflammatory TH17 cells. Nature 448, 484–487. Korn, T., Bettelli, E., Oukka, M., Kuchroo, V.K., 2009. IL-17 and Th17 cells. Annu. Rev. Immunol. 27, 485–517. Kreymborg, K., Etzensperger, R., Dumoutier, L., Haak, S., Rebollo, A., Buch, T., Heppner, F.L., Renauld, J.-C., Becher, B., 2007. IL-22 is expressed by Th17 cells in an IL-23-dependent fashion, but not required for the development of autoimmune encephalomyelitis. J. Immunol. 179, 8098–8104. Kroenke, M.A., Carlson, T.J., Andjelkovic, A.V., Segal, B.M., 2008. IL-12- and IL-23modulated T cells induce distinct types of EAE based on histology, CNS chemokine profile, and response to cytokine inhibition. J. Exp. Med. 205, 1535–1541. Lampropoulou, V., Hoehlig, K., Roch, T., Neves, P., Gómez, E.C., Sweenie, C.H., Hao, Y., Freitas, A.A., Steinhoff, U., Anderton, S.M., Fillatreau, S., 2008. TLR-activated B cells suppress T cell-mediated autoimmunity. J. Immunol. 180, 4763–4773. Langrish, C.L., Chen, Y., Blumenschein, W.M., Mattson, J., Basham, B., Sedgwick, J.D., McClanahan, T., Kastelein, R.A., Cua, D.J., 2005. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J. Exp. Med. 201, 233–240. Lavasani, S., Dzhambazov, B., Nouri, M., Fåk, F., Buske, S., Molin, G., Thorlacius, H., Alenfall, J., Jeppsson, B., Weström, B., Unutmaz, D., 2010. A novel probiotic mixture exerts a therapeutic effect on experimental autoimmune encephalomyelitis mediated by IL-10 producing regulatory T cells. PLoS One 5, e9009. Lazarevic, V., Chen, X., Shim, J.-H., Hwang, E.-S., Jang, E., Bolm, A.N., Oukka, M., Kuchroo, V. K., Glimcher, L.H., 2011. T-bet represses TH17 differentiation by preventing Runx1mediated activation of the gene encoding ROR[gamma]t. Nat. Immunol. 12, 96–104.

N

C

O

R

R

E

C

T

interleukin 17-producing cells originate from a CD161 + CD4+ T cell precursor. J. Exp. Med. 205, 1903–1916. Croxford, A.L., Mair, F., Becher, B., 2012. IL-23: one cytokine in control of autoimmunity. Eur. J. Immunol. 42, 2263–2273. Cua, D.J., Sherlock, J., Chen, Y., Murphy, C.A., Joyce, B., Seymour, B., Lucian, L., To, W., Kwan, S., Churakova, T., Zurawski, S., Wiekowski, M., Lira, S.A., Gorman, D., Kastelein, R.A., Sedgwick, J.D., 2003. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature 421, 744–748. Dardalhon, V., Korn, T., Kuchroo, V.K., Anderson, A.C., 2008. Role of Th1 and Th17 cells in organ-specific autoimmunity. J. Autoimmun. 31, 252–256. Das Sarma, J., Ciric, B., Marek, R., Sadhukhan, S., Caruso, M.L., Shafagh, J., Fitzgerald, D.C., Shindler, K.S., Rostami, A., 2009. Functional interleukin-17 receptor A is expressed in central nervous system glia and upregulated in experimental autoimmune encephalomyelitis. J. Neuroinflammation 6, 14. Duhen, R., Glatigny, S., Arbelaez, C.A., Blair, T.C., Oukka, M., Bettelli, E., 2013. Cutting edge: the pathogenicity of IFN-producing Th17 cells is independent of T-bet. J. Immunol. 190, 4478–4482. Durelli, L., Conti, L., Clerico, M., Boselli, D., Contessa, G., Ripellino, P., Ferrero, B., Eid, P., Novelli, F., 2009. T-helper 17 cells expand in multiple sclerosis and are inhibited by interferon-beta. Ann. Neurol. 65, 499–509. El-behi, M., Ciric, B., Dai, H., Yan, Y., Cullimore, M., Safavi, F., Zhang, G.-X., Dittel, B.N., Rostami, A., 2011. The encephalitogenicity of T(H)17 cells is dependent on IL-1and IL-23-induced production of the cytokine GM-CSF. Nat. Immunol. 12, 568–575. Ely, L.K., Fischer, S., Garcia, K.C., 2009. Structural basis of receptor sharing by interleukin 17 cytokines. Nat. Immunol. 10, 1245–1251. Elyaman, W., Bradshaw, E.M., Uyttenhove, C., Dardalhon, V., Awasthi, A., Imitola, J., Bettelli, E., Oukka, M., Van Snick, J., Renauld, J.-C., Kuchroo, V.K., Khoury, S.J., 2009. IL-9 induces differentiation of TH17 cells and enhances function of FoxP3+ natural regulatory T cells. Proc. Natl. Acad. Sci. 106, 12885–12890. Engelhardt, B., Ransohoff, R.M., 2012. Capture, crawl, cross: the T cell code to breach the blood–brain barriers. Trends Immunol. 33, 579–589. Fang, Y., Yu, S., Ellis, J.S., Sharav, T., Braley-Mullen, H., 2010. Comparison of sensitivity of Th1, Th2, and Th17 cells to Fas-mediated apoptosis. J. Leukoc. Biol. 87, 1019–1028. Ferber, I.A., Brocke, S., Taylor-Edwards, C., Ridgway, W., Dinisco, C., Steinman, L., Dalton, D., Fathman, C.G., 1996. Mice with a disrupted IFN-gamma gene are susceptible to the induction of experimental autoimmune encephalomyelitis (EAE). J. Immunol. 156, 5–7. Filipovic, R., Jakovcevski, I., Zecevic, N., 2003. GRO-alpha and CXCR2 in the human fetal brain and multiple sclerosis lesions. Dev. Neurosci. 25, 279–290. Fletcher, J.M., Lalor, S.J., Sweeney, C.M., Tubridy, N., Mills, K.H.G., 2010. T cells in multiple sclerosis and experimental autoimmune encephalomyelitis. Clin. Exp. Immunol. 162, 1–11. Gaboriau-Routhiau, V., Rakotobe, S., Lécuyer, E., Mulder, I., Lan, A., Bridonneau, C., Rochet, V., Pisi, A., De Paepe, M., Brandi, G., Eberl, G., Snel, J., Kelly, D., Cerf-Bensussan, N., 2009. The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses. Immunity 31, 677–689. Gaffen, S.L., 2009. Structure and signalling in the IL-17 receptor family. Nat. Rev. Immunol. 9, 556–567. Ghoreschi, K., Laurence, A., Yang, X.-P., Tato, C.M., McGeachy, M.J., Konkel, J.E., Ramos, H.L., Wei, L., Davidson, T.S., Bouladoux, N., Grainger, J.R., Chen, Q., Kanno, Y., Watford, W.T., Sun, H.-W., Eberl, G., Shevach, E.M., Belkaid, Y., Cua, D.J., Chen, W., O'Shea, J.J., 2010. Generation of pathogenic TH17 cells in the absence of TGFbeta signalling. Nature 467, 967–971. Glatigny, S., Duhen, R., Oukka, M., Bettelli, E., 2011. Cutting edge: loss of α4 integrin expression differentially affects the homing of Th1 and Th17 cells. J. Immunol. 187, 6176–6179. Gold, R., Linington, C., Lassmann, H., 2006. Understanding pathogenesis and therapy of multiple sclerosis via animal models: 70 years of merits and culprits in experimental autoimmune encephalomyelitis research. Brain 129, 1953–1971. Goswami, R., Kaplan, M.H., 2011. A brief history of IL-9. J. Immunol. 186, 3283–3288. Gran, B., Zhang, G.-X., Yu, S., Li, J., Chen, X.-H., Ventura, E.S., Kamoun, M., Rostami, A., 2002. IL-12p35-deficient mice are susceptible to experimental autoimmune encephalomyelitis: evidence for redundancy in the IL-12 system in the induction of central nervous system autoimmune demyelination. J. Immunol. 169, 7104–7110. Gu, Y., Yang, Jianfei, Ouyang, X., Liu, W., Li, H., Yang, Jianjun, Bromberg, J., Chen, S.-H., Mayer, L., Unkeless, J.C., Xiong, H., 2008. Interleukin 10 suppresses Th17 cytokines secreted by macrophages and T cells. Eur. J. Immunol. 38, 1807–1813. Haak, S., Croxford, A.L., Kreymborg, K., Heppner, F.L., Pouly, S., Becher, B., Waisman, A., 2008. IL-17A and IL-17F do not contribute vitally to autoimmune neuro-inflammation in mice. J. Clin. Invest. 119 (1), 61–69. Hamilton, J.A., 2008. Colony-stimulating factors in inflammation and autoimmunity. Nat. Rev. Immunol. 8, 533–544. Harrington, L.E., Hatton, R.D., Mangan, P.R., Turner, H., Murphy, T.L., Murphy, K.M., Weaver, C.T., 2005. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat. Immunol. 6, 1123–1132. Hedegaard, C.J., Krakauer, M., Bendtzen, K., Lund, H., Sellebjerg, F., Nielsen, C.H., 2008. T helper cell type 1 (Th1), Th2 and Th17 responses to myelin basic protein and disease activity in multiple sclerosis. Immunology 125, 161–169. Henao-Mejia, J., Elinav, E., Jin, C., Hao, L., Mehal, W.Z., Strowig, T., Thaiss, C.A., Kau, A.L., Eisenbarth, S.C., Jurczak, M.J., Camporez, J.-P., Shulman, G.I., Gordon, J.I., Hoffman, H. M., Flavell, R.A., 2012. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 482, 179–185. Hirahara, K., Poholek, A., Vahedi, G., Laurence, A., Kanno, Y., Milner, J.D., O'Shea, J.J., 2013. Mechanisms underlying helper T-cell plasticity: Implications for immune-mediated disease. J. Allergy Clin. Immunol. 131, 1276–1287. Hirota, K., Duarte, J.H., Veldhoen, M., Hornsby, E., Li, Y., Cua, D.J., Ahlfors, H., Wilhelm, C., Tolaini, M., Menzel, U., Garefalaki, A., Potocnik, A.J., Stockinger, B., 2011. Fate mapping of IL-17-producing T cells in inflammatory responses. Nat. Immunol. 12, 255–263.

U

716 717 718 719 720 721 722 723 724 725 726 727 728 729 730 731 732 733 734 735 736 737 738 739 740 741 742 743 744 745 746 747 748 749 750 751 752 753 754 755 756 757 758 759 760 761 762 763 764 765 766 767 768 769 770 771 772 773 774 775 776 777 778 779 780 781 782 783 784 785 786 787 788 789 790 791 792 793 794 795 796 797 798 799 800 801

C. Sie et al. / Experimental Neurology xxx (2014) xxx–xxx

E

8

Please cite this article as: Sie, C., et al., Th17 cells in central nervous system autoimmunity, Exp. Neurol. (2014), http://dx.doi.org/10.1016/ j.expneurol.2014.03.009

802 803 804 805 806 807 808 809 810 811 812 813 814 815 816 817 818 819 820 821 822 823 824 825 826 827 828 829 830 831 832 833 834 835 836 837 838 839 840 841 842 843 844 845 846 847 848 849 850 851 852 853 854 855 856 857 858 859 860 861 862 863 864 865 866 867 868 869 870 871 872 873 874 875 876 877 878 879 880 881 882 883 884 885 886 887

C. Sie et al. / Experimental Neurology xxx (2014) xxx–xxx

N C O

R

R

E

C

D

P

R O

O

F

Parham, C., Chirica, M., Timans, J., Vaisberg, E., Travis, M., Cheung, J., Pflanz, S., Zhang, R., Singh, K.P., Vega, F., To, W., Wagner, J., O'Farrell, A.-M., McClanahan, T., Zurawski, S., Hannum, C., Gorman, D., Rennick, D.M., Kastelein, R.A., de Waal Malefyt, R., Moore, K.W., 2002. A receptor for the heterodimeric cytokine IL-23 is composed of IL12Rβ1 and a novel cytokine receptor subunit, IL-23R. J. Immunol. 168, 5699– 5708. Peng, H., Guerau-de-Arellano, M., Mehta, V.B., Yang, Y., Huss, D.J., Papenfuss, T.L., LovettRacke, A.E., Racke, M.K., 2012. Dimethyl fumarate inhibits dendritic cell maturation via nuclear factor κB (NF-κB) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) and mitogen stress-activated kinase 1 (MSK1) signaling. J. Biol. Chem. 287, 28017–28026. Perdew, G.H., Babbs, C.F., 1991. Production of ah receptor ligands in rat fecal suspensions containing tryptophan or indole-3-carbinol. Nutr. Cancer 16, 209–218. Peters, A., Pitcher, L.A., Sullivan, J.M., Mitsdoerffer, M., Acton, S.E., Franz, B., Wucherpfennig, K., Turley, S., Carroll, M.C., Sobel, R.A., Bettelli, E., Kuchroo, V.K., 2011. Th17 cells induce ectopic lymphoid follicles in central nervous system tissue inflammation. Immunity 35, 986–996. Quintana, F.J., Basso, A.S., Iglesias, A.H., Korn, T., Farez, M.F., Bettelli, E., Caccamo, M., Oukka, M., Weiner, H.L., 2008. Control of Treg and TH17 cell differentiation by the aryl hydrocarbon receptor. Nature 453, 65–71. Ramgolam, V.S., Sha, Y., Jin, J., Zhang, X., Markovic-Plese, S., 2009. IFN-beta inhibits human Th17 cell differentiation. J. Immunol. 183, 5418–5427. Rangachari, M., Kuchroo, V.K., 2013. Using EAE to better understand principles of immune function and autoimmune pathology. J. Autoimmun. 45, 31–39. Reboldi, A., Coisne, C., Baumjohann, D., Benvenuto, F., Bottinelli, D., Lira, S., Uccelli, A., Lanzavecchia, A., Engelhardt, B., Sallusto, F., 2009. C–C chemokine receptor 6-regulated entry of TH-17 cells into the CNS through the choroid plexus is required for the initiation of EAE. Nat. Immunol. 10, 514–523. Rothhammer, V., Heink, S., Petermann, F., Srivastava, R., Claussen, M.C., Hemmer, B., Korn, T., 2011. Th17 lymphocytes traffic to the central nervous system independently of α4 integrin expression during EAE. J. Exp. Med. 208, 2465–2476. Sawcer, S., Hellenthal, G., Pirinen, M., Spencer, C.C.A., Patsopoulos, N.A., et al., 2011. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature 476, 214–219. Schluesener, H.J., Wekerle, H., 1985. Autoaggressive T lymphocyte lines recognizing the encephalitogenic region of myelin basic protein: in vitro selection from unprimed rat T lymphocyte populations. J. Immunol. 135, 3128–3133. Segal, B.M., Dwyer, B.K., Shevach, E.M., 1998. An interleukin (IL)-10/IL-12 immunoregulatory circuit controls susceptibility to autoimmune disease. J. Exp. Med. 187, 537–546. Segal, B.M., Constantinescu, C.S., Raychaudhuri, A., Kim, L., Fidelus-Gort, R., Kasper, L.H., Investigators, U.M.S., 2008. Repeated subcutaneous injections of IL12/23 p40 neutralising antibody, ustekinumab, in patients with relapsing–remitting multiple sclerosis: a phase II, double-blind, placebo-controlled, randomised, dose-ranging study. Lancet Neurol. 7, 796–804. Serafini, B., Rosicarelli, B., Magliozzi, R., Stigliano, E., Aloisi, F., 2004. Detection of ectopic B-cell follicles with germinal centers in the meninges of patients with secondary progressive multiple sclerosis. Brain Pathol. 14, 164–174. Siffrin, V., Radbruch, H., Glumm, R., Niesner, R., Paterka, M., Herz, J., Leuenberger, T., Lehmann, S.M., Luenstedt, S., Rinnenthal, J.L., Laube, G., Luche, H., Lehnardt, S., Fehling, H.-J., Griesbeck, O., Zipp, F., 2010. In vivo imaging of partially reversible th17 cell-induced neuronal dysfunction in the course of encephalomyelitis. Immunity 33, 424–436. Sonderegger, I., Iezzi, G., Maier, R., Schmitz, N., Kurrer, M., Kopf, M., 2008a. GM-CSF mediates autoimmunity by enhancing IL-6-dependent Th17 cell development and survival. J. Exp. Med. 205, 2281–2294. Sonderegger, I., Kisielow, J., Meier, R., King, C., Kopf, M., 2008b. IL-21 and IL-21R are not required for development of Th17 cells and autoimmunity in vivo. Eur. J. Immunol. 38, 1833–1838. Stromnes, I.M., Cerretti, L.M., Liggitt, D., Harris, R.A., Goverman, J.M., 2008. Differential regulation of central nervous system autoimmunity by TH1 and TH17 cells. Nat. Med. 14, 337–342. Sweeney, C.M., Lonergan, R., Basdeo, S.A., Kinsella, K., Dungan, L.S., Higgins, S.C., Kelly, P.J., Costelloe, L., Tubridy, N., Mills, K.H.G., Fletcher, J.M., 2011. IL-27 mediates the response to IFN-β therapy in multiple sclerosis patients by inhibiting Th17 cells. Brain Behav. Immun. 25, 1170–1181. Tigno-Aranjuez, J.T., Jaini, R., Tuohy, V.K., Lehmann, P.V., Tary-Lehmann, M., 2009. Encephalitogenicity of complete Freund's adjuvant relative to CpG is linked to induction of Th17 cells. J. Immunol. 183, 5654–5661. Traugott, U., Lebon, P., 1988a. Interferon-gamma and Ia antigen are present on astrocytes in active chronic multiple sclerosis lesions. J. Neurol. Sci. 84, 257–264. Traugott, U., Lebon, P., 1988b. Multiple sclerosis: involvement of interferons in lesion pathogenesis. Ann. Neurol. 24, 243–251. Tzartos, J.S., Friese, M.A., Craner, M.J., Palace, J., Newcombe, J., Esiri, M.M., Fugger, L., 2008. Interleukin-17 production in central nervous system-infiltrating T cells and glial cells is associated with active disease in multiple sclerosis. Am. J. Pathol. 172, 146–155. Uyttenhove, C., Van Snick, J., 2006. Development of an anti-IL-17A auto-vaccine that prevents experimental auto-immune encephalomyelitis. Eur. J. Immunol. 36, 2868–2874. Vaknin-Dembinsky, A., Balashov, K., Weiner, H.L., 2006. IL-23 is increased in dendritic cells in multiple sclerosis and down-regulation of IL-23 by antisense oligos increases dendritic cell IL-10 production. J. Immunol. 176, 7768–7774. Varrin-Doyer, M., Spencer, C.M., Schulze-Topphoff, U., Nelson, P.A., Stroud, R.M., Cree, B.A.C., Zamvil, S.S., 2012. Aquaporin 4-specific T cells in neuromyelitis optica exhibit a Th17 bias and recognize Clostridium ABC transporter. Ann. Neurol. 72, 53–64.

E

T

Lee, Y.K., Turner, H., Maynard, C.L., Oliver, J.R., Chen, D., Elson, C.O., Weaver, C.T., 2009. Late developmental plasticity in the T helper 17 lineage. Immunity 30, 92–107. Lee, Y.K., Menezes, J.S., Umesaki, Y., Mazmanian, S.K., 2011. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc. Natl. Acad. Sci. 108, 4615–4622. Lee, Y., Awasthi, A., Yosef, N., Quintana, F.J., Xiao, S., Peters, A., Wu, C., Kleinewietfeld, M., Kunder, S., Hafler, D.A., Sobel, R.A., Regev, A., Kuchroo, V.K., 2012. Induction and molecular signature of pathogenic TH17 cells. Nat. Immunol. 13, 991–999. Lees, J.R., Golumbek, P.T., Sim, J., Dorsey, D., Russell, J.H., 2008. Regional CNS responses to IFN-gamma determine lesion localization patterns during EAE pathogenesis. J. Exp. Med. 205, 2633–2642. Li, M.O., Flavell, R.A., 2008. Contextual regulation of inflammation: a duet by transforming growth factor-beta and interleukin-10. Immunity 28, 468–476. Liang, S.C., Long, A.J., Bennett, F., Whitters, M.J., Karim, R., Collins, M., Goldman, S.J., Dunussi-Joannopoulos, K., Williams, C.M.M., Wright, J.F., Fouser, L.A., 2007. An IL17F/A heterodimer protein is produced by mouse Th17 cells and induces airway neutrophil recruitment. J. Immunol. 179, 7791–7799. Liu, S., Song, X., Chrunyk, B.A., Shanker, S., Hoth, L.R., Marr, E.S., Griffor, M.C., 2013. Crystal structures of interleukin 17A and its complex with IL-17 receptor A. Nat. Commun. 4, 1888. Lock, C., Hermans, G., Pedotti, R., Brendolan, A., Schadt, E., Garren, H., Langer-Gould, A., Strober, S., Cannella, B., Allard, J., Klonowski, P., Austin, A., Lad, N., Kaminski, N., Galli, S.J., Oksenberg, J.R., Raine, C.S., Heller, R., Steinman, L., 2002. Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat. Med. 8, 500–508. Magliozzi, R., Howell, O., Vora, A., Serafini, B., Nicholas, R., Puopolo, M., Reynolds, R., Aloisi, F., 2007. Meningeal B-cell follicles in secondary progressive multiple sclerosis associate with early onset of disease and severe cortical pathology. Brain 130, 1089–1104. Mangan, P.R., Harrington, L.E., O'Quinn, D.B., Helms, W.S., Bullard, D.C., Elson, C.O., Hatton, R.D., Wahl, S.M., Schoeb, T.R., Weaver, C.T., 2006. Transforming growth factor-[beta] induces development of the TH17 lineage. Nature 441, 231–234. Matusevicius, D., Kivisäkk, P., He, B., Kostulas, N., Ozenci, V., Fredrikson, S., Link, H., 1999. Interleukin-17 mRNA expression in blood and CSF mononuclear cells is augmented in multiple sclerosis. Mult. Scler. 5, 101–104. McGeachy, M.J., Bak-Jensen, K.S., Chen, Y., Tato, C.M., Blumenschein, W., McClanahan, T., Cua, D.J., 2007. TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nat. Immunol. 8, 1390–1397. McGeachy, M.J., Chen, Y., Tato, C.M., Laurence, A., Joyce-Shaikh, B., Blumenschein, W.M., McClanahan, T.K., O'Shea, J.J., Cua, D.J., 2009. The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo. Nat. Immunol. 10, 314–324. McQualter, J.L., Darwiche, R., Ewing, C., Onuki, M., Kay, T.W., Hamilton, J.A., Reid, H.H., Bernard, C.C.A., 2001. Granulocyte macrophage colony-stimulating factor: a new putative therapeutic target in multiple sclerosis. J. Exp. Med. 194, 873–882. Mehling, M., Lindberg, R., Raulf, F., Kuhle, J., Hess, C., Kappos, L., Brinkmann, V., 2010. Th17 central memory T cells are reduced by FTY720 in patients with multiple sclerosis. Neurology 75, 403–410. Minagar, A., Alexander, J.S., 2003. Blood–brain barrier disruption in multiple sclerosis. Mult. Scler. 9, 540–549. Mitsdoerffer, M., Lee, Y., Jäger, A., Kim, H.-J., Korn, T., Kolls, J.K., Cantor, H., Bettelli, E., Kuchroo, V.K., 2010. Proinflammatory T helper type 17 cells are effective B-cell helpers. Proc. Natl. Acad. Sci. 107, 14292–14297. Montes, M., Zhang, X., Berthelot, L., Laplaud, D.-A., Brouard, S., Jin, J., Rogan, S., Armao, D., Jewells, V., Soulillou, J.-P., Markovic-Plese, S., 2009. Oligoclonal myelin-reactive T-cell infiltrates derived from multiple sclerosis lesions are enriched in Th17 cells. Clin. Immunol. 130, 133–144. Mosmann, T.R., Cherwinski, H., Bond, M.W., Giedlin, M.A., Coffman, R.L., 1986. Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J. Immunol. 136, 2348–2357. Murphy, E., Shibuya, K., Hosken, N., Openshaw, P., Maino, V., Davis, K., Murphy, K., O'Garra, A., 1996. Reversibility of T helper 1 and 2 populations is lost after longterm stimulation. J. Exp. Med. 183, 901–913. Nowak, E.C., Weaver, C.T., Turner, H., Begum-Haque, S., Becher, B., Schreiner, B., Coyle, A.J., Kasper, L.H., Noelle, R.J., 2009. IL-9 as a mediator of Th17-driven inflammatory disease. J. Exp. Med. 206, 1653–1660. Nurieva, R., Yang, X.O., Martinez, G., Zhang, Y., Panopoulos, A.D., Ma, L., Schluns, K., Tian, Q. , Watowich, S.S., Jetten, A.M., Dong, C., 2007. Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature 448, 480–483. Ochoa-Repáraz, J., Mielcarz, D.W., Wang, Y., Begum-Haque, S., Dasgupta, S., Kasper, D.L., Kasper, L.H., 2010. A polysaccharide from the human commensal Bacteroides fragilis protects against CNS demyelinating disease. Mucosal Immunol. 3, 487–495. Odoardi, F., Sie, C., Streyl, K., Ulaganathan, V.K., Schläger, C., Lodygin, D., Heckelsmiller, K., Nietfeld, W., Ellwart, J., Klinkert, W.E.F., Lottaz, C., Nosov, M., Brinkmann, V., Spang, R., Lehrach, H., Vingron, M., Wekerle, H., Flügel-Koch, C., Flügel, A., 2012. T cells become licensed in the lung to enter the central nervous system. Nature 488, 675–679. Oppmann, B., Lesley, R., Blom, B., Timans, J.C., Xu, Y., Hunte, B., Vega, F., Yu, N., Wang, J., Singh, K., Zonin, F., Vaisberg, E., Churakova, T., Liu, M., Gorman, D., Wagner, J., Zurawski, S., Liu, Y.-J., Abrams, J.S., Moore, K.W., Rennick, D., de Waal-Malefyt, R., Hannum, C., Bazan, J.F., Kastelein, R.A., 2000. Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL12. Immunity 13, 715–725. Ouyang, W., Kolls, J.K., Zheng, Y., 2008. The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity 28, 454–467. Paintlia, M.K., Paintlia, A.S., Singh, A.K., Singh, I., 2011. Synergistic activity of interleukin17 and tumor necrosis factor-α enhances oxidative stress-mediated oligodendrocyte apoptosis. J. Neurochem. 116, 508–521.

U

888 889 890 891 892 893 894 895 896 897 898 899 900 901 902 903 904 905 906 907 908 909 910 911 912 913 914 915 916 917 918 919 920 921 922 923 924 925 926 927 928 929 930 931 932 933 934 935 936 937 938 939 940 941 942 943 944 945 946 947 948 949 950 951 952 953 954 955 956 957 958 959 960 961 962 963 964 965 966 967 968 969 970 971 972 973

9

Please cite this article as: Sie, C., et al., Th17 cells in central nervous system autoimmunity, Exp. Neurol. (2014), http://dx.doi.org/10.1016/ j.expneurol.2014.03.009

974 975 976 977 978 979 980 981 982 983 984 985 986 987 988 989 990 991 992 993 994 995 996 997 998 999 1000 1001 1002 1003 1004 1005 1006 1007 1008 1009 1010 1011 1012 1013 1014 1015 1016 1017 1018 1019 1020 1021 1022 1023 1024 1025 1026 1027 1028 1029 1030 1031 1032 1033 1034 1035 1036 1037 1038 1039 1040 1041 1042 1043 1044 1045 1046 1047 1048 1049 1050 1051 1052 1053 1054 1055 1056 1057 Q4 1058 1059

10

1060 1061 1062 1063 1064 1065 1066 1067 1068 1069 1070 1071 1072 1073 1074 1075

C. Sie et al. / Experimental Neurology xxx (2014) xxx–xxx

Veldhoen, M., Hocking, R.J., Atkins, C.J., Locksley, R.M., Stockinger, B., 2006. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17producing T cells. Immunity 24, 179–189. Veldhoen, M., Hirota, K., Westendorf, A.M., Buer, J., Dumoutier, L., Renauld, J.-C., Stockinger, B., 2008. The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature 453, 106–109. Wang, H.H., Dai, Y.Q., Qiu, W., Lu, Z.Q., Peng, F.H., Wang, Y.G., Bao, J., Li, Y., Hu, X.Q., 2011. Interleukin-17-secreting T cells in neuromyelitis optica and multiple sclerosis during relapse. J. Clin. Neurosci. 18, 1313–1317. Wikoff, W.R., Anfora, A.T., Liu, J., Schultz, P.G., Lesley, S.A., Peters, E.C., Siuzdak, G., 2009. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc. Natl. Acad. Sci. 106, 3698–3703. Willenborg, D.O., Fordham, S., Bernard, C.C., Cowden, W.B., Ramshaw, I.A., 1996. IFNgamma plays a critical down-regulatory role in the induction and effector phase of myelin oligodendrocyte glycoprotein-induced autoimmune encephalomyelitis. J. Immunol. 157, 3223–3227.

Wu, C., Yosef, N., Thalhamer, T., Zhu, C., Xiao, S., Kishi, Y., Regev, A., Kuchroo, V.K., 2013. Induction of pathogenic TH17 cells by inducible salt-sensing kinase SGK1. Nature 496, 513–517. Zamvil, S.S., Steinman, L., 1990. The T lymphocyte in experimental allergic encephalomyelitis. Annu. Rev. Immunol. 8, 579–621. Zhang, G.-X., Gran, B., Yu, S., Li, J., Siglienti, I., Chen, X., Kamoun, M., Rostami, A., 2003. Induction of experimental autoimmune encephalomyelitis in IL-12 receptor-β2deficient mice: IL-12 responsiveness is not required in the pathogenesis of inflammatory demyelination in the central nervous system. J. Immunol. 170, 2153–2160. Zhang, Y., Xu, G., Zhang, L., Roberts, A.I., Shi, Y., 2008. Th17 cells undergo Fasmediated activation-induced cell death independent of IFN-gamma. J. Immunol. 181, 190–196. Zhou, L., Ivanov, I.I., Spolski, R., Min, R., Shenderov, K., Egawa, T., Levy, D.E., Leonard, W.J., Littman, D.R., 2007. IL-6 programs TH-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat. Immunol. 8, 967–974.

U

N

C

O

R

R

E

C

T

E

D

P

R O

O

F

1092

Please cite this article as: Sie, C., et al., Th17 cells in central nervous system autoimmunity, Exp. Neurol. (2014), http://dx.doi.org/10.1016/ j.expneurol.2014.03.009

1076 1077 1078 1079 1080 1081 1082 1083 1084 1085 1086 1087 1088 1089 1090 1091

Th17 cells in central nervous system autoimmunity.

Multiple sclerosis (MS) is the most important autoimmune disease of the central nervous system (CNS). Its animal model experimental autoimmune encepha...
743KB Sizes 5 Downloads 6 Views