Th17 Cells in autoimmune diseases

Lei Han , Jing Yang , Xiuwen Wang , Dan Li , Ling Lv , Bin Li

Front. Med. ›› 2015, Vol. 9 ›› Issue (1) : 10 -19.

PDF (173KB)
Front. Med. ›› 2015, Vol. 9 ›› Issue (1) : 10 -19. DOI: 10.1007/s11684-015-0388-9
REVIEW
REVIEW

Th17 Cells in autoimmune diseases

Author information +
History +
PDF (173KB)

Abstract

Th17 cells are a new subset of CD4+ T cells involved in the clearance of extracellular pathogens and fungi. Accumulating evidence suggests that Th17 cells and their signature cytokines have a pivotal role in the pathogenesis of multiple autoimmune-mediated inflammatory diseases. Here, we summarize recent research progress on Th17 function in the development and pathogenesis of autoimmune diseases. We also propose to identify new small molecule compounds to manipulate Th17 function for potential therapeutic application to treat human autoimmune diseases, including rheumatoid arthritis, systemic lupus erythematosus, Sjögren’s syndrome, inflammatory bowel disease, and multiple sclerosis.

Keywords

IL-17 / Th17 cells / RORγt / autoimmune diseases / posttranslational modification / inhibitors

Cite this article

Download citation ▾
Lei Han, Jing Yang, Xiuwen Wang, Dan Li, Ling Lv, Bin Li. Th17 Cells in autoimmune diseases. Front. Med., 2015, 9(1): 10-19 DOI:10.1007/s11684-015-0388-9

登录浏览全文

4963

注册一个新账户 忘记密码

Introduction

Th17 cells have been identified as a new CD4+ T helper lymphocyte lineage following the promising discovery of the role of IL-23 in experimental autoimmune encephalomyelitis (EAE) [ 1] and collagen-induced arthritis (CIA) [ 2]. They are characterized according to their capacity to produce interleukin (IL)-17A (also called IL-17), IL-17F, and IL-22 [ 3]. Th17 cells are currently assumed to mediate host defense, especially against extracellular bacterial infections, and play an important role in the pathogenesis of autoimmune diseases [ 4]. In this review, we discuss our current understanding of the role of Th17 cells in autoimmune diseases, with a focus on rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), Sjögren’s syndrome (SS), inflammatory bowel disease (IBD), and multiple sclerosis (MS).

Th17 cells

Naïve CD4+ T cells differentiate into diverse subsets of T helper cells depending on different cytokine milieu upon T-cell receptor (TCR) stimulation. Unlike the required cytokines for Th1 and Th2 differentiation, the combination of transforming growth factor-β (TGF-β) and IL-6 initiates the development of Th17 lineage in mice [ 5, 6]. TGF-β favors the induction of Th17 under inflammatory milieu despite its role in the development of regulatory T cells [ 7]. IL-6 induces the production of IL-21, which subsequently amplifies the differentiation in an autocrine way [ 8]. IL-23 stabilizes the Th17 phenotype and maintains its ability to produce related cytokines [ 9]. By contrast, the role of TGF-β in the differentiation of human Th17 cells is controversial. Previous research suggested that human Th17 cells were induced by IL-1β plus IL-6 and suppressed by TGF-β [ 10]. More recent studies show that a combination of TGF-β, IL-1β, IL-6, and IL-23 is critical for human Th17 development [ 11, 12].

The retinoic acid-related orphan receptor γt (RORγt), as the master transcription factor, orchestrates the differentiation of Th17 cells [ 13]. RORγt directs the transcription of IL-17A by directly binding to the IL-17A promoter, which is essential for the functions of this cell lineage [ 14]. Moreover, another related nuclear receptor RORα cooperates with RORγt to trigger greater Th17 response and upregulates the expression of IL-17A and IL-17F [ 15]. Moreover, other key transcription factors, including signal transducer and activator of transcription 3 (STAT3), interferon regulatory factor 4 (IRF4), and aryl hydrocarbon receptor (Ahr), have been identified as the pivotal factors during the differentiation and function of Th17 cells [ 1618].

The key effector cytokine of Th17 cells is IL-17A [ 9]. The IL-17/IL-17R complex recruits the U-box-like E3 ubiquitin ligase Act1 to trigger intracellular signaling pathways through homotypic interactions. Binding of Act1 to tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) and TGF-β-associated kinase 1 (TAK1) ultimately activates the canonical NF-κB pathway [ 19]. However, not all the Th17 are pathogenic; thus, TGF-β3 and IL-6 are attributed to the full pathogenic phenotype, which is IL-23 dependent [ 20].

Th17 and RA

RA is an autoimmune rheumatic disease characterized by inflammation of multiple joints, which eventually leads to progressive joint destruction and deformity. Immune cells infiltrate the synovium of RA following aberrant cytokine and chemokine signaling [ 21].

IL-17A cytokines are involved in the pathogenesis of RA, which has been well established both in animal models of autoimmune arthritis and RA patients. IL-17A has been detected both in the serum and synovial fluid (SF) of RA patients [ 2224]. IL-17A induces the expression of receptor activator of the nuclear factor kappa B ligand (RANKL) to enhance osteoclastogenesis [ 25]. Anti-IL-17A treatment reduces the expression of RANKL, alleviates bone structural damage, and reduces disease progression in models [ 26]. Moreover, antibodies against IL-17A, such as secukinumab, have already been used in clinical trials [ 27, 28]. Enhanced epigenetic modifications in RA patients lead to the production of IL-17A, which sustains the survival of synoviocytes and inflammatory cells. These effects initiate the expansion of Th17 cells [ 21]. However, mast cells were also found as the main producer of IL-17A in the synovial tissue of RA patients in a clinical study [ 23].

IL-17F, another Th17-specific cytokine, was significantly increased in RA patients versus healthy control. IL-17F, but not IL-17A, was reduced after methotrexate and anti-TNF treatment [ 29]. This result indirectly proved the role of Th17 cells in the development of RA. Spontaneous development of autoimmune arthritis in SKG mice, which resembles RA in humans, is accompanied with enhanced arthritogenic Th17 cells [ 30]. Higher frequencies of Th17 cells are observed in RA patients as compared with those from healthy groups [ 3136] (Table 1). Th17 frequencies in RA SF mononuclear cells (SFMC) are higher than paired RA peripheral blood mononuclear cells (PBMC) [ 37]. Moreover, CCR6+ memory Th17 cells from early RA patients trigger pathogenic activation of RASFs, which elevates the production of proinflammatory cytokines and other relevant enzymes [ 33]. Interestingly, the frequency of Th17 cells in RA joints is low, which suggests that Th17 cells might not be the main producer of IL-17A within RA joints [ 37]. This finding might be explained by the proposal that Th17 cells are unstable and spontaneously convert to a Th1 phenotype [ 38]. The adoptive transfer of polarized Th17 cells induces severe, destructive polyarthritis in mice, and IL-17A produced by Th17 cells is attributed to the osteoclastogenic effects of Th17 cells [ 39]. In addition, Th17 cells could assist the B cell in producing autoantibodies [ 40]. A small fraction of Th17 cell lineage, which originates from CD25loFoxp3+CD4+ T cells, possesses more potent osteoclastogenic capability than conventional Th17 cells under arthritic conditions [ 41].

Th17 and SLE

SLE is a chronic systemic autoimmune disease characterized by dysregulation of the immune system, which leads to loss of self-tolerance with activation of autoreactive T and B cells [ 42]. Production of autoantibodies contributes to deposition of immune complexes and tissue injury.

In the past years, numerous studies have reported elevated serum IL-17A levels in SLE patients compared with healthy controls [ 4346]. However, a weak correlation was found between IL-17A levels and SLE disease activity index (SLEDAI) [ 46]. Moreover, IL-17A-deficient mice are prevented from the induction of SLE by pristane [ 47], which suggested that IL-17A contributes to the pathogenesis of SLE. Consistent with the increasing serum IL-17A, expansion of Th17 population in SLE patients is also observed and correlated with SLEDAI in most studies (Table 2). One study indicated that Th17 cells in lupus nephritis (LN) patients were higher than those in conventional SLE patients [ 48]. Th17 cells were suggested as a predominant resource of IL-17A in SLE patients in a clinical study [ 49].

However, the alteration of IL-17A levels and Th17 cell frequencies has not been confirmed in larger groups. In the peripheral blood of SLE patients, data about the source of IL-17A are inconclusive, with several studies reporting the importance of CD3+CD4CD8 double-negative (DN) T cells (expressed as either αβ- or γδ-TCR). In patients with SLE, the capacity of IL-17A production by activated CD4+ T cells was equal to that observed in DN T cells. These IL-17A-producing TCRαβ+ DN T cells are detected in kidney biopsies of LN patients [ 50]. Furthermore, reduced frequency of DN T cells (not mentioned as either TCRαβ+ or γδ+) was observed in IL-17A-deficient mice, which were protected from the induction of SLE by pristane [ 47]. IL-2 mitigates inflammation in MRL/lpr mice by reducing IL-17A-producing TCRαβ+ DN T cell [ 51]. By contrast, rapamycin inhibits IL-17A production most in CD4+ T cells [ 49]. Above all, the role of Th17 in SLE needs to be investigated further.

Th17 and SS

Primary SS is an autoimmune disease primarily characterized by chronic inflammation of exocrine glands, which leads to glandular atrophy and deficient function [ 57, 58]. The infiltration of lymphocytes is mainly responsible for the inflammation, and CD4+ T cells constitute the majority of infiltrating lymphocytes in the salivary glands [ 59, 60].

IL-17A knockout (KO) mice are resistant to the induction of SS. Adoptive transfer of Th17 cells into the inducible IL-17A KO mice could rapidly acquire a SS profile [ 61]. C57BL/6J mice that receive adenovirus serotype 5 (Ad5) -IL17A vector, an SS model, express high levels of IL-17A, which correlates with recruitment of inflammatory cells [ 62]. In patients with Sjögren’s syndrome, both systematically elevated levels of IL-17 [ 63, 64] and presence of Th17 cells [ 6567] in inflamed tissue have been observed in multiple studies. Th17 cells are also observed as the predominant infiltrating T cells in the salivary glands [ 65]. High levels of IL-7 secreted from SS patients have the potential to promote Th17 polarization [ 59]. Furthermore, Th17 cells are hypothesized to promote the generation of germinal centers and function as B-cell helpers [ 68]. This evidence indicates the role of Th17 cells in the pathogenesis of Sjögren’s syndrome.

CD3+CD4CD8 DN T cells (either TCRαβ+ or γδ+) were reportedly a predominant producer of IL-17A in SS and infiltrated the salivary gland of SS patients, and TCRαβ+ DN T cells were associated with disease progression [ 69]. Circulating DN T cells are not expanded and inversely correlated with Th17 cells in the early stage of SS. In addition, minor salivary gland infiltrating DN T cells are inversely correlated with circulating DN T cells [ 70]. These results might support the hypothesis that DN T cells might derive from Th17 cells and acquire the ability to migrate into the salivary glands continuously in early SS. Furthermore, IL-17A+ DN T cells from SS were almost resistant to dexamethasone (Dex) treatment, while Dex was capable of significantly reducing IL-17A production from activated CD4+ T cells [ 69]; this condition might be the reason for the poor response of SS patients to common Dex treatment.

Overall, the following hypothesis is developed according to the above evidence: Th17 cells and their production are mainly involved in the initiation of SS. Subsequently, Th17 cells lose expression of surface molecules and give rise to the DN T cell upon the chronic inflammatory milieu. Finally, these IL-17A-producing DN T cells infiltrate the target glands and promote glandular damage.

Th17 and IBD

IBDs, including ulcerative colitis (UC) and Crohn’s disease, are chronic relapsing inflammatory diseases that mainly destroy the gastrointestinal tract [ 71]. The etiology of IBD remains unclear.

Before the discovery of Th17 cells, the Th1/Th2 paradigm was considered critical for mediating mucosal inflammation [ 72]. The main mechanism of UC was thought to be a Th2 response, while CD was thought to be caused by Th1 cells [ 73]. Recently, higher levels of IL-17A and IL-17F have been observed both in IBD mucosa and lamina propria compared with those in a normal gut [ 74, 75]. Massive infiltration of Th17 cells was also detected in IBD patients [ 76]. Based on the accumulating evidence, Th17 cells also contribute to the pathogenesis of IBD and show more potent capacity than Th1 cells in the induction of colitis [ 77]. This finding might be explained by the conversion of Th17 cells to Th1 cells via induction of mucosal IL-23 production in IBD patients [ 77]. Consistent with the aforementioned results, several genes involved in Th17 signaling, such as IL23R and STAT3, could confer susceptibility to IBD, according to the analysis by genome-wide association studies [ 78]. A monoclonal antibody against TNF infliximab downregulates IL-17A expression, which is one of the mechanisms by which infliximab alleviates IBD-related mucosal inflammation [ 79]. IL-21, a cytokine produced by Th17 cells, is upregulated in inflamed mucosal tissue [ 80].

However, confusing results on dextran sulfate sodium-induced colitis revealed that IL-17A deficiency exacerbates colitis, whereas a lack of IL-17F alleviates colitis in mice [ 81]. Adoptive transfer of CD45RBhi T cells from IL-17A-knockout mice induces a more aggressive colitis, which has higher IFN-γ expression [ 82]. By contrast, IL-17RA-deficient mice develop mild colitis [ 83]. These results support the finding that Th17 cytokine can also have protective functions in the intestine and that the pathogenic effect is mainly IL-17F mediated [ 84]. Sarra et al. proposed that whether Th17 cells were inflammatory or protective would depend on the cytokine milieu, especially IFN-γ [ 85]; it could also be explained by the existence of ex-Th17 cells, a type of Th17 cells that was found in Helicobacter hepaticus (Hh)-induced intestinal inflammation. In Hh-induced colitis, CD4+ T cells differentiated into Th1 and Th17 cells, and IL-17A+ cells partly turned into IFN-γ+ IL-17A+ cells, then became IFN-γ+ ex-Th17 cells [ 86]. IL-23 has been confirmed as a key driver in this process [ 87]. Moreover, anti-IL-17A antibody secukinumab failed to attenuate clinical activity in CD patients [ 88]. In conclusion, how Th17 exert functions in the intestinal inflammation needs further investigation.

Th17 and MS

MS is a progressive autoimmune-mediated inflammation of the central nervous system (CNS), and its pathological feature is demuelination, which ultimately leads to neurological impairment [ 89]. The pathogenic mechanisms are widely explored through EAE in mice, which resembles MS in humans.

Over the past decades, EAE induction has been demonstrated to be dominated by Th1 cells [ 90]. Th1 and Th17 cells are identified as the encephalitogenic mediators in MS and its mice models with the discovery of IL-23 [ 3]. However, determining which cell has the dominant role in the pathogenesis of MS remains a controversial topic [ 91, 92]. Increased frequencies of circulating Th17 cells and serum IL-17A have been observed in MS [ 93, 94]. Moreover, in vitro differentiation of myelin oligodendrocyte glycoprotein-specific Th17 cells could induce EAE. Certain costimulatory pathways have also been demonstrated to restrain EAE development by suppressing the generation of Th17 cells and their infiltration into the CNS [ 95].

Nevertheless, none of the Th17 signature cytokines have been identified as the indispensable component for the development of EAE [ 9698]. Only when cultured with IL-23 could Th17 recipients exert inflammatory functions by inducing ectopic accumulation of lymphocytes [ 92]. Deficiency of granulocyte-macrophage colony-stimulating factor (GM-CSF) in autoreactive Th cells is resistant to the induction of EAE despite the presence of IL-17A or interferon (IFN)-γ [ 99]. Encephalitogenic GM-CSF also contributes to the maintenance of the Th17 phenotype [ 100]. Furthermore, a high-salt diet leads to a more severe form of EAE in mice, which are accompanied with a highly pathogenic Th17 population. These pathogenic Th17 cells could upregulate the expression of proinflammatory cytokines, such as TNF-α, IL-2, and GM-CSF [ 101]. Overall, the aforementioned results indicate that Th17 cells might exert their encephalitogenic capacity mainly through GM-CSF and other molecules [ 102] but not classical Th17 signature cytokines.

Treatment against Th17 cells in autoimmune diseases

On account of the Th17 cell pathogenicity in autoimmune diseases, extensive research efforts focus on investigating therapeutic strategies against Th17 cells. The relevant mechanisms that target Th17 cells are faced with an obstacle to achieving differentiation and amplification of the Th17 cell, neutralization of Th17 signature cytokines, and inhibition of the Th17-specific transcription factor [ 103]. Ustekinumab [ 104, 105], which targets the IL-12/IL-23 pathway, or anti-IL-17A monoclonal antibodies AIN457 [ 106], LY2439821 [ 107], and secukinumab [ 88] have shown promising therapeutic effects for the treatment of autoimmune diseases, such as RA, psoriasis, and CD. Nevertheless, the high cost of the monoclonal antibody has to be taken into consideration because the majority of patients could not afford it. Therefore, effective small molecule compounds should be investigated.

RORγt plays a central role in the development and function of Th17 cells. Therefore, RORγt would be a potential pharmacologic target for the treatment. Based on a small molecule screen, several RORγt inhibitors are identified, including digoxin, ML209, SR1001, SR2211, and ursolic acid [ 108]. Treating animals with these small molecules not only delays the onset but also alleviates the severity of models of EAE and CIA [ 109112]. Recently, TMP778, TM920, and GSK805, which were screened from a benzhydryl amide group, have been identified as highly potent and selective RORγt inhibitors [ 113]. Moreover, TMP778 and GSK805 ameliorate the progression of EAE dramatically by inhibiting Th17 cell generation. AS101 (trichloro (dioxoethylene-O, O′) tellurate), an organotellurium compound, has been found to be able to alleviate the progression of EAE by blocking the activation of STAT3 and RORγt [ 114]. All these inhibitors and their derivatives might be used as therapeutic agents that treat autoimmune-mediated inflammatory diseases.

Targeting other modulators of RORγt, such as deubiquitinases, could be a novel strategy for the treatment of autoimmune diseases. The deubiquitinases have been indicated as important modulators in Th17 cells. For example, deficiency of ubiquitin-specific protease 25 (USP25) exacerbates the severity of EAE by enhancing IL-17A-mediated responses [ 115]. Moreover, we have recently identified deubiquitinase USP17 as a novel regulator of RORγt [ 116]. USP17 affects the functions of Th17 cells by stabilizing RORγt via deubiquitination. Furthermore, USP17 level is increased in SLE patients. Based on the potential application of deubiquitinase inhibitors in cancer therapy [ 117], further efforts need to be explored to test whether these inhibitors could also have therapeutic applications in the treatment of autoimmune diseases.

Conclusions

The function of Th17 cells has proven to be indispensable for the development of autoimmune diseases. However, Th17 cells obviously exert pathogenic capacity through multiple molecular pathways in each specific autoimmune disease. Numerous unresolved scientific questions need to be investigated in the development and function of Th17 cells in health and disease. An enhanced understanding of Th17 functions in autoimmunity could promote more effective therapies in clinical practice.

References

[1]

Cua DJ, Sherlock J, Chen Y, Murphy CA, Joyce B, Seymour B, Lucian L, To W, Kwan S, Churakova T, Zurawski S, Wiekowski M, Lira SA, Gorman D, Kastelein RA, Sedgwick JD. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature2003; 421(6924): 744–748

[2]

Murphy CA, Langrish CL, Chen Y, Blumenschein W, McClanahan T, Kastelein RA, Sedgwick JD, Cua DJ. Divergent pro- and antiinflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation. J Exp Med2003; 198(12): 1951–1957

[3]

Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH, Wang Y, Hood L, Zhu Z, Tian Q, Dong C. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol2005; 6(11): 1133–1141

[4]

Ouyang W, Kolls JK, Zheng Y. The biological functions of T helper 17 cell effector cytokines in inflammation. Immunity2008; 28(4): 454–467

[5]

Mangan PR, Harrington LE, O’Quinn DB, Helms WS, Bullard DC, Elson CO, Hatton RD, Wahl SM, Schoeb TR, Weaver CT. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature2006; 441(7090): 231–234

[6]

Bedoya SK, Lam B, Lau K, Larkin J3rd. Th17 cells in immunity and autoimmunity. Clin Dev Immunol2013; 2013: 986789

[7]

Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, Kuchroo VK. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature2006; 441(7090): 235–238

[8]

Wei L, Laurence A, Elias KM, O’Shea JJ. IL-21 is produced by Th17 cells and drives IL-17 production in a STAT3-dependent manner. J Biol Chem2007; 282(48): 34605–34610

[9]

Bettelli E, Korn T, Oukka M, Kuchroo VK. Induction and effector functions of T(H)17 cells. Nature2008; 453(7198): 1051–1057

[10]

Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F. Interleukins 1β and 6 but not transforming growth factor-β are essential for the differentiation of interleukin 17-producing human T helper cells. Nat Immunol2007; 8(9): 942–949

[11]

Volpe E, Servant N, Zollinger R, Bogiatzi SI, Hupé P, Barillot E, Soumelis V. A critical function for transforming growth factor-β, interleukin 23 and proinflammatory cytokines in driving and modulating human T(H)-17 responses. Nat Immunol2008; 9(6): 650–657

[12]

Manel N, Unutmaz D, Littman DR. The differentiation of human T(H)-17 cells requires transforming growth factor-β and induction of the nuclear receptor RORgt. Nat Immunol2008; 9(6): 641–649

[13]

Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ, Littman DR. The orphan nuclear receptor RORgt directs the differentiation program of proinflammatory IL-17+ T helper cells. Cell2006; 126(6): 1121–1133

[14]

Ichiyama K, Yoshida H, Wakabayashi Y, Chinen T, Saeki K, Nakaya M, Takaesu G, Hori S, Yoshimura A, Kobayashi T. Foxp3 inhibits RORgt-mediated IL-17A mRNA transcription through direct interaction with RORgt. J Biol Chem2008; 283(25): 17003–17008

[15]

Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, Chung Y, Ma L, Shah B, Panopoulos AD, Schluns KS, Watowich SS, Tian Q, Jetten AM, Dong C. T helper 17 lineage differentiation is programmed by orphan nuclear receptors RORα and RORg. Immunity2008; 28(1): 29–39

[16]

Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, Watowich SS, Dong C. STAT3 regulates cytokine-mediated generation of inflammatory helper T cells. J Biol Chem2007; 282(13): 9358–9363

[17]

Brüstle A, Heink S, Huber M, Rosenplänter C, Stadelmann C, Yu P, Arpaia E, Mak TW, Kamradt T, Lohoff M. The development of inflammatory T(H)-17 cells requires interferon-regulatory factor 4. Nat Immunol2007; 8(9): 958–966

[18]

Quintana FJ, Basso AS, Iglesias AH, Korn T, Farez MF, Bettelli E, Caccamo M, Oukka M, Weiner HL. Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature2008; 453(7191): 65–71

[19]

Liu C, Qian W, Qian Y, Giltiay NV, Lu Y, Swaidani S, Misra S, Deng L, Chen ZJ, Li X. Act1, a U-box E3 ubiquitin ligase for IL-17 signaling. Sci Signal2009; 2(92): ra63

[20]

Lee Y, Awasthi A, Yosef N, Quintana FJ, Xiao S, Peters A, Wu C, Kleinewietfeld M, Kunder S, Hafler DA, Sobel RA, Regev A, Kuchroo VK. Induction and molecular signature of pathogenic TH17 cells. Nat Immunol2012; 13(10): 991–999

[21]

Benedetti G, Miossec P. Interleukin 17 contributes to the chronicity of inflammatory diseases such as rheumatoid arthritis. Eur J Immunol2014; 44(2): 339–347

[22]

Metawi SA, Abbas D, Kamal MM, Ibrahim MK. Serum and synovial fluid levels of interleukin-17 in correlation with disease activity in patients with RA. Clin Rheumatol2011; 30(9): 1201–1207

[23]

Suurmond J, Dorjée AL, Boon MR, Knol EF, Huizinga TW, Toes RE, Schuerwegh AJ. Mast cells are the main interleukin 17-positive cells in anticitrullinated protein antibody-positive and-negative rheumatoid arthritis and osteoarthritis synovium. Arthritis Res Ther2011; 13(5): R150

[24]

Park JS, Park MK, Lee SY, Oh HJ, Lim MA, Cho WT, Kim EK, Ju JH, Park YW, Park SH, Cho ML, Kim HY. TWEAK promotes the production of interleukin-17 in rheumatoid arthritis. Cytokine2012; 60(1): 143–149

[25]

Lubberts E, Koenders MI, van den Berg WB. The role of T-cell interleukin-17 in conducting destructive arthritis: lessons from animal models. Arthritis Res Ther2005; 7(1): 29–37

[26]

Chao CC, Chen SJ, Adamopoulos IE, Davis N, Hong K, Vu A, Kwan S, Fayadat-Dilman L, Asio A, Bowman EP. Anti-IL-17A therapy protects against bone erosion in experimental models of rheumatoid arthritis. Autoimmunity2011; 44(3): 243–252

[27]

Kellner H. Targeting interleukin-17 in patients with active rheumatoid arthritis: rationale and clinical potential. Ther Adv Musculoskelet Dis; 5(3): 141–152

[28]

Patel DD, Lee DM, Kolbinger F, Antoni C. Effect of IL-17A blockade with secukinumab in autoimmune diseases. Ann Rheum Dis2013; 72(Suppl 2): ii116–ii123

[29]

Jain M, Attur M, Furer V, Todd J, Ramirez R, Lock M, Lu QA, Abramson SB, Greenberg JD. Increased plasma IL-17F levels in rheumatoid arthritis patients are responsive to methotrexate, anti-TNF, and T Cell costimulatory modulation. Inflammation2014<month>Sep</month><day>21</day>. [Epub ahead of print]

[30]

Hirota K, Hashimoto M, Yoshitomi H, Tanaka S, Nomura T, Yamaguchi T, Iwakura Y, Sakaguchi N, Sakaguchi S. T cell self-reactivity forms a cytokine milieu for spontaneous development of IL-17+ Th cells that cause autoimmune arthritis. J Exp Med2007; 204(1): 41–47

[31]

Leipe J, Schramm MA, Prots I, Schulze-Koops H, Skapenko A. Increased Th17 cell frequency and poor clinical outcome in rheumatoid arthritis are associated with a genetic variant in the IL4R gene, rs1805010. Arthritis Rheum (Munch)2014; 66(5): 1165–1175

[32]

Shen H, Goodall JC, Hill Gaston JS. Frequency and phenotype of peripheral blood Th17 cells in ankylosing spondylitis and rheumatoid arthritis. Arthritis Rheum2009; 60(6): 1647–1656

[33]

van Hamburg JP, Asmawidjaja PS, Davelaar N, Mus AM, Colin EM, Hazes JM, Dolhain RJ, Lubberts E. Th17 cells, but not Th1 cells, from patients with early rheumatoid arthritis are potent inducers of matrix metalloproteinases and proinflammatory cytokines upon synovial fibroblast interaction, including autocrine interleukin-17A production. Arthritis Rheum2011; 63(1): 73–83

[34]

Zhang L, Li YG, Li YH, Qi L, Liu XG, Yuan CZ, Hu NW, Ma DX, Li ZF, Yang Q, Li W, Li JM. Increased frequencies of Th22 cells as well as Th17 cells in the peripheral blood of patients with ankylosing spondylitis and rheumatoid arthritis. PLoS ONE2012; 7(4): e31000

[35]

van Hamburg JP, Corneth OB, Paulissen SM, Davelaar N, Asmawidjaja PS, Mus AM, Lubberts E. IL-17/Th17 mediated synovial inflammation is IL-22 independent. Ann Rheum Dis2013; 72(10): 1700–1707

[36]

Kim J, Kang S, Kim J, Kwon G, Koo S. Elevated levels of T helper 17 cells are associated with disease activity in patients with rheumatoid arthritis. Ann Lab Med2013; 33(1): 52–59

[37]

Church LD, Filer AD, Hidalgo E, Howlett KA, Thomas AM, Rapecki S, Scheel-Toellner D, Buckley CD, Raza K. Rheumatoid synovial fluid interleukin-17-producing CD4 T cells have abundant tumor necrosis factor-α co-expression, but little interleukin-22 and interleukin-23R expression. Arthritis Res Ther2010; 12(5): R184

[38]

Nistala K, Adams S, Cambrook H, Ursu S, Olivito B, de Jager W, Evans JG, Cimaz R, Bajaj-Elliott M, Wedderburn LR. Th17 plasticity in human autoimmune arthritis is driven by the inflammatory environment. Proc Natl Acad Sci USA2010; 107(33): 14751–14756

[39]

Sato K, Suematsu A, Okamoto K, Yamaguchi A, Morishita Y, Kadono Y, Tanaka S, Kodama T, Akira S, Iwakura Y, Cua DJ, Takayanagi H. Th17 functions as an osteoclastogenic helper T cell subset that links T cell activation and bone destruction. J Exp Med2006; 203(12): 2673–2682

[40]

Hickman-Brecks CL, Racz JL, Meyer DM, LaBranche TP, Allen PM. Th17 cells can provide B cell help in autoantibody induced arthritis. J Autoimmun2011; 36(1): 65–75

[41]

Komatsu N, Okamoto K, Sawa S, Nakashima T, Oh-hora M, Kodama T, Tanaka S, Bluestone JA, Takayanagi H. Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat Med2014; 20(1): 62–68

[42]

Shlomchik MJ, Craft JE, Mamula MJ. From T to B and back again: positive feedback in systemic autoimmune disease. Nat Rev Immunol2001; 1(2): 147–153

[43]

Wong CK, Lit LC, Tam LS, Li EK, Wong PT, Lam CW. Hyperproduction of IL-23 and IL-17 in patients with systemic lupus erythematosus: implications for Th17-mediated inflammation in auto-immunity. Clin Immunol2008; 127(3): 385–393

[44]

Zhao XF, Pan HF, Yuan H, Zhang WH, Li XP, Wang GH, Wu GC, Su H, Pan FM, Li WX, Li LH, Chen GP, Ye DQ. Increased serum interleukin 17 in patients with systemic lupus erythematosus. Mol Biol Rep2010; 37(1): 81–85

[45]

Cheng F, Guo Z, Xu H, Yan D, Li Q. Decreased plasma IL22 levels, but not increased IL17 and IL23 levels, correlate with disease activity in patients with systemic lupus erythematosus. Ann Rheum Dis2009; 68(4): 604–606

[46]

Vincent FB, Northcott M, Hoi A, Mackay F, Morand EF. Clinical associations of serum interleukin-17 in systemic lupus erythematosus. Arthritis Res Ther2013; 15(4): R97

[47]

Amarilyo G, Lourenço EV, Shi FD, La Cava A. IL-17 promotes murine lupus. J Immunol2014; 193(2): 540–543

[48]

Xing Q, Wang B, Su H, Cui J, Li J. Elevated Th17 cells are accompanied by FoxP3+ Treg cells decrease in patients with lupus nephritis. Rheumatol Int2012; 32(4): 949–958

[49]

Kato H, Perl A. Mechanistic target of rapamycin complex 1 expands Th17 and IL-4+ CD4CD8 double-negative T cells and contracts regulatory T cells in systemic lupus erythematosus. J Immunol2014; 192(9): 4134–4144

[50]

Crispín JC, Oukka M, Bayliss G, Cohen RA, Van Beek CA, Stillman IE, Kyttaris VC, Juang YT, Tsokos GC. Expanded double negative T cells in patients with systemic lupus erythematosus produce IL-17 and infiltrate the kidneys. J Immunol2008; 181(12): 8761–8766

[51]

Mizui M, Koga T, Lieberman LA, Beltran J, Yoshida N, Johnson MC, Tisch R, Tsokos GC. IL-2 protects lupus-prone mice from multiple end-organ damage by limiting CD4CD8 IL-17-producing T cells. J Immunol2014; 193(5): 2168–2177

[52]

Shah K, Lee WW, Lee SH, Kim SH, Kang SW, Craft J, Kang I. Dysregulated balance of Th17 and Th1 cells in systemic lupus erythematosus. Arthritis Res Ther2010; 12(2): R53

[53]

Yang XY, Wang HY, Zhao XY, Wang LJ, Lv QH, Wang QQ. Th22, but not Th17 might be a good index to predict the tissue involvement of systemic lupus erythematosus. J Clin Immunol2013; 33(4): 767–774

[54]

Yang J, Chu Y, Yang X, Gao D, Zhu L, Yang X, Wan L, Li M. Th17 and natural Treg cell population dynamics in systemic lupus erythematosus. Arthritis Rheum2009; 60(5): 1472–1483

[55]

Chen DY, Chen YM, Wen MC, Hsieh TY, Hung WT, Lan JL. The potential role of Th17 cells and Th17-related cytokines in the pathogenesis of lupus nephritis. Lupus2012; 21(13): 1385–1396

[56]

Dolff S, Bijl M, Huitema MG, Limburg PC, Kallenberg CG, Abdulahad WH. Disturbed Th1, Th2, Th17 and T(reg) balance in patients with systemic lupus erythematosus. Clin Immunol2011; 141(2): 197–204

[57]

Voulgarelis M, Tzioufas AG. Pathogenetic mechanisms in the initiation and perpetuation of Sjögren’s syndrome. Nat Rev Rheumatol2010; 6(9): 529–537

[58]

Jonsson R, Vogelsang P, Volchenkov R, Espinosa A, Wahren-Herlenius M, Appel S. The complexity of Sjögren’s syndrome: novel aspects on pathogenesis. Immunol Lett2011; 141(1): 1–9

[59]

Singh N, Cohen PL. The T cell in Sjogren’s syndrome: force majeure, not spectateur. J Autoimmun2012; 39(3): 229–233

[60]

Fox RI, Adamson TC 3rd, Fong S, Young C, Howell FV. Characterization of the phenotype and function of lymphocytes infiltrating the salivary gland in patients with primary Sjögren syndrome. Diagn Immunol1983; 1(3): 233–239

[61]

Lin X, Tian J, Rui K, Ma KY, Ko KH, Wang S, Lu L. The role of T helper 17 cell subsets in Sjögren’s syndrome: similarities and differences between mouse model and humans. Ann Rheum Dis2014; 73(7): e43

[62]

Nguyen CQ, Yin H, Lee BH, Carcamo WC, Chiorini JA, Peck AB. Pathogenic effect of interleukin-17A in induction of Sjögren’s syndrome-like disease using adenovirus-mediated gene transfer. Arthritis Res Ther2010; 12(6): R220

[63]

Ciccia F, Guggino G, Rizzo A, Ferrante A, Raimondo S, Giardina A, Dieli F, Campisi G, Alessandro R, Triolo G. Potential involvement of IL-22 and IL-22-producing cells in the inflamed salivary glands of patients with Sjögren’s syndrome. Ann Rheum Dis2012; 71(2): 295–301

[64]

Nguyen CQ, Hu MH, Li Y, Stewart C, Peck AB. Salivary gland tissue expression of interleukin-23 and interleukin-17 in Sjögren’s syndrome: findings in humans and mice. Arthritis Rheum2008; 58(3): 734–743

[65]

Sakai A, Sugawara Y, Kuroishi T, Sasano T, Sugawara S. Identification of IL-18 and Th17 cells in salivary glands of patients with Sjögren’s syndrome, and amplification of IL-17-mediated secretion of inflammatory cytokines from salivary gland cells by IL-18. J Immunol2008; 181(4): 2898–2906

[66]

Katsifis GE, Rekka S, Moutsopoulos NM, Pillemer S, Wahl SM. Systemic and local interleukin-17 and linked cytokines associated with Sjögren’s syndrome immunopathogenesis. Am J Pathol2009; 175(3): 1167–1177

[67]

Fei Y, Zhang W, Lin D, Wu C, Li M, Zhao Y, Zeng X, Zhang F. Clinical parameter and Th17 related to lymphocytes infiltrating degree of labial salivary gland in primary Sjögren’s syndrome. Clin Rheumatol2014; 33(4): 523–529

[68]

Youinou P, Pers JO. Disturbance of cytokine networks in Sjögren’s syndrome. Arthritis Res Ther2011; 13(4): 227

[69]

Alunno A, Bistoni O, Bartoloni E, Caterbi S, Bigerna B, Tabarrini A, Mannucci R, Falini B, Gerli R. IL-17-producing CD4CD8 T cells are expanded in the peripheral blood, infiltrate salivary glands and are resistant to corticosteroids in patients with primary Sjögren’s syndrome. Ann Rheum Dis2013; 72(2): 286–292

[70]

Alunno A, Carubbi F, Bistoni O, Caterbi S, Bartoloni E, Bigerna B, Pacini R, Beghelli D, Cipriani P, Giacomelli R, Gerli R. CD4(-)CD8(-) T-cells in primary Sjögren’s syndrome: association with the extent of glandular involvement. J Autoimmun2014; 51: 38–43

[71]

Kaser A, Zeissig S, Blumberg RS. Inflammatory bowel disease. Annu Rev Immunol2010; 28(1): 573–621

[72]

Di Sabatino A, Biancheri P, Rovedatti L, MacDonald TT, Corazza GR. New pathogenic paradigms in inflammatory bowel disease. Inflamm Bowel Dis2012; 18(2): 368–371

[73]

Podolsky DK. Inflammatory bowel disease. N Engl J Med2002; 347(6): 417–429

[74]

Fujino S, Andoh A, Bamba S, Ogawa A, Hata K, Araki Y, Bamba T, Fujiyama Y. Increased expression of interleukin 17 in inflammatory bowel disease. Gut2003; 52(1): 65–70

[75]

Seiderer J, Elben I, Diegelmann J, Glas J, Stallhofer J, Tillack C, Pfennig S, Jürgens M, Schmechel S, Konrad A, Göke B, Ochsenkühn T, Müller-Myhsok B, Lohse P, Brand S. Role of the novel Th17 cytokine IL-17F in inflammatory bowel disease (IBD): upregulated colonic IL-17F expression in active Crohn’s disease and analysis of the IL17F p. His161Arg polymorphism in IBD. Inflamm Bowel Dis2008; 14(4): 437–445

[76]

Zenewicz LA, Antov A, Flavell RA. CD4 T-cell differentiation and inflammatory bowel disease. Trends Mol Med 2009; 15(5): 199–207

[77]

Feng T, Qin H, Wang L, Benveniste EN, Elson CO, Cong Y. Th17 cells induce colitis and promote Th1 cell responses through IL-17 induction of innate IL-12 and IL-23 production. J Immunol2011; 186(11): 6313–6318

[78]

Lees CW, Barrett JC, Parkes M, Satsangi J. New IBD genetics: common pathways with other diseases. Gut 2011; 60(12): 1739–1753

[79]

Caprioli F, Bosè F, Rossi RL, Petti L, Viganò C, Ciafardini C, Raeli L, Basilisco G, Ferrero S, Pagani M, Conte D, Altomare G, Monteleone G, Abrignani S, Reali E. Reduction of CD68+ macrophages and decreased IL-17 expression in intestinal mucosa of patients with inflammatory bowel disease strongly correlate with endoscopic response and mucosal healing following infliximab therapy. Inflamm Bowel Dis2013; 19(4): 729–739

[80]

Geremia A, Biancheri P, Allan P, Corazza GR, Di Sabatino A. Innate and adaptive immunity in inflammatory bowel disease. Autoimmun Rev2014; 13(1): 3–10

[81]

Yang XO, Chang SH, Park H, Nurieva R, Shah B, Acero L, Wang YH, Schluns KS, Broaddus RR, Zhu Z, Dong C. Regulation of inflammatory responses by IL-17F. J Exp Med2008; 205(5): 1063–1075

[82]

O’Connor W Jr, Kamanaka M, Booth CJ, Town T, Nakae S, Iwakura Y, Kolls JK, Flavell RA. A protective function for interleukin 17A in T cell-mediated intestinal inflammation. Nat Immunol2009; 10(6): 603–609

[83]

Zhang Z, Zheng M, Bindas J, Schwarzenberger P, Kolls JK. Critical role of IL-17 receptor signaling in acute TNBS-induced colitis. Inflamm Bowel Dis2006; 12(5): 382–388

[84]

Troncone E, Marafini I, Pallone F, Monteleone G. Th17 cytokines in inflammatory bowel diseases: discerning the good from the bad. Int Rev Immunol2013; 32(5–6): 526–533

[85]

Sarra M, Pallone F, Macdonald TT, Monteleone G. IL-23/IL-17 axis in IBD. Inflamm Bowel Dis2010; 16(10): 1808–1813

[86]

Morrison PJ, Bending D, Fouser LA, Wright JF, Stockinger B, Cooke A, Kullberg MC. Th17-cell plasticity in Helicobacter hepaticus-induced intestinal inflammation. Mucosal Immunol2013; 6(6): 1143–1156

[87]

Morrison PJ, Ballantyne SJ, Kullberg MC. Interleukin-23 and T helper 17-type responses in intestinal inflammation: from cytokines to T-cell plasticity. Immunology2011; 133(4): 397–408

[88]

Hueber W, Sands BE, Lewitzky S, Vandemeulebroecke M, Reinisch W, Higgins PD, Wehkamp J, Feagan BG, Yao MD, Karczewski M, Karczewski J, Pezous N, Bek S, Bruin G, Mellgard B, Berger C, Londei M, Bertolino AP, Tougas G, Travis SP; Secukinumab in Crohn’s Disease Study Group. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: unexpected results of a randomised, double-blind placebo-controlled trial. Gut2012; 61(12): 1693–1700

[89]

McFarland HF, Martin R. Multiple sclerosis: a complicated picture of autoimmunity. Nat Immunol2007; 8(9): 913–919

[90]

Voskuhl RR, Martin R, Bergman C, Dalal M, Ruddle NH, McFarland HF. T helper 1 (Th1) functional phenotype of human myelin basic protein-specific T lymphocytes. Autoimmunity1993; 15(2): 137–143

[91]

Kroenke MA, Chensue SW, Segal BM. EAE mediated by a non-IFN-γ/non-IL-17 pathway. Eur J Immunol2010; 40(8): 2340–2348

[92]

Jäger A, Dardalhon V, Sobel RA, Bettelli E, Kuchroo VK. Th1, Th17, and Th9 effector cells induce experimental autoimmune encephalomyelitis with different pathological phenotypes. J Immunol2009; 183(11): 7169–7177

[93]

Romme Christensen J, Börnsen L, Ratzer R, Piehl F, Khademi M, Olsson T, Sørensen PS, Sellebjerg F. Systemic inflammation in progressive multiple sclerosis involves follicular T-helper, Th17- and activated B-cells and correlates with progression. PLoS ONE2013; 8(3): e57820

[94]

Tao Y, Zhang X, Chopra M, Kim MJ, Buch KR, Kong D, Jin J, Tang Y, Zhu H, Jewells V, Markovic-Plese S. The role of endogenous IFN-β in the regulation of Th17 responses in patients with relapsing-remitting multiple sclerosis. J Immunol2014; 192(12): 5610–5617

[95]

Coquet JM, Middendorp S, van der Horst G, Kind J, Veraar EA, Xiao Y, Jacobs H, Borst J. The CD27 and CD70 costimulatory pathway inhibits effector function of T helper 17 cells and attenuates associated autoimmunity. Immunity2013; 38(1): 53–65

[96]

Haak S, Croxford AL, Kreymborg K, Heppner FL, Pouly S, Becher B, Waisman A. IL-17A and IL-17F do not contribute vitally to autoimmune neuro-inflammation in mice. J Clin Invest2009; 119(1): 61–69

[97]

Kreymborg K, Etzensperger R, Dumoutier L, Haak S, Rebollo A, Buch T, Heppner FL, Renauld JC, Becher B. IL-22 is expressed by Th17 cells in an IL-23-dependent fashion, but not required for the development of autoimmune encephalomyelitis. J Immunol2007; 179(12): 8098–8104

[98]

Sonderegger I, Kisielow J, Meier R, King C, Kopf M. IL-21 and IL-21R are not required for development of Th17 cells and autoimmunity in vivo. Eur J Immunol2008; 38(7): 1833–1838

[99]

Codarri L, Gyülvészi G, Tosevski V, Hesske L, Fontana A, Magnenat L, Suter T, Becher B. RORγt drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation. Nat Immunol2011; 12(6): 560–567

[100]

El-Behi M, Ciric B, Dai H, Yan Y, Cullimore M, Safavi F, Zhang GX, Dittel BN, Rostami A. The encephalitogenicity of T(H)17 cells is dependent on IL-1- and IL-23-induced production of the cytokine GM-CSF. Nat Immunol2011; 12(6): 568–575

[101]

Kleinewietfeld M, Manzel A, Titze J, Kvakan H, Yosef N, Linker RA, Muller DN, Hafler DA. Sodium chloride drives autoimmune disease by the induction of pathogenic TH17 cells. Nature2013; 496(7446): 518–522

[102]

Reboldi A, Coisne C, Baumjohann D, Benvenuto F, Bottinelli D, Lira S, Uccelli A, Lanzavecchia A, Engelhardt B, Sallusto F. 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 Immunol2009; 10(5): 514–523

[103]

Maddur MS, Miossec P, Kaveri SV, Bayry J. Th17 cells: biology, pathogenesis of autoimmune and inflammatory diseases, and therapeutic strategies. Am J Pathol2012; 181(1): 8–18

[104]

Krueger GG, Langley RG, Leonardi C, Yeilding N, Guzzo C, Wang Y, Dooley LT, Lebwohl M; CNTO 1275 Psoriasis Study Group. A human interleukin-12/23 monoclonal antibody for the treatment of psoriasis. N Engl J Med2007; 356(6): 580–592

[105]

Sandborn WJ, Feagan BG, Fedorak RN, Scherl E, Fleisher MR, Katz S, Johanns J, Blank M, Rutgeerts P; Ustekinumab Crohn’s Disease Study Group.A randomized trial of Ustekinumab, a human interleukin-12/23 monoclonal antibody, in patients with moderate-to-severe Crohn’s disease. Gastroenterology2008; 135(4): 1130–1141

[106]

Hueber W, Patel DD, Dryja T, Wright AM, Koroleva I, Bruin G, Antoni C, Draelos Z, Gold MH; Psoriasis Study Group, Durez P, Tak PP, Gomez-Reino JJ; Rheumatoid Arthritis Study Group, Foster CS, Kim RY, Samson CM, Falk NS, Chu DS, Callanan D, Nguyen QD; Uveitis Study Group, Rose K, Haider A, Di Padova F. Effects of AIN457, a fully human antibody to interleukin-17A, on psoriasis, rheumatoid arthritis, and uveitis. Sci Transl Med2010; 2(52): 52ra72

[107]

Genovese MC, Van den Bosch F, Roberson SA, Bojin S, Biagini IM, Ryan P, Sloan-Lancaster J. LY2439821, a humanized anti-interleukin-17 monoclonal antibody, in the treatment of patients with rheumatoid arthritis: a phase I randomized, double-blind, placebo-controlled, proof-of-concept study. Arthritis Rheum2010; 62(4): 929–939

[108]

Huh JR, Littman DR. Small molecule inhibitors of RORγt: targeting Th17 cells and other applications. Eur J Immunol2012; 42(9): 2232–2237

[109]

Huh JR, Leung MW, Huang P, Ryan DA, Krout MR, Malapaka RR, Chow J, Manel N, Ciofani M, Kim SV, Cuesta A, Santori FR, Lafaille JJ, Xu HE, Gin DY, Rastinejad F, Littman DR. Digoxin and its derivatives suppress TH17 cell differentiation by antagonizing RORγt activity. Nature2011; 472(7344): 486–490

[110]

Solt LA, Kumar N, Nuhant P, Wang Y, Lauer JL, Liu J, Istrate MA, Kamenecka TM, Roush WR, Vidović D, Schürer SC, Xu J, Wagoner G, Drew PD, Griffin PR, Burris TP. Suppression of TH17 differentiation and autoimmunity by a synthetic ROR ligand. Nature2011; 472(7344): 491–494

[111]

Xu T, Wang X, Zhong B, Nurieva RI, Ding S, Dong C. Ursolic acid suppresses interleukin-17 (IL-17) production by selectively antagonizing the function of RORgt protein. J Biol Chem2011; 286(26): 22707–22710

[112]

Cascão R, Vidal B, Raquel H, Neves-Costa A, Figueiredo N, Gupta V, Fonseca JE, Moita LF. Effective treatment of rat adjuvant-induced arthritis by celastrol. Autoimmun Rev2012; 11(12): 856–862

[113]

Xiao S, Yosef N, Yang J, Wang Y, Zhou L, Zhu C, Wu C, Baloglu E, Schmidt D, Ramesh R, Lobera M, Sundrud MS, Tsai PY, Xiang Z, Wang J, Xu Y, Lin X, Kretschmer K, Rahl PB, Young RA, Zhong Z, Hafler DA, Regev A, Ghosh S, Marson A, Kuchroo VK. Small-molecule RORγt antagonists inhibit T helper 17 cell transcriptional network by divergent mechanisms. Immunity2014; 40(4): 477–489

[114]

Xie L, Chen J, McMickle A, Awar N, Nady S, Sredni B, Drew PD, Yu S. The immunomodulator AS101 suppresses production of inflammatory cytokines and ameliorates the pathogenesis of experimental autoimmune encephalomyelitis. J Neuroimmunol2014; 273(1–2): 31–41

[115]

Zhong B, Liu X, Wang X, Chang SH, Liu X, Wang A, Reynolds JM, Dong C. Negative regulation of IL-17-mediated signaling and inflammation by the ubiquitin-specific protease USP25. Nat Immunol2012; 13(11): 1110–1117

[116]

Han L, Yang J, Wang X, Wu Q, Yin S, Li Z, Zhang J, Xing Y, Chen Z, Tsun A, Li D, Piccioni M, Zhang Y, Guo Q, Jiang L, Bao L, Lv L, Li B. The E3 deubiquitinase USP17 is a positive regulator of retinoic acid-related orphan nuclear receptor γt (RORγt) in Th17 cells. J Biol Chem2014; 289(37): 25546–25555

[117]

Pal A, Young MA, Donato NJ. Emerging potential of therapeutic targeting of ubiquitin-specific proteases in the treatment of cancer. Cancer Res2014; 74(18): 4955–4966

RIGHTS & PERMISSIONS

Higher Education Press and Springer-Verlag Berlin Heidelberg

AI Summary AI Mindmap
PDF (173KB)

3528

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/