Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Mar 1.
Published in final edited form as: Int Immunopharmacol. 2020 Jan 10;80:106187. doi: 10.1016/j.intimp.2020.106187

Molecular control of pathogenic Th17 cells in autoimmune diseases

Bing Wu 1,2,*, Yisong Wan 1,2,*
PMCID: PMC7031035  NIHMSID: NIHMS1549288  PMID: 31931372

Abstract

IL-17A-producing CD4+ T helper cells (Th17) are crucial for the development of inflammatory and autoimmune diseases and thus are exploited for clinical immunotherapies. Emerging evidence suggests Th17 cells are heterogeneous and able to adopt both pathogenic and non-pathogenic phenotypes which are shaped by environmental and genetic factors. On one hand, IL-6 in concert with TGFβ1 can induce non-pathogenic Th17 cells (non-pTh17), which are not effective in inducing tissue inflammation. On the other hand, IL-6, IL-1β with IL-23 induce pathogenic Th17 cells (pTh17) to induce immune pathologies in various tissues. Th17 cells could be both pathogenic and non-pathogenic in a content-dependent manner in vivo. Understanding how the generation and pathogenicity of pTh17 cells are regulated will aid us to devise more effective immunotherapy. In this review, we summarize recent advances in the differentiation and regulation of Th17 cells especially pTh17 cells in vitro and in vivo. The emerging results revealing the specific molecular control of pTh17 cells are highlighted.

Keywords: TGFβ1, IL-10, CD5L, IL-23, IL-23R, non-pathogenic Th17 cells, pathogenic Th17 cells, EAE, autoimmune diseases

1. Introduction

Th17 cells that produce the signature cytokine interleukin 17A (IL-17A) and lineage-specific transcriptional factor retinoid-related orphan receptor gamma t (RORγt) have attracted great attention since their identification in 2005 [1, 2]. Th17 responses contribute to the tissue inflammation and pathogenesis of diverse autoimmune diseases, including psoriasis, rheumatoid arthritis, inflammatory bowel disease (IBD) and multiple sclerosis [36]. Intrigued by the strong correlation of Th17 cells with immuno-pathology in autoimmune diseases, Th17 cells have been exploited in the intervention of these diseases [4, 5, 717].

However, clinical trials targeting IL-17A signaling with Secukinumab (anti-IL-17A antibody) or Brodalumab (anti-IL-17RA antibody) show inconsistent effects. Secukinumab provides encouraging improvement in patients with multiple sclerosis, active ankylosing spondylitis (AS) or psoriasis [18, 19]. In contrast, Secukinumab/Brodalumab failed to improve Th17 cell-mediated Crohn’s and arthritis diseases [20, 21], which is in line with a report that IL-17A might play a protective role in a mouse colitis model [22]. A series of studies suggest that the blockade of IL-17A signaling may even exacerbate disease in intestinal and bone tissues [11, 2224]. Indeed, although Th17 cells are essential for driving inflammatory disorders, Th17 cells also benefit the homeostasis of tissues in certain conditions [2527]. Thus, in vivo generated Th17 cells can be both pathogenic and non-pathogenic to the host in a context-dependent manner. In addition, how naive T cells become non-pathogenic or pathogenic Th17 cells depends on signals from different cytokine milieus in vitro [28, 29]. In light of the dichotomy of Th17 cells, increasing efforts have been devoted to reveal how the generation and function of pathogenic and non-pathogenic Th17 cells are controlled.

2. Differentiation of “non-pathogenic” and “pathogenic” Th17 Cells

The phenotypic and functional properties of Th17 cells have been extensively investigated in vitro and in vivo. Detailed studies suggest that different environmental factors direct naïve CD4+ T cells to differentiation into pTh17 cells or non-pTh17 cells [28, 30, 31].

2.1. Generation and function of “non-pathogenic” Th17 cells

Current knowledge on the Th17-specifying molecular program is largely obtained from studying TGF-β1+IL-6 induced Th17 cells [32]. IL-6 is the key factor that directly regulates the balance of Th17 and Foxp3+ Treg cells by inhibiting TGF-β1-induced Foxp3 expression [33]. In addition, IL-6-induced activation of STAT-3 is essential for the differentiation of Th17 cells from naïve T cells (Figure. 1) [3437]. STAT3-deficient T cells did not respond to IL-6 and failed to elicit a downstream signal [38]. However, in the absence of IL-6, pleiotropic cytokine IL-21 produced by Th17 cells can promote the development of Th17 cells via the activation of STAT3 [3941]. And yet, the relevance and necessity of IL-21 in the polarization of Th17 cells remains uncertain in vivo. The development and function of Th17 cells are unaltered during experimental autoimmune encephalomyelitis (EAE) in IL-21−/− or IL-21R−/− mice [42, 43], suggesting a dispensable role of IL-21 signaling in driving Th17 differentiation and tissue inflammation in vivo.

Figure 1. The differentiation and regulation of Th17 cells.

Figure 1.

Th17 cells are subtyped based on cytokine conditions that define pathogenic versus non-pathogenic function.

While TGF-β1+IL-6-induced Th17 cells may promote inflammation to a certain degree, they are largely considered as non-pathogenic Th17 cells and even demonstrate a protective role under certain circumstances [4447]. Non-pTh17 cells exhibit a regulatory program to produce the immune regulatory cytokines IL-4 and IL-10, as well as critical negative regulators of a range of pathophysiological responses including CD5L (CD5 like molecule), IL-9 and GATA3 [22, 34, 48, 49]. For instance, in steady-state, segmented filamentous bacteria (SFB)-elicited Th17 cells produce a large amount of IL-10 in lamina propria in the intestines, contributing to the immune homeostasis and tissue integrity of the gut [50, 51]. Also, stimulation of MOG peptide-specific T cells with TGF-β1/IL-6 abrogates their pathogenic function during EAE induction and progression [34]. In agreement with the critical role of IL-10, IL-27 and IL-6 trigger STAT3-dependent IL-10 production in Th17 cells and restrain the progression of endometriosis and EAE [48, 52]. While the importance of IL-10 in non-pTh17 cells have been demonstrated, it remains to be addressed as how IL-10 expression is regulated at the transcriptional and epigenetic levels during non-pTh17 generation. Recently, IL-10+ Th17 cells are found to be controlled by c-Maf and can be characterized as a tissue-resident non-pTh17 population [53]. Mechanistic study elucidates c-Maf modulates the immunoregulatory and tissue-residency program by binding to genes such as Pdcd1, Ctla4, and Cd69, suggesting c-Maf maybe an important factor to distinguish between non-pTh17 and pTh17 cells [53]. Furthermore, while the deletion of CD5L does not affect the differentiation of Th17 cells, CD5L regulates lipid metabolism, especially the balance of PUFA/SFA level, to alter RORγt ligand availability in non-pTh17 cells [49]. Compared to enhanced lipid metabolism in Treg cells, Th17 cells favor glycolysis and glutaminolysis pathways [5457]. Inhibition of mitochondrial function and oxidative phosphorylation (OXPHS) is associated with non-pathogenic Th17 gene expression and impaired pTh17 cell function, suggesting that fine-tuning cell fitness and metabolic activity may be used to balance the function of pTh17 and non-pTh17 cells [5862]. While regulatory molecules such as IL-4 and IL-10 participate in the process of metabolism [6365], whether the increased regulatory program modulates the metabolic program in Th17 cells to restrain Th17 cell-mediated inflammatory disorders remains largely unknown and is of great interest.

2.2. Generation and function of “pathogenic” Th17 cells

Researchers have made a substantial effort to interpret the differentiation, maintenance and function of pTh17 cells owing to their critical role in tissue inflammation. In humans, an IL-23R polymorphism has been genetically linked to many autoimmune diseases such as Crohn’s disease and psoriasis [66, 67]. Notably, mice deficient in the p19 subunit of IL-23 fail to produce Th17 cells and were resistant to autoimmune diseases such as EAE, IBD and arthritis [6870]. In line with this notion, the development of pathogenic Th17 cells relies on the cytokine combination of IL-1β/IL-6/IL-23 in vitro. Such pTh17 cells exhibit a detrimental pro-inflammatory program (Il23r, Csf2, Tbx21, Il17a, Il17f, etc) and decreased regulatory modules such as Il4, Il10 and Cd5l [28, 29, 7173] (Figure. 1). In addition, Th17 cells generated via TGF-β1 plus IL-6 do not readily promote EAE disease until further exposure to IL-23 [30, 74], suggesting an essential role for IL-23 signaling in pTh17 cell-induced inflammation. However, whether there are cell intrinsic factors regulated by IL-23 and/or other cytokines that can drive the differentiation of pTh17 in vivo is still unclear and warrants further investigation.

Interestingly, IL-23 is not the inducer of Th17 cells due to the lack of IL-23 receptor (IL-23R) on naïve CD4+ T cells. Nevertheless, IL-23/IL-23R signal promotes the stability and survival of Th17 cells and is indispensable for Th17 cells to gain pathogenic features [5, 30, 68, 74, 75]. IL-23 does so through multiple mechanisms. Transcription factor Blimp-1, downstream of IL-23, drives the pathogenic program of pTh17 cells while suppresses IL2 and Bcl6 that are suppressors of Th17 cells [76]. In addition, GM-CSF is highly produced by pTh17 cells and is essential for the pathogenicity of Th17 cells but not required for Th17 cell generation [7779]. Two mechanisms are involved in increased GM-CSF in Th17 cells: 1) IL-23 and RORγt drive the production of GM-CSF in Th cells [79]; 2) GM-CSF also acts on dendritic cells (DCs) to enhance their production of IL-23, which in turn promotes further activation of Th17 cells and GM-CSF production [80]. Additionally, IL-23 suppresses CD5L expression and regulates the metabolic status of Th17 cells, which is required for its pathogenicity [49]. p-STAT3/STAT4 is shown to be required for IL-23-IL-23R mediated CNS autoimmunity and such STAT4 activation is IL-12 independent. IL-12 signal disruption did not ameliorate EAE progression [81]. However, STAT4-deficient mice are resistant to EAE and colitis [8286]. Global gene expression analysis indicates that in the absence of STAT4, the levels of pathogenic Th17 genes including Tbx21, Il22 and Cxcl3 are significantly reduced, while the expression of non-pathogenic genes including Il10 and Ahr is increased. Given the importance of IL-23 signal in pTh17 cells, specifically interfering with the IL-23 signal with Isankizumab, Guselkumab or Ustekinumab blockade antibodies led to promising outcomes in several clinical trials in patients with rheumatoid arthritis, psoriasis and Crohn’s disease [12, 8791]. However, anti-IL-23 monoclonal antibody treatment did not achieve favorable outcome in patients with advanced relapsing-remitting multiple sclerosis or active ankylosing spondylitis, suggesting a more specific approach to target pTh17 cells is required [9294].

Current studies indicate TGFβ1 is not always required for IL-17A production. IL-17A producing CD4+ T cells are still detectable in the gut from CD4dnTGF-βRII and TGFbr1fl/fl;Cd4Cre mice in which TGFβR signaling is abrogated in T cells [30]. However, acute deletion with tamoxifen in ERcre;TGFβRIIfl/fl mice suggest a requirement of TGFβRII signal in the development of Th17 cells [95]. These findings indicate the requirement of TGF-βRII but not TGF-βRI in the generation of Th17 cells. This is not happening by chance as CD4+ T cells gradually lose TGFβRI but not TGFβRII expression after activation [96], which could also explain the generation of pathogenic Th17 cells in the absence of TGFβ1. TGF-β superfamily members (e.g., TGFβs, activins, and BMPs) regulate diverse developmental and physiological processes. More interestingly, compared to TGF-β1, TGF-β3, another TGF-β family cytokine, is produced by activated Th17 cells and further increased by the addition of IL-23. TGF-β3 plus IL-6 can also induce pTh17 cells (Figure. 1) [28]. Although TGFβ3 shares the same TGFβRII with TGFβ1, TGFβ3 induced the activation of Smad1/5 but not canonical Smad2/3 signal in Th17 cells, further indicating that TGFβ3-induced pTh17 cells are developmentally distinct from non-pTh17 cells. Currently, it is still unclear whether other molecules are involved in the generation of pathogenic Th17 cells beyond TGFβ3 and IL-23. Nonetheless, there are numerous TGF‐β superfamily members that share similar structures and functional overlap and yet distinct receptors [9799]. Indeed, Activin A, a member of the TGF-β superfamily that regulates tissue homeostasis, cell proliferation, and tissue inflammation, is able to promote Th17 differentiation in concert with IL-6 [95]. In addition, the activity of phosphatase PP2A, a factor modulates TGFβ/Activin A/Nodal signaling, controls Th17 cell generation and mediated EAE by its differential effect on the phosphorylation of Smad2 vs. Smad3 [100]. These findings suggest a much broader function of the TGF-β superfamily in the generation and function of Th17 cells through more intricate molecular networks than we previously thought. It therefore would be interesting to know whether and how TGF-β superfamily members besides TGFβ may contribute to the generation and functional specification of distinct Th17 cell subsets.

3. Specific molecular control of pathogenic Th17 cells

Emerging evidence suggests that non-pTh17 and pTh17 cells are distinct cell subsets that controlled by a distinct molecular program. One would expect unique transcriptional factors to direct the generation and function of pTh17 cells. Nonetheless, the identified molecules including RORγt, BATF and IRF4 are indistinguishably required for the generation of both pathogenic- and non-pathogenic-Th17 cells [35, 101103]. Therefore, increasing efforts have been made to shed light on the specific molecular control of pTh17 cells.

3.1. Transcriptional analysis of non-pathogenic and pathogenic Th17 cells

Studies of the transcriptional signatures of non-pathogenic and pathogenic Th17 cells enable us to better understand the molecular network of these two subsets. Lee et. al analyzed the molecular programs associated with pTh17 cells compared to the non-pTh17 cell by genome wide-microarray analysis [28]. To further address the signaling network of pTh17 cell development in vivo, Gaublomme et. al used single-cell RNA sequence technology to investigate the molecular network governing the heterogeneity and the pathogenicity of Th17 cells isolated from EAE diseased mice [49, 104]. Results showed that TGFβ3- and IL-1β/IL-6/IL-23-induced pTh17 cells reveal similar transcriptional gene profiles distinct from that of TGFβ1-induced non-pTh17 cells. The establishment of pathogenicity not only increased a pro-inflammatory genes module (Il23r, Csf2, Tbx21, Il17a, and Il17f) but also reduced the expression of immune-suppressive genes (Il10, Il4, Cd5l, Ahr and cmaf). Single-cell transcriptomics analysis from in vivo isolated IL-17A+ Th17 cells shows that there is a zone of overlapping cell states between non-pTh17 and pTh17 cells [104]. Through co-variation module analysis, a proinflammatory module co-variance with IL-17A and a regulatory module correlation with IL-10 are further established. Intrigued by the sequence data, some putative regulators are selected showing a potential role in the regulation of pathogenicity of Th17 cells: Gpr65, Toso, Plzp, and Cd5l.

3.2. Molecular control of pathogenic Th17 cells

GPR65, Toso, and Plzp were found to promote pTh17 cells and are essential for the progress of EAE, with up-regulated pro-inflammatory genes [104]. For instance, Gpr65-deficient CD4+ T cells show comparable IL-17A+ percentage with WT counterpart under TGF-β1/IL-6 condition but exhibit defective differentiation of Th17 cells under IL-1β/IL-6/IL-23 condition, indicating a differential requirement of Gpr65 in non-pTh17 and pTh17 subsets. However, little is known about whether and how IL-23 regulates those molecular factors. Additional studies focus on other top-ranked candidates such as Hif1a, Fosl2, Stat4, Med12, Etv6, Gem, Foxp1, Rbpj and Procr. Hif1α is a key metabolic sensor and induced in CD4+ T cells in a STAT3/mTOR-dependent manner which is also critical for IL-17A production, through the binding with RORγt and recruiting p300 in non-pTh17 cells [105, 106]. In addition, Notch signaling mediator RBPJ drives the expression of IL-23R and reciprocally suppresses IL-10 expression during Th17 differentiation [107]. Overexpression of IL-23R could rescue the defective pathogenicity of RBPJ-deficient Th17 cells. The role of another candidate molecule, protein C receptor (PROCR), in correlation with IL-10 production in Th17 cells, was also identified in the pathogenicity of Th17 cells [108]. Overexpression of PROCR can reduce the pro-inflammatory gene profile including Il1r and Il23r, indicating a negative role of PROCR in the pathogenicity of Th17 cells. Additionally, microRNA-183 cluster C, which is upregulated by IL-6 and downregulated by TGF-β1 [109], promotes the pathogenic module of pTh17 cells by repressing Foxo1, a suppresser of Th17 cells [109, 110]. Recently, RIP2 has been reported to be involved in the balancing of homeostatic and pathogenic features of Th17 cells [111]. Deletion of RIP2 reduced IL-17A production in non-pTh17 cells. In contrast, RIP2 deficiency enhanced IL-17A expression under pTh17 cell condition via the increase of RORα that promotes Th17 cell differentiation [111].

Are there any molecules that can function in both development and pathogenicity adaption for pTh17 cells but not in non-pTh17 cells? Recently, our group revealed that RASA3 (RAS p21 protein activator 3), a GTPase activating protein of GAP1 sub-family, is specifically required for both the generation and pathogenicity of pTh17 cells and dispensable for non-pTh17 cells (Figure. 1). RASA3 does so by balancing the reciprocal molecular programs of pTh17-Th2 cells via a RASA3-IRF4-Cbl-b axis under pTh17 condition [112]. Although IRF4 is essential for the development of both non-pTh17 and pTh17 cells, our finding revealed that the levels of IRF4 expression appeared to be critical: high levels of IRF4 expression mediated Th2 program suppressed and medium levels of IRF4 expression enhanced the pathogenic-Th17 cell generation of RASA3-deficient T cells. These findings suggest that IRF4 controls the pathogenic-Th17 program in a biphasic, dose-dependent manner.

The above studies suggest that unique molecular programs exist to dictate the generation of pTh17. Both lineage development and pathogenic function adaption contribute to the generation of pTh17 cells in vitro and in vivo. Other molecules and pathways are likely to be discovered to specifically control pTh17 cell generation and immune pathologies. The studies to unveil critical factors in controlling pTh17 cell function will provide valuable therapeutic targets to interfere with the lineage commitment and/or the pathogenicity of Th17 cells and to treat autoimmune diseases.

4. Perspectives and Conclusion

While the past knowledge of Th17 cell function is mainly acquired from TGF-β1/IL-6-induced non-pathogenic Th17 cells, a discrete pathogenic Th17 subset induced by IL-1β/IL-6/IL-23 has attracted great attention for its critical role in controlling autoimmune diseases. Although increasing findings have extended the understanding of pTh17 cells, several questions must be addressed in the future: (1) Are there any specific transcriptional factors or surface makers that can be utilized to distinguish pTh17 cells during pathological conditions? Can these potential candidates be manipulated for the treatment of pTh17-related diseases? (2) Non-pTh17 cell subset is functionally plastic and can be converted into Th1 and Treg cells [113]. If and how the function of pTh17 cells are similarly plastic remains to be investigated. (3) Increasing evidence indicates that Th17 cells can establish transient or long-term residency in non-lymphoid tissue [51, 114, 115] to control immunity, homeostasis and pathology. Nonetheless, the definition and function of Th17 Trm cells in vivo remain poorly defined. In addition, whether Th17 Trm cells share similar molecular controls as CD8+ Trm T cells are interesting questions to be addressed.

In summary, while great strides have been made in understanding the function and regulation of Th17 cells in the past, much work is needed to reveal specific molecular mechanisms underlying the generation and function pTh17 vs. non-pTh17 cells. This will shed light on how Th17 cells impact inflammatory diseases. Additionally, it will benefit the development of therapeutic approaches to treat Th17 cell related diseases.

Highlights:

  1. Systematical summary of non-pathogenic Th17 cells vs. pathogenic Th17 cells

  2. Specific molecular controlling of the development and pathogenicity of Th17 cells

  3. Clinical interventions of Th17 cells in autoimmune diseases

Acknowledgements

We thank the critical reading of this manuscript by Dr. Song Zhang (Nankai University, China) and Dr. Zengli Guo and Dr. Nazanin Kiapour (University of North Carolina at Chapel Hill, USA). We thank the supports from NIH (AI123193) and National Multiple Sclerosis Society (RG-1802-30483) for Y.Y.W.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

Conflict of interest: The authors declare that they have no conflict of interest.

References

  • [1].Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, Weaver CT, Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages, Nature immunology 6(11) (2005) 1123–32. [DOI] [PubMed] [Google Scholar]
  • [2].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, Nature immunology 6(11) (2005) 1133–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Dong C, TH17 cells in development: an updated view of their molecular identity and genetic programming, Nature reviews. Immunology 8(5) (2008) 337–48. [DOI] [PubMed] [Google Scholar]
  • [4].Korn T, Bettelli E, Oukka M, Kuchroo VK, IL-17 and Th17 Cells, Annual review of immunology 27 (2009) 485–517. [DOI] [PubMed] [Google Scholar]
  • [5].Patel DD, Kuchroo VK, Th17 Cell Pathway in Human Immunity: Lessons from Genetics and Therapeutic Interventions, Immunity 43(6) (2015) 1040–51. [DOI] [PubMed] [Google Scholar]
  • [6].Zou W, Restifo NP, T(H)17 cells in tumour immunity and immunotherapy, Nature reviews. Immunology 10(4) (2010) 248–56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [7].Flannigan KL, Ngo VL, Geem D, Harusato A, Hirota SA, Parkos CA, Lukacs NW, Nusrat A, Gaboriau-Routhiau V, Cerf-Bensussan N, Gewirtz AT, Denning TL, IL-17A-mediated neutrophil recruitment limits expansion of segmented filamentous bacteria, Mucosal immunology 10(3) (2017) 673–684. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [8].Nakai K, He YY, Nishiyama F, Naruse F, Haba R, Kushida Y, Katsuki N, Moriue T, Yoneda K, Kubota Y, IL-17A induces heterogeneous macrophages, and it does not alter the effects of lipopolysaccharides on macrophage activation in the skin of mice, Scientific reports 7(1) (2017) 12473. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Wojkowska DW, Szpakowski P, Ksiazek-Winiarek D, Leszczynski M, Glabinski A, Interactions between neutrophils, Th17 cells, and chemokines during the initiation of experimental model of multiple sclerosis, Mediators of inflammation 2014 (2014) 590409. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Ouyang W, Kolls JK, Zheng Y, The biological functions of T helper 17 cell effector cytokines in inflammation, Immunity 28(4) (2008) 454–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [11].Floudas A, Saunders SP, Moran T, Schwartz C, Hams E, Fitzgerald DC, Johnston JA, Ogg GS, McKenzie AN, Walsh PT, Fallon PG, IL-17 Receptor A Maintains and Protects the Skin Barrier To Prevent Allergic Skin Inflammation, Journal of immunology 199(2) (2017) 707–717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Fotiadou C, Lazaridou E, Sotiriou E, Ioannides D, Targeting IL-23 in psoriasis: current perspectives, Psoriasis (Auckl) 8 (2018) 1–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Komiyama Y, Nakae S, Matsuki T, Nambu A, Ishigame H, Kakuta S, Sudo K, Iwakura Y, IL-17 plays an important role in the development of experimental autoimmune encephalomyelitis, Journal of immunology 177(1) (2006) 566–73. [DOI] [PubMed] [Google Scholar]
  • [14].Mizutani N, Nabe T, Yoshino S, IL-17A promotes the exacerbation of IL-33-induced airway hyperresponsiveness by enhancing neutrophilic inflammation via CXCR2 signaling in mice, Journal of immunology 192(4) (2014) 1372–84. [DOI] [PubMed] [Google Scholar]
  • [15].van der Fits L, Mourits S, Voerman JS, Kant M, Boon L, Laman JD, Cornelissen F, Mus AM, Florencia E, Prens EP, Lubberts E, Imiquimod-induced psoriasis-like skin inflammation in mice is mediated via the IL-23/IL-17 axis, Journal of immunology 182(9) (2009) 5836–45. [DOI] [PubMed] [Google Scholar]
  • [16].Sarkar S, Cooney LA, White P, Dunlop DB, Endres J, Jorns JM, Wasco MJ, Fox DA, Regulation of pathogenic IL-17 responses in collagen-induced arthritis: roles of endogenous interferon-gamma and IL-4, Arthritis research & therapy 11(5) (2009) R158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Yang J, Sundrud MS, Skepner J, Yamagata T, Targeting Th17 cells in autoimmune diseases, Trends in pharmacological sciences 35(10) (2014) 493–500. [DOI] [PubMed] [Google Scholar]
  • [18].Papp KA, Leonardi C, Menter A, Ortonne JP, Krueger JG, Kricorian G, Aras G, Li J, Russell CB, Thompson EH, Baumgartner S, Brodalumab, an anti-interleukin-17-receptor antibody for psoriasis, The New England journal of medicine 366(13) (2012) 1181–9. [DOI] [PubMed] [Google Scholar]
  • [19].Jacques P, Van Praet L, Carron P, Van den Bosch F, Elewaut D, Pathophysiology and role of the gastrointestinal system in spondyloarthritides, Rheum Dis Clin North Am 38(3) (2012) 569–82. [DOI] [PubMed] [Google Scholar]
  • [20].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, G. Secukinumab in Crohn’s Disease Study, 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, Gut 61(12) (2012) 1693–700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Genovese MC, Durez P, Richards HB, Supronik J, Dokoupilova E, Mazurov V, Aelion JA, Lee SH, Codding CE, Kellner H, Ikawa T, Hugot S, Mpofu S, Efficacy and safety of secukinumab in patients with rheumatoid arthritis: a phase II, dose-finding, double-blind, randomised, placebo controlled study, Annals of the rheumatic diseases 72(6) (2013) 863–9. [DOI] [PubMed] [Google Scholar]
  • [22].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, Nature immunology 10 (2009) 603. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Esplugues E, Huber S, Gagliani N, Hauser AE, Town T, Wan YY, O’Connor W Jr., Rongvaux A, Van Rooijen N, Haberman AM, Iwakura Y, Kuchroo VK, Kolls JK, Bluestone JA, Herold KC, Flavell RA, Control of TH17 cells occurs in the small intestine, Nature 475(7357) (2011) 514–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Kumar P, Monin L, Castillo P, Elsegeiny W, Horne W, Eddens T, Vikram A, Good M, Schoenborn AA, Bibby K, Montelaro RC, Metzger DW, Gulati AS, Kolls JK, Intestinal Interleukin-17 Receptor Signaling Mediates Reciprocal Control of the Gut Microbiota and Autoimmune Inflammation, Immunity 44(3) (2016) 659–671. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Omenetti S, Bussi C, Metidji A, Iseppon A, Lee S, Tolaini M, Li Y, Kelly G, Chakravarty P, Shoaie S, Gutierrez MG, Stockinger B, The Intestine Harbors Functionally Distinct Homeostatic Tissue-Resident and Inflammatory Th17 Cells, Immunity (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Schwartz S, Beaulieu JF, Ruemmele FM, Interleukin-17 is a potent immuno-modulator and regulator of normal human intestinal epithelial cell growth, Biochemical and biophysical research communications 337(2) (2005) 505–9. [DOI] [PubMed] [Google Scholar]
  • [27].Blaschitz C, Raffatellu M, Th17 cytokines and the gut mucosal barrier, J Clin Immunol 30(2) (2010) 196–203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].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, Nature immunology 13(10) (2012) 991–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Peters A, Lee Y, Kuchroo VK, The many faces of Th17 cells, Current opinion in immunology 23(6) (2011) 702–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Ghoreschi K, Laurence A, Yang XP, Tato CM, McGeachy MJ, Konkel JE, Ramos HL, Wei L, Davidson TS, Bouladoux N, Grainger JR, Chen Q, Kanno Y, Watford WT, Sun HW, Eberl G, Shevach EM, Belkaid Y, Cua DJ, Chen W, O’Shea JJ, Generation of pathogenic T(H)17 cells in the absence of TGF-beta signalling, Nature 467(7318) (2010) 967–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Zhou L, Chong MM, Littman DR, Plasticity of CD4+ T cell lineage differentiation, Immunity 30(5) (2009) 646–55. [DOI] [PubMed] [Google Scholar]
  • [32].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, Nature 441(7090) (2006) 235–8. [DOI] [PubMed] [Google Scholar]
  • [33].Korn T, Mitsdoerffer M, Croxford AL, Awasthi A, Dardalhon VA, Galileos G, Vollmar P, Stritesky GL, Kaplan MH, Waisman A, Kuchroo VK, Oukka M, IL-6 controls Th17 immunity in vivo by inhibiting the conversion of conventional T cells into Foxp3+ regulatory T cells, Proceedings of the National Academy of Sciences of the United States of America 105(47) (2008) 18460–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, McClanahan T, Cua DJ, 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, Nature immunology 8(12) (2007) 1390–7. [DOI] [PubMed] [Google Scholar]
  • [35].Ivanov II, McKenzie BS, Zhou L, Tadokoro CE, Lepelley A, Lafaille JJ, Cua DJ, Littman DR, The orphan nuclear receptor RORgammat directs the differentiation program of proinflammatory IL-17+ T helper cells, Cell 126(6) (2006) 1121–33. [DOI] [PubMed] [Google Scholar]
  • [36].Yang XO, Panopoulos AD, Nurieva R, Chang SH, Wang D, Watowich SS, Dong C, STAT3 regulates cytokine-mediated generation of inflammatory helper T cells, The Journal of biological chemistry 282(13) (2007) 9358–63. [DOI] [PubMed] [Google Scholar]
  • [37].Zhao M, Tan Y, Peng Q, Huang C, Guo Y, Liang G, Zhu B, Huang Y, Liu A, Wang Z, Li M, Gao X, Wu R, Wu H, Long H, Lu Q, IL-6/STAT3 pathway induced deficiency of RFX1 contributes to Th17-dependent autoimmune diseases via epigenetic regulation, Nature communications 9(1) (2018) 583. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Akira S, Roles of STAT3 defined by tissue-specific gene targeting, Oncogene 19(21) (2000) 2607–11. [DOI] [PubMed] [Google Scholar]
  • [39].Spolski R, Leonard WJ, Interleukin-21: basic biology and implications for cancer and autoimmunity, Annual review of immunology 26 (2008) 57–79. [DOI] [PubMed] [Google Scholar]
  • [40].Yang L, Anderson DE, Baecher-Allan C, Hastings WD, Bettelli E, Oukka M, Kuchroo VK, Hafler DA, IL-21 and TGF-beta are required for differentiation of human T(H)17 cells, Nature 454(7202) (2008) 350–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Shi Y, Chen Z, Zhao Z, Yu Y, Fan H, Xu X, Bu X, Gu J, IL-21 Induces an Imbalance of Th17/Treg Cells in Moderate-to-Severe Plaque Psoriasis Patients, Frontiers in immunology 10 (2019) 1865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Coquet JM, Chakravarti S, Smyth MJ, Godfrey DI, Cutting edge: IL-21 is not essential for Th17 differentiation or experimental autoimmune encephalomyelitis, Journal of immunology 180(11) (2008) 7097–101. [DOI] [PubMed] [Google Scholar]
  • [43].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, European journal of immunology 38(7) (2008) 1833–8. [DOI] [PubMed] [Google Scholar]
  • [44].Zenewicz LA, Yancopoulos GD, Valenzuela DM, Murphy AJ, Stevens S, Flavell RA, Innate and adaptive interleukin-22 protects mice from inflammatory bowel disease, Immunity 29(6) (2008) 947–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].Ogawa A, Andoh A, Araki Y, Bamba T, Fujiyama Y, Neutralization of interleukin-17 aggravates dextran sulfate sodium-induced colitis in mice, Clinical immunology 110(1) (2004) 55–62. [DOI] [PubMed] [Google Scholar]
  • [46].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, Nature immunology 10(6) (2009) 603–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Dankers W, Davelaar N, van Hamburg JP, van de Peppel J, Colin EM, Lubberts E, Human Memory Th17 Cell Populations Change Into Anti-inflammatory Cells With Regulatory Capacity Upon Exposure to Active Vitamin D, Frontiers in immunology 10 (2019) 1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Stumhofer JS, Silver JS, Laurence A, Porrett PM, Harris TH, Turka LA, Ernst M, Saris CJ, O’Shea JJ, Hunter CA, Interleukins 27 and 6 induce STAT3-mediated T cell production of interleukin 10, Nature immunology 8(12) (2007) 1363–71. [DOI] [PubMed] [Google Scholar]
  • [49].Wang C, Yosef N, Gaublomme J, Wu C, Lee Y, Clish CB, Kaminski J, Xiao S, Meyer Zu Horste G, Pawlak M, Kishi Y, Joller N, Karwacz K, Zhu C, Ordovas-Montanes M, Madi A, Wortman I, Miyazaki T, Sobel RA, Park H, Regev A, Kuchroo VK, CD5L/AIM Regulates Lipid Biosynthesis and Restrains Th17 Cell Pathogenicity, Cell 163(6) (2015) 1413–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Ivanov II, Atarashi K, Manel N, Brodie EL, Shima T, Karaoz U, Wei D, Goldfarb KC, Santee CA, Lynch SV, Tanoue T, Imaoka A, Itoh K, Takeda K, Umesaki Y, Honda K, Littman DR, Induction of intestinal Th17 cells by segmented filamentous bacteria, Cell 139(3) (2009) 485–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Omenetti S, Bussi C, Metidji A, Iseppon A, Lee S, Tolaini M, Li Y, Kelly G, Chakravarty P, Shoaie S, Gutierrez MG, Stockinger B, The Intestine Harbors Functionally Distinct Homeostatic Tissue-Resident and Inflammatory Th17 Cells, Immunity 51(1) (2019) 77–89 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Chang KK, Liu LB, Jin LP, Zhang B, Mei J, Li H, Wei CY, Zhou WJ, Zhu XY, Shao J, Li DJ, Li MQ, IL-27 triggers IL-10 production in Th17 cells via a c-Maf/RORgammat/Blimp-1 signal to promote the progression of endometriosis, Cell death & disease 8(3) (2017) e2666. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].Aschenbrenner D, Foglierini M, Jarrossay D, Hu D, Weiner HL, Kuchroo VK, Lanzavecchia A, Notarbartolo S, Sallusto F, An immunoregulatory and tissue-residency program modulated by c-MAF in human TH17 cells, Nature immunology 19(10) (2018) 1126–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Carr EL, Kelman A, Wu GS, Gopaul R, Senkevitch E, Aghvanyan A, Turay AM, Frauwirth KA, Glutamine uptake and metabolism are coordinately regulated by ERK/MAPK during T lymphocyte activation, Journal of immunology 185(2) (2010) 1037–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Shi LZ, Wang R, Huang G, Vogel P, Neale G, Green DR, Chi H, HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells, The Journal of experimental medicine 208(7) (2011) 1367–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Johnson MO, Wolf MM, Madden MZ, Andrejeva G, Sugiura A, Contreras DC, Maseda D, Liberti MV, Paz K, Kishton RJ, Johnson ME, de Cubas AA, Wu P, Li G, Zhang Y, Newcomb DC, Wells AD, Restifo NP, Rathmell WK, Locasale JW, Davila ML, Blazar BR, Rathmell JC, Distinct Regulation of Th17 and Th1 Cell Differentiation by Glutaminase-Dependent Metabolism, Cell 175(7) (2018) 1780–1795 e19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [57].Michalek RD, Gerriets VA, Jacobs SR, Macintyre AN, MacIver NJ, Mason EF, Sullivan SA, Nichols AG, Rathmell JC, Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets, Journal of immunology 186(6) (2011) 3299–303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Gulen MF, Kang Z, Bulek K, Youzhong W, Kim TW, Chen Y, Altuntas CZ, Sass Bak-Jensen K, McGeachy MJ, Do JS, Xiao H, Delgoffe GM, Min B, Powell JD, Tuohy VK, Cua DJ, Li X, The receptor SIGIRR suppresses Th17 cell proliferation via inhibition of the interleukin-1 receptor pathway and mTOR kinase activation, Immunity 32(1) (2010) 54–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Deason K, Troutman TD, Jain A, Challa DK, Mandraju R, Brewer T, Ward ES, Pasare C, BCAP links IL-1R to the PI3K-mTOR pathway and regulates pathogenic Th17 cell differentiation, The Journal of experimental medicine 215(9) (2018) 2413–2428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Ren W, Yin J, Duan J, Liu G, Tan B, Yang G, Wu G, Bazer FW, Peng Y, Yin Y, mTORC1 signaling and IL-17 expression: Defining pathways and possible therapeutic targets, European journal of immunology 46(2) (2016) 291–9. [DOI] [PubMed] [Google Scholar]
  • [61].Kaufmann U, Kahlfuss S, Yang J, Ivanova E, Koralov SB, Feske S, Calcium Signaling Controls Pathogenic Th17 Cell-Mediated Inflammation by Regulating Mitochondrial Function, Cell Metab 29(5) (2019) 1104–1118 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Kalim KW, Yang JQ, Li Y, Meng Y, Zheng Y, Guo F, Reciprocal Regulation of Glycolysis-Driven Th17 Pathogenicity and Regulatory T Cell Stability by Cdc42, Journal of immunology 200(7) (2018) 2313–2326. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [63].Shiau MY, Chuang PH, Yang CP, Hsiao CW, Chang SW, Chang KY, Liu TM, Chen HW, Chuang CC, Yuan SY, Chang YH, Mechanism of Interleukin-4 Reducing Lipid Deposit by Regulating Hormone-Sensitive Lipase, Scientific reports 9(1) (2019) 11974. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [64].Ricardo-Gonzalez RR, Red Eagle A, Odegaard JI, Jouihan H, Morel CR, Heredia JE, Mukundan L, Wu D, Locksley RM, Chawla A, IL-4/STAT6 immune axis regulates peripheral nutrient metabolism and insulin sensitivity, Proceedings of the National Academy of Sciences of the United States of America 107(52) (2010) 22617–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Ip WKE, Hoshi N, Shouval DS, Snapper S, Medzhitov R, Anti-inflammatory effect of IL-10 mediated by metabolic reprogramming of macrophages, Science 356(6337) (2017) 513–519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Xu WD, Xie QB, Zhao Y, Liu Y, Association of Interleukin-23 receptor gene polymorphisms with susceptibility to Crohn’s disease: A meta-analysis, Scientific reports 5 (2015) 18584. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Nair RP, Ruether A, Stuart PE, Jenisch S, Tejasvi T, Hiremagalore R, Schreiber S, Kabelitz D, Lim HW, Voorhees JJ, Christophers E, Elder JT, Weichenthal M, Polymorphisms of the IL12B and IL23R genes are associated with psoriasis, The Journal of investigative dermatology 128(7) (2008) 1653–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Ahern PP, Schiering C, Buonocore S, McGeachy MJ, Cua DJ, Maloy KJ, Powrie F, Interleukin-23 drives intestinal inflammation through direct activity on T cells, Immunity 33(2) (2010) 279–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].McGeachy MJ, Chen Y, Tato CM, Laurence A, Joyce-Shaikh B, Blumenschein WM, McClanahan TK, O’Shea JJ, Cua DJ, The interleukin 23 receptor is essential for the terminal differentiation of interleukin 17-producing effector T helper cells in vivo, Nature immunology 10(3) (2009) 314–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Gaffen SL, Jain R, Garg AV, Cua DJ, The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing, Nature reviews. Immunology 14(9) (2014) 585–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Lee JS, Tato CM, Joyce-Shaikh B, Gulen MF, Cayatte C, Chen Y, Blumenschein WM, Judo M, Ayanoglu G, McClanahan TK, Li X, Cua DJ, Interleukin-23-Independent IL-17 Production Regulates Intestinal Epithelial Permeability, Immunity 43(4) (2015) 727–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Acosta-Rodriguez EV, Napolitani G, Lanzavecchia A, Sallusto F, Interleukins 1beta and 6 but not transforming growth factor-beta are essential for the differentiation of interleukin 17-producing human T helper cells, Nature immunology 8(9) (2007) 942–9. [DOI] [PubMed] [Google Scholar]
  • [73].Wilson NJ, Boniface K, Chan JR, McKenzie BS, Blumenschein WM, Mattson JD, Basham B, Smith K, Chen T, Morel F, Lecron JC, Kastelein RA, Cua DJ, McClanahan TK, Bowman EP, de Waal Malefyt R, Development, cytokine profile and function of human interleukin 17-producing helper T cells, Nature immunology 8(9) (2007) 950–7. [DOI] [PubMed] [Google Scholar]
  • [74].Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, McClanahan T, Kastelein RA, Cua DJ, IL-23 drives a pathogenic T cell population that induces autoimmune inflammation, The Journal of experimental medicine 201(2) (2005) 233–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Stritesky GL, Yeh N, Kaplan MH, IL-23 promotes maintenance but not commitment to the Th17 lineage, Journal of immunology 181(9) (2008) 5948–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Jain R, Chen Y, Kanno Y, Joyce-Shaikh B, Vahedi G, Hirahara K, Blumenschein WM, Sukumar S, Haines CJ, Sadekova S, McClanahan TK, McGeachy MJ, O’Shea JJ, Cua DJ, Interleukin-23-Induced Transcription Factor Blimp-1 Promotes Pathogenicity of T Helper 17 Cells, Immunity 44(1) (2016) 131–142. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].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, Nature immunology 12(6) (2011) 568–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [78].Komuczki J, Tuzlak S, Friebel E, Hartwig T, Spath S, Rosenstiel P, Waisman A, Opitz L, Oukka M, Schreiner B, Pelczar P, Becher B, Fate-Mapping of GM-CSF Expression Identifies a Discrete Subset of Inflammation-Driving T Helper Cells Regulated by Cytokines IL-23 and IL-1beta, Immunity 50(5) (2019) 1289–1304 e6. [DOI] [PubMed] [Google Scholar]
  • [79].Codarri L, Gyulveszi G, Tosevski V, Hesske L, Fontana A, Magnenat L, Suter T, Becher B, RORgammat drives production of the cytokine GM-CSF in helper T cells, which is essential for the effector phase of autoimmune neuroinflammation, Nature immunology 12(6) (2011) 560–7. [DOI] [PubMed] [Google Scholar]
  • [80].Sonderegger I, Iezzi G, Maier R, Schmitz N, Kurrer M, Kopf M, GM-CSF mediates autoimmunity by enhancing IL-6-dependent Th17 cell development and survival, The Journal of experimental medicine 205(10) (2008) 2281–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].Zhang GX, Gran B, Yu S, Li J, Siglienti I, Chen X, Kamoun M, Rostami A, Induction of experimental autoimmune encephalomyelitis in IL-12 receptor-beta 2-deficient mice: IL-12 responsiveness is not required in the pathogenesis of inflammatory demyelination in the central nervous system, Journal of immunology 170(4) (2003) 2153–60. [DOI] [PubMed] [Google Scholar]
  • [82].Chitnis T, Najafian N, Benou C, Salama AD, Grusby MJ, Sayegh MH, Khoury SJ, Effect of targeted disruption of STAT4 and STAT6 on the induction of experimental autoimmune encephalomyelitis, The Journal of clinical investigation 108(5) (2001) 739–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Becher B, Durell BG, Noelle RJ, Experimental autoimmune encephalitis and inflammation in the absence of interleukin-12, The Journal of clinical investigation 110(4) (2002) 493–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [84].Xu J, Yang Y, Qiu G, Lal G, Yin N, Wu Z, Bromberg JS, Ding Y, Stat4 is critical for the balance between Th17 cells and regulatory T cells in colitis, Journal of immunology 186(11) (2011) 6597–606. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Lee PW, Smith AJ, Yang Y, Selhorst AJ, Liu Y, Racke MK, Lovett-Racke AE, IL-23R-activated STAT3/STAT4 is essential for Th1/Th17-mediated CNS autoimmunity, JCI insight 2(17) (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Harbour SN, Maynard CL, Zindl CL, Schoeb TR, Weaver CT, Th17 cells give rise to Th1 cells that are required for the pathogenesis of colitis, Proceedings of the National Academy of Sciences of the United States of America 112(22) (2015) 7061–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Haugh IM, Preston AK, Kivelevitch DN, Menter AM, Risankizumab: an anti-IL-23 antibody for the treatment of psoriasis, Drug Des Devel Ther 12 (2018) 3879–3883. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Dolgin E, New anti-IL-23 drugs raise hopes for psoriasis plaque clearance, Nat Biotechnol 34(12) (2016) 1218–1219. [DOI] [PubMed] [Google Scholar]
  • [89].Koutruba N, Emer J, Lebwohl M, Review of ustekinumab, an interleukin-12 and interleukin-23 inhibitor used for the treatment of plaque psoriasis, Ther Clin Risk Manag 6 (2010) 123–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Siebert S, Millar NL, McInnes IB, Why did IL-23p19 inhibition fail in AS: a tale of tissues, trials or translation?, Annals of the rheumatic diseases (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Sands BE, Chen J, Feagan BG, Penney M, Rees WA, Danese S, Higgins PDR, Newbold P, Faggioni R, Patra K, Li J, Klekotka P, Morehouse C, Pulkstenis E, Drappa J, van der Merwe R, Gasser RA Jr., Efficacy and Safety of MEDI2070, an Antibody Against Interleukin 23, in Patients With Moderate to Severe Crohn’s Disease: A Phase 2a Study, Gastroenterology 153(1) (2017) 77–86 e6. [DOI] [PubMed] [Google Scholar]
  • [92].Longbrake EE, Racke MK, Why did IL-12/IL-23 antibody therapy fail in multiple sclerosis?, Expert Rev Neurother 9(3) (2009) 319–21. [DOI] [PubMed] [Google Scholar]
  • [93].Vollmer TL, Wynn DR, Alam MS, Valdes J, A phase 2, 24-week, randomized, placebo-controlled, double-blind study examining the efficacy and safety of an anti-interleukin-12 and −23 monoclonal antibody in patients with relapsing-remitting or secondary progressive multiple sclerosis, Mult Scler 17(2) (2011) 181–91. [DOI] [PubMed] [Google Scholar]
  • [94].Baeten D, Ostergaard M, Wei JC, Sieper J, Jarvinen P, Tam LS, Salvarani C, Kim TH, Solinger A, Datsenko Y, Pamulapati C, Visvanathan S, Hall DB, Aslanyan S, Scholl P, Padula SJ, Risankizumab, an IL-23 inhibitor, for ankylosing spondylitis: results of a randomised, double-blind, placebo-controlled, proof-of-concept, dose-finding phase 2 study, Annals of the rheumatic diseases 77(9) (2018) 1295–1302. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Zhang S, Takaku M, Zou L, Gu AD, Chou WC, Zhang G, Wu B, Kong Q, Thomas SY, Serody JS, Chen X, Xu X, Wade PA, Cook DN, Ting JPY, Wan YY, Reversing SKI-SMAD4-mediated suppression is essential for TH17 cell differentiation, Nature 551(7678) (2017) 105–109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Tu E, Chia CPZ, Chen W, Zhang D, Park SA, Jin W, Wang D, Alegre ML, Zhang YE, Sun L, Chen W, T Cell Receptor-Regulated TGF-beta Type I Receptor Expression Determines T Cell Quiescence and Activation, Immunity 48(4) (2018) 745–759 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Massague J, Blain SW, Lo RS, TGFbeta signaling in growth control, cancer, and heritable disorders, Cell 103(2) (2000) 295–309. [DOI] [PubMed] [Google Scholar]
  • [98].Moustakas A, Souchelnytskyi S, Heldin CH, Smad regulation in TGF-beta signal transduction, J Cell Sci 114(Pt 24) (2001) 4359–69. [DOI] [PubMed] [Google Scholar]
  • [99].Tengroth L, Arebro J, Larsson O, Bachert C, Georen SK, Cardell LO, Activation of Activin receptor-like kinases curbs mucosal inflammation and proliferation in chronic rhinosinusitis with nasal polyps, Scientific reports 8(1) (2018) 1561. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [100].Xu Q, Jin X, Zheng M, Rohila D, Fu G, Wen Z, Lou J, Wu S, Sloan R, Wang L, Hu H, Gao X, Lu L, Phosphatase PP2A is essential for TH17 differentiation, Proceedings of the National Academy of Sciences of the United States of America 116(3) (2019) 982–987. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [101].Brustle A, Heink S, Huber M, Rosenplanter 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, Nature immunology 8(9) (2007) 958–66. [DOI] [PubMed] [Google Scholar]
  • [102].Schraml BU, Hildner K, Ise W, Lee WL, Smith WA, Solomon B, Sahota G, Sim J, Mukasa R, Cemerski S, Hatton RD, Stormo GD, Weaver CT, Russell JH, Murphy TL, Murphy KM, The AP-1 transcription factor Batf controls T(H)17 differentiation, Nature 460(7253) (2009) 405–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [103].Huber M, Lohoff M, IRF4 at the crossroads of effector T-cell fate decision, European journal of immunology 44(7) (2014) 1886–95. [DOI] [PubMed] [Google Scholar]
  • [104].Gaublomme JT, Yosef N, Lee Y, Gertner RS, Yang LV, Wu C, Pandolfi PP, Mak T, Satija R, Shalek AK, Kuchroo VK, Park H, Regev A, Single-Cell Genomics Unveils Critical Regulators of Th17 Cell Pathogenicity, Cell 163(6) (2015) 1400–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [105].Dang EV, Barbi J, Yang HY, Jinasena D, Yu H, Zheng Y, Bordman Z, Fu J, Kim Y, Yen HR, Luo W, Zeller K, Shimoda L, Topalian SL, Semenza GL, Dang CV, Pardoll DM, Pan F, Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1, Cell 146(5) (2011) 772–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [106].Chou TF, Chuang YT, Hsieh WC, Chang PY, Liu HY, Mo ST, Hsu TS, Miaw SC, Chen RH, Kimchi A, Lai MZ, Tumour suppressor death-associated protein kinase targets cytoplasmic HIF-1alpha for Th17 suppression, Nature communications 7 (2016) 11904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [107].Meyer Zu Horste G, Wu C, Wang C, Cong L, Pawlak M, Lee Y, Elyaman W, Xiao S, Regev A, Kuchroo VK, RBPJ Controls Development of Pathogenic Th17 Cells by Regulating IL-23 Receptor Expression, Cell reports 16(2) (2016) 392–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [108].Kishi Y, Kondo T, Xiao S, Yosef N, Gaublomme J, Wu C, Wang C, Chihara N, Regev A, Joller N, Kuchroo VK, Protein C receptor (PROCR) is a negative regulator of Th17 pathogenicity, The Journal of experimental medicine 213(11) (2016) 2489–2501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [109].Ichiyama K, Gonzalez-Martin A, Kim BS, Jin HY, Jin W, Xu W, Sabouri-Ghomi M, Xu S, Zheng P, Xiao C, Dong C, The MicroRNA-183-96-182 Cluster Promotes T Helper 17 Cell Pathogenicity by Negatively Regulating Transcription Factor Foxo1 Expression, Immunity 44(6) (2016) 1284–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Laine A, Martin B, Luka M, Mir L, Auffray C, Lucas B, Bismuth G, Charvet C, Foxo1 Is a T Cell-Intrinsic Inhibitor of the RORgammat-Th17 Program, Journal of immunology 195(4) (2015) 1791–803. [DOI] [PubMed] [Google Scholar]
  • [111].Shimada K, Porritt RA, Markman JL, O’Rourke JG, Wakita D, Noval Rivas M, Ogawa C, Kozhaya L, Martins GA, Unutmaz D, Baloh RH, Crother TR, Chen S, Arditi M, T-Cell-Intrinsic Receptor Interacting Protein 2 Regulates Pathogenic T Helper 17 Cell Differentiation, Immunity 49(5) (2018) 873–885 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [112].Wu B, Zhang S, Guo Z, Wang G, Zhang G, Xie L, Lou J, Chen X, Wu D, Bergmeier W, Zheng J, Wan YY, RAS P21 Protein Activator 3 (RASA3) Specifically Promotes Pathogenic T Helper 17 Cell Generation by Repressing T-Helper-2-Cell-Biased Programs, Immunity 49(5) (2018) 886–898 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [113].Stadhouders R, Lubberts E, Hendriks RW, A cellular and molecular view of T helper 17 cell plasticity in autoimmunity, J Autoimmun 87 (2018) 1–15. [DOI] [PubMed] [Google Scholar]
  • [114].Christensen D, Mortensen R, Rosenkrands I, Dietrich J, Andersen P, Vaccine-induced Th17 cells are established as resident memory cells in the lung and promote local IgA responses, Mucosal immunology 10(1) (2017) 260–270. [DOI] [PubMed] [Google Scholar]
  • [115].Amezcua Vesely MC, Pallis P, Bielecki P, Low JS, Zhao J, Harman CCD, Kroehling L, Jackson R, Bailis W, Licona-Limon P, Xu H, Iijima N, Pillai PS, Kaplan DH, Weaver CT, Kluger Y, Kowalczyk MS, Iwasaki A, Pereira JP, Esplugues E, Gagliani N, Flavell RA, Effector TH17 Cells Give Rise to Long-Lived TRM Cells that Are Essential for an Immediate Response against Bacterial Infection, Cell 178(5) (2019) 1176–1188 e15. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES