Abstract
Systemic sclerosis is characterized by widespread fibrosis of the skin and internal organs, vascular impairment, and dysregulation of innate and adaptive immune system. Growing evidence indicates that T-cell proliferation and cytokine secretion play a major role in the initiation of systemic sclerosis, but the role of T helper 17 cells and of interleukin-17 cytokines in the development and progression of the disease remains controversial. In particular, an equally distributed body of literature supports both pro-fibrotic and anti-fibrotic effects of interleukin-17, suggesting a complex and nuanced role of this cytokine in systemic sclerosis pathogenesis that may vary depending on disease stage, target cells in affected organs, and inflammatory milieu. Although interleukin-17 already represents an established therapeutic target for several immune-mediated inflammatory diseases, more robust experimental evidence is required to clarify whether it may become an attractive therapeutic target for systemic sclerosis as well.
Keywords: Systemic sclerosis, interleukin-17, fibrosis, T helper 17, fibroblast
Introduction
Systemic sclerosis (SSc) is an autoimmune connective tissue disease of unknown etiology characterized by widespread fibrosis of the skin and internal organs, vascular impairment, and dysregulation of innate and adaptive immune system.1,2 SSc has an unpredictable course, and available treatments only delay disease progression. In analogy to other autoimmune disorders, environmental, genetic, and immunological factors are all involved in the pathogenesis of SSc, but their relative contribution to the spectrum of scleroderma manifestations is not fully clarified. In view of its interplay with the immune system, the human microbiota also represents a potential player in the onset and progression of SSc. 3
Growing evidence indicates that T-cell proliferation and cytokine secretion play a major role in the initiation of SSc,4–6 suggesting that T lymphocytes may orchestrate disease pathogenesis and the characteristic tissue fibrosis. 5 In particular, T helper 17 (Th17) cells represent a subset of T lymphocytes of major interest in SSc because of established roles in different fibro-inflammatory disorders. Th17 cells are increased in the peripheral blood of patients with SSc, accumulate at disease sites, and secrete the pro-inflammatory cytokine interleukin (IL)-17.7,8 Yet, the exact contribution of Th17 cells and IL-17 to disease pathogenesis—whether pro-fibrotic or anti-fibrotic—remains a matter of debate.9–11
Th17 cells may, in fact, display functionally distinctive features, and their inflammatory potential can be largely influenced by a number of environmental and immunological triggers. Looking at the gastrointestinal tract and at the commensal flora, for instance, non-inflammatory Th17 cells and IL-17A production are elicited by segmented filamentous bacteria via the engagement of the ILC3/IL-22/SAA1/2 axis, while Th17 cells with pro-inflammatory activity are induced by Citrobacter species. 12 Hence, because of these nuanced inflammatory properties, it is possible that the role of Th17 cells and IL-17 in the pathogenesis of SSc and tissue fibrosis is not uniformly stimulatory or inhibitory, and likely depends on a complex and tightly regulated network of signals yet to be identified.
In the present review, we aim to discuss the most recent advances in our understanding of the contribution of Th17 cells in the pathogenesis of SSc, with particular focus on the debated pro-fibrotic and anti-fibrotic role of IL-17. A brief overview of IL-17 biology and of the potential therapeutic implications of blocking the IL-17 pathway will be also provided.
Immunobiology of IL-17
The IL-17 family comprises six cytokines: IL-17A (also named as IL-17, the first discovered and most studied member), IL17B, IL-17C, IL-17D (known as IL-27), IL-17E (also named as IL-25), and IL-17F.13–15 IL-17 cytokines bind to their receptors (IL-17Rs) on target cells and exert pleiotropic functions13,14,16,17 being involved in the sustainment of inflammation, autoimmunity, and tissue repair.16,17 The IL-17Rs consist of five single-pass transmembrane receptors with conserved domains: IL-17RA, IL-17RB, IL-17RC, IL-17RD, and IL-17RE. 18 Functionally, the IL-17R is a heterodimeric complex composed of the IL-17RA subunit in combination with other subunits. IL-17A as well as IL-17F signal through the IL-17RA/IL-17RC heterodimer, but the IL-17A interaction with its receptors appears to be stronger. 18 IL-17RA is also used by IL-17B, IL-17C, and IL-17E. Of note, while the IL-17RA subunit is ubiquitously expressed, IL-17RC is mainly present on non-hematopoietic epithelial and mesenchymal cells. IL-17 exerts its in vivo functions through canonical and non-canonical signaling pathways. Canonical signaling induces both transcriptional and post-transcriptional mechanisms involved in autoimmunity and metabolic reprogramming of lymphoid tissues. Non-canonical signaling acts in synergy with other receptor systems—including transforming growth factor β (TGFβ), interferon γ (IFNγ), and tumor necrosis factor α (TNFα)—and is mainly responsible for tissue repair and regeneration. 18
IL-17 is primarily produced by Th17 cells, but several other cell types are able to produce it such as gamma delta (γδ) T cells, invariant natural killer T cells, group 3 innate lymphoid cells, natural killer cells, double-negative T cells, B lymphocytes, and mast cells.7,8,19,20 Th17 cells arise from naive CD4+ T cells, and their maturation is influenced by TGFβ, IL-6, and IL-1β.13,21 Conversely, cytokines secreted by Th1 and Th2 cells, such as IFN-c and IL-4, respectively, exert an inhibitory action on Th17 cells. 21 IL-23 is also crucial for Th17 differentiation, and the IL-23/IL-17 axis has been long regarded as a redundant pathway for perpetuating chronic inflammation. 21 Indeed, IL-23 has been shown to cause inflammation through IL-17-dependent and IL-17-independent pathways, increasing the complexity around Th17 pathophysiology and IL-17 signaling. 22 Th17 cells synthesize the IL-17A and IL-17F isoforms of IL-17 as well as additional pro-inflammatory cytokines such as IL-21 and IL-22. 14 Dual role of this cytokine armamentarium is to perpetuate inflammation via IL-17 and IL-22, and to inhibit regulatory T cells (Tregs) through IL-21, thus sustaining inflammatory responses. 14 In view of these pro-inflammatory properties, Th17 cells participate in a number of physiological processes including (1) neutrophil recruitment in inflammatory niches; (2) response to fungi, mycobacteria, and viruses; and (3) remodeling of the extracellular matrix (ECM).13,15 Th17 cells have been also implicated in autoimmune disorders such as psoriasis and inflammatory bowel diseases, but conflicting data exist regarding their engagement in SSc. 23 Similarly, regardless of its cellular source, the relevance of IL-17 in the pathogenesis of scleroderma remains to be fully addressed and the question whether it would be beneficial or detrimental to neutralize it in this specific pathological setting needs to be unequivocally answered. Animal models of scleroderma, in fact, essentially support a pro-fibrotic and a pro-inflammatory activity of IL-17A, but in vitro studies point toward an anti-fibrotic action.24,25 Yet, although experiments with human fibroblasts in plastic plates have intrinsic limitations due to nonphysiological conditions of cell culture and, hence, result interpretation, in vivo murine models also require careful consideration because of intrinsic differences between humans and rodents with regard to immunological responses and fibrogenesis.
Evidence in favor of a pro-fibrotic role of IL-17 in SSc
Involvement of IL-17 and of its downstream pathways in the pathogenesis of SSc has been first hypothesized following the accumulation of experimental evidence showing the pro-fibrotic effects of this cytokine on a series of organ-specific chronic disorders.26,27 In experimental models of liver cirrhosis, idiopathic pulmonary fibrosis, and heart failure, in fact, IL-17 participates in fibrogenesis via different mechanisms including direct activation of resident stromal cells and amplification of the inflammatory response.
In liver fibrosis, IL-17 and IL-17-producing cells contribute to disease pathology both by perpetuating inflammation in synergy with IL-1β, IL-6, and IL-23, and by inducing the expression of TGFβ and TGFβ receptor by hepatic stellate cells.28–31 IL-17 also promotes the release of periostin by hepatocytes leading to fibroblast activation and collagen production. 32 Accordingly, inactivation of IL-17 signaling in hepatocytes reduces liver fibrosis in mouse models of hepatitis-induced liver injury. 33 In the bronchial mucosa of patients with moderate-to-severe asthma, IL-17 is highly expressed and drives epithelial-to-mesenchymal transition when used to stimulate human small airway epithelial cells in in vitro cultures.34,35 In patients with idiopathic pulmonary fibrosis as well as in bleomycin-induced murine models of lung fibrosis, Th17 cells increase the production of TGFβ and IL-17A, and co-culture of these cells with human lung fibroblasts results in increased deposition of collagen and other ECM proteins. 36 Indeed, blocking IL-17 ameliorates lung fibrosis in murine models of post-bone marrow transplant pulmonary disease. 37 Finally, IL-17 produced by γδ T cells and Th17 cells plays a role in different models of cardiac fibrosis, likely representing a central pro-fibrotic mediator of multiple mechanical, toxic, and inflammatory insults in these organs. IL-17, in fact, activates cardiac myofibroblasts in ischemia- or isoproterenol-induced mouse models of heart failure as well as in spontaneous or angiotensin II–induced models of hypertension.38–44 Blockade of IL-17, of its receptor, or of IL-17 signaling in these experimental scenarios all reduces cardiac fibrosis along with improvement of cardiac contractile function.38–44
Initial observation in support of a pathogenic role of IL-17 in SSc came from the pathological evidence of IL-17 producing cells in the inflammatory infiltrate of scleroderma lesions, including Th17 cells, topoisomerase-specific CD4+ Th cells, and γδ T cells.27,45–47 Circulating Th17 cells and serum IL-17 concentrations were also found to be significantly increased in active scleroderma patients compared to healthy controls, and to correlate with disease activity as defined by the Valentini score.48–50 Yet, when addressing direct pro-fibrotic effects of IL-17, only two in vitro studies out of a dozen were able to show a stimulatory activity on fibroblast growth and collagen production.48,51 Most in vitro studies, in fact, seem to support a general pro-inflammatory role of IL-17 in SSc rather than an intrinsic pro-fibrotic one. In particular, when stimulated with IL-17A, dermal fibroblasts as well as endothelial cells increase the expression of adhesion molecules and the production of a variety of pro-inflammatory mediators such as chemokine ligand (CCL)2, CCL8, IL-1, IL-6, and matrix metalloproteinase 1 (MMP1).25,27,48,49,52,53 Indeed, these cytokines and chemokines are highly expressed in the skin of scleroderma patients, enhance the recruitment of inflammatory cells, and ultimately amplify pro-fibrotic responses in fibroblasts.54–56
Various mouse models of SSc also indicate that IL-17 consistently favors the development of tissue fibrosis primarily through the amplification of a fibro-inflammatory response. Lung fibrosis induced by Saccharopolyspora rectivirgula and bleomycin, for instance, was attenuated in IL-17−/− mice with less collagen deposition and expression of ECM-associated genes.36,57 In the bleomycin-induced murine model of cutaneous scleroderma, administration of bleomycin increased the recruitment of Th17 cells to the skin as well as the local secretion of IL-17. 58 Of note, IL-21 and B cell activating factor (BAFF)—two key cytokines in the proliferation and activation of T and B cells—also seemed to boost IL-17-mediated fibrosis downstream to IL-17 in this model.59,60 In addition, IL-17 both promoted collagen deposition and prevented collagen degradation in a TGFβ-independent manner. 61 These findings were confirmed in the “tight skin mouse 1” (TSK-1) mouse model, wherein blockade of IL-17 reduced the severity of skin fibrosis. 62 In other models of IL-1 receptor antagonist deficient and “graft versus host disease” murine models of scleroderma, severe dermal and pulmonary fibrosis induced by bleomycin or splenocyte injection was reversed by knocking down IL-17. 58 Similarly, IL-1β-treated IL-17A-deficient mice showed reduced pulmonary inflammation and fibrosis, and delivery of IL-17A resulted in increased TGFβ-dependent collagen deposition, suggesting that IL-1β and IL-17 exerted a synergistic effect on the expression of pro-fibrotic and inflammatory mediators. 36
Taken together, this in vitro and in vivo evidence indicates that IL-17 clearly contributes to SSc pathogenesis and, specifically, to the characteristic fibrotic outcomes but likely sustains tissue fibrosis indirectly by amplifying inflammation and by enhancing TGFβ-driven collagen secretion. Indeed, pro-inflammatory properties of IL-17 are well established in literature and fibroblasts are known to respond to IL-17 by increasing the expression of inflammatory mediators, adhesion molecules, and MMPs.25,27,48,49,52,53 In theory, IL-17 may also contribute to SSc pathogenesis beside its effects on stromal cells by boosting the production of autoantibodies.63,64 By enhancing the retention of B cells in germinal centers, in fact, IL-17 can favor the emergence of autoreactive clones and the generation of high-affinity autoantibodies with pro-fibrotic activity.63,64
In conclusion, a body of experimental data sustain a role of IL-17 in the initiation and progression of scleroderma. However, two major points need to be stressed when considering translation of these results from murine models of SSc to humans. On one hand, in fact, because most of these models are focused on skin involvement, it is possible that different IL-17-mediated mechanisms—such as those described in the context of heart failure and liver and pulmonary fibrosis—may contribute to tissue fibrosis in the multiple organs potentially affected by SSc depending on the inflammatory context and on the distribution of IL-17 receptors on resident cells. On the other hand, because most evidence obtained in murine models points toward an indirect role of IL-17 in modulating tissue fibrosis, partial to no amelioration of clinical outcomes should be taken into account when targeting only this cytokine within a complex system of redundant and interconnected pathways.
Evidence in favor of an anti-fibrotic role of IL-17 in SSc
The role of IL-17 in SSc is complex, incompletely understood, and controversial. Although there is consistent evidence regarding the pro-fibrotic role of this cytokine, there are also data suggesting an opposite, anti-fibrotic effect. The role of IL-17 in regulating ECM expression is yet unexplained.
In a 2012 study, Nakashima et al. 65 compared IL-17A, IL-17 receptor (IL-17RA), and IL-17F expression in patients with SSc and healthy controls. In addition to serum measurements, skin biopsies obtained from patients with SSc, dermatopolymyositis, and healthy individuals were analyzed. The results showed an increased level of serum IL-17A in patients with SSc compared to healthy subjects, while IL-17F levels were similar among different conditions. Furthermore, there was no correlation between IL-17A and IL-17F levels in individual patients. Correlations between IL-17A levels and certain clinical features have also been identified. In patients with an elevated concentration of IL-17A, the prevalence of pitting scars was significantly higher compared to patients with normal levels of IL-17A. The authors also identified increased expression of IL-17A mRNA in the affected skin of patients with SSc (but not in the normal skin of these patients) compared to controls, both in the superficial and deep dermis. However, this was not observed for IL-17F mRNA. Expression of IL-17RA in the skin of patients with SSc both in vitro (in fibroblast cultures) and in vivo (on skin biopsies) was lower than that observed in healthy individuals. In particular, in vitro expression of IL-17RA in SSc fibroblasts was downregulated by TGFβ1 signaling, and IL-17A levels increased as a result of a negative-feedback mechanism. This led to an increased accumulation of collagen and to fibrosis. Another observation of this study was that the expression of connective tissue growth factor (CTGF) mRNA—a factor involved in the induction of fibrosis and the pathogenesis of SSc—decreased under the influence of IL-17A but not of IL-17F. 65
Although several studies have shown a higher Th17 cell count and increased IL-17 expression in the affected skin of patients with SSc compared to healthy subjects, it is noteworthy that no differences were observed in IL-17 expression between patients with diffuse or limited forms of the disease or between those with early forms or those with late forms.52,65 There are also several studies that have shown that the modified Rodnan skin thickness score (mRSS) is lower in patients with elevated serum levels of IL-17A.52,66
Other arguments in favor of the anti-fibrotic role of IL-17A are provided by data suggesting that the synthesis of α1 (I) collagen by fibroblasts is reduced under the influence of this cytokine. 65 In particular, microRNA analysis showed that by increasing miR-129-5p—an inhibitor of α1 (I) collagen SSc fibroblasts—IL-17A decreases collagen expression. 67 These effects were not observed using IL-17F. 65 IL-17A also reduces the differentiation of fibroblasts into myofibroblasts under the action of TGFβ and directly stimulates the production of MMP1 by dermal fibroblasts.52,68
In addition to type I collagen and other components of the ECM, activated fibroblasts in the skin of patients with SSc, especially those in the deep dermis, express α-smooth muscle actin (α-SMA). 69 Data have shown that IL-17A does not induce the expression of this protein, but inhibits the stimulatory effect of TGFβ, both in fibroblasts of patients with SSc and in healthy individuals, 52 suggesting a direct negative influence on dermal fibrosis. 52 Whether IL-17 increases the expression of type I and type III procollagen mRNA in human fibroblast cultures has not been unequivocally demonstrated. 27 By stimulating the synthesis of MMP1 by dermal fibroblasts, but not type I collagen, IL-17A also increases turnover of the ECM and alters collagen metabolism in SSc. 70
Skin homeostasis is tightly regulated by the cooperation of keratinocytes and fibroblasts. Disruption of this homeostasis can lead to fibrosis. 71 Recent data from a study that used an original organotypic skin model proposed that IL-17A may influence the interaction between keratinocytes and fibroblasts in SSc. 72 Under the influence of IL-17 and keratinocyte-conditioned media, in fact, fibroblasts from patients with SSc increased MMP1 production and ECM degradation. This effect was reversed by the addition of TGFβ, supporting opposite functions of IL-17 and TGFβ in this specific model. 72 The authors concluded that despite its strong pro-inflammatory effect, IL-17 also has anti-fibrotic effects, which may be of particular importance in the pathogenesis of SSc.
All together, the above evidence suggests that IL-17A can play a dual role in inflammation (Figure 1). Although there is a body of literature supporting its pro-fibrotic effects, IL-17A appears at the same time able to limit, if not fully prevent, fibrosis in SSc patients.70,73,74
Figure 1.
Dual effect of IL-17 in the pathogenesis of SSc: pro-fibrotic and anti-fibrotic. IL-17A is secreted by different immune cell types but prominently by Th17 cells. IL-17A may instruct plasma cell production of autoantibodies and, indirectly, activate quiescent fibroblasts.
IL-17A: interleukin-17A; CCL: chemokine (C-C motif) ligand; MMP: matrix metalloproteinase; IL-6: interleukin-6; IL-1: interleukin-1; CTGF: connective tissue growth factor; TGF: transforming growth factor; IL-17RA: interleukin-17 receptor; SMA: smooth muscle actin; ECM: extracellular matrix, ILC3: innate L-cell3; NK: natural killer; EMT: epithelial-mesenchymal transition.
Treatment perspectives
IL-17 represents an interesting therapeutic target for autoimmune diseases, and its direct neutralization is currently approved for treating psoriasis, psoriatic arthritis, inflammatory bowel diseases, and ankylosing spondylitis.75–78 Indeed, IL-17 is also indirectly targeted by other immunosuppressive drugs already available in immunological settings, thus widening their anti-inflammatory activity. Anti-TNFα therapy, for instance, decreases IL-17 in rheumatoid arthritis and ankylosing spondylitis79–81 by inducing Treg cells and by modulating the IL-6 signaling.82,83 Rituximab does not affect TNFα or Treg cell responses, but strongly reduces IL-17 and IL-22 levels in patient sera. 84 Allogeneic adipose-derived stem cells (ADSCs) in patients with active systemic lupus erythematosus reduced the number of Th17 cells and their ability to release IL-17. 85 On the other hand, conflicting data have been reported on the ability of methotrexate to modify IL-17 levels. 86
As reported above, IL-17 represents a therapeutic target of great interest also for SSc, and modulation of Th17 cells and IL17 pathways correlate with the clinical response to currently used therapeutic strategies. Truchetet et al., 87 for instance, showed that prostacyclin analogs significantly increase IL-17A and IL-22 production in vitro and the frequency of Th17 cells in vivo. Yet, clinical information regarding the effects of targeting IL-17 in patients with SSc is still limited. The promising results obtained with tocilizumab in SSc and the comparative experience of anti-IL-17 and IL-6 monoclonal antibodies in other immune-mediated disorders may, indeed, anticipate a potential argument against the utility of blocking IL-17 in SSc.88–90 Targeting IL-17, in fact, did not prove effective in diseases responsive to anti-IL-6 biologic agents such as rheumatoid arthritis, while showing positive results in disorders that failed to respond to anti-IL-6 therapy such as psoriatic arthritis.91–93 At present, there are two ongoing phase I (Clinicaltrials.gov identifier: NCT04368403) and phase III (Clinicaltrials.gov identifier: NCT03957681) clinical trials assessing IL-17 inhibition in SSc with brodalumab. Brodalumab is a human anti-IL-17 receptor monoclonal antibody binding to human IL-17RA and blocking the biological activity of IL-17A and IL-17F.94,95 The phase I trial is an open-label Japanese study of 210-mg subcutaneous brodalumab every 2 weeks in patients with moderate to severe progressive skin thickening and an mRSS of 10–30 but without severe lung disease. The primary and secondary aims of the study are to evaluate serum concentrations of brodalumab at different intervals until completion and to assess changes in the mRSS compared to baseline, respectively. The phase III trial is a placebo-controlled, double-blind comparative Japanese study of brodalumab with an open-label extension period in SSc patients with moderate to severe skin thickening. The primary aim of the study is to assess changes in the mRSS at week 24 and 52. Results of both trials are expected to be released in 2024. The two other anti-IL-17 drugs available on the market (ixekizumab and secukinumab) have not been tested in SSc yet. Looking ahead, because of the central role of IL-23 in the development of Th17 cells and in view of the non-redundant and independent functions of these two cytokines, dual inhibition of both IL-23 and IL-17 could offer even greater efficacy for treating SSc relative to targeting IL-17 alone.
Finally, due to potential influence of microbiota on the immune system and on IL-17 production, the efficacy of probiotics on gastrointestinal symptoms and immune responses in SSc patients has been recently evaluated. Maringhela TF et al. reported that the use of probiotics reduced Th17 cells in patients with SSc after 8 weeks of treatment. This finding indicates a possible effect of probiotics on the systemic immune system and might represent an innovative therapeutic approach for patients with SSc. 96
Conclusive remarks
Dissecting the pleiotropic role of IL-17 is of crucial importance to drive definitive conclusions about the therapeutic potential of blocking its effects in scleroderma patients. In particular, additional in vivo data are eagerly awaited to better understand whether interfering with IL-17 might be beneficial to arrest collagen deposition and tissue fibrosis or, rather, detrimental in SSc. In this regard, understanding what regulates the pro-fibrotic as well as the anti-fibrotic outcomes of IL-17 is pivotal to properly tune the in vivo biology of this cytokine. As this dual activity possibly depends also on the distribution of IL-17 receptors on target organs and on different intracellular pathways on target cells, useful information may be derived in the future by clinical studies assessing the efficacy of IL-17 inhibitors on organ-specific fibrotic disorders such as liver and lung fibrosis.
Footnotes
Author contributions: All authors contributed to the design of the work, acquisition, analysis, and interpretation of data. They revised the work critically for important intellectual content and approved the final version of the article. They agreed to be accountable for all aspects of the work by ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved. They contributed equally.
Declaration of conflicting interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) received no financial support for the research, authorship, and/or publication of this article.
Ethical approval and patient consent: This study did not require an approval by an institutional review board (IRB) because this is not an interventional or observational study on humans or animals and no diagnostic or therapeutic procedures were performed.
ORCID iD: Emanuel Della-Torre
https://orcid.org/0000-0002-9192-4270
References
- 1. Meier FM, Frommer KW, Dinser R, et al. Update on the profile of the EUSTAR cohort: an analysis of the EULAR Scleroderma Trials and Research group database. Ann Rheum Dis 2012; 71(8): 1355–1360. [DOI] [PubMed] [Google Scholar]
- 2. Varga J, Trojanowska M, Kuwana M. Pathogenesis of systemic sclerosis: recent insights of molecular and cellular mechanisms and therapeutic opportunities. J Scleroderma Relat Disord 2017; 2(3): 137–152. [Google Scholar]
- 3. Scher JU, Abramson SB. The microbiome and rheumatoid arthritis. Nat Rev Rheumatol 2011; 7: 569–578. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Baraut J, Michel L, Verrecchia F, et al. Relationship between cytokine profiles and clinical outcomes in patients with systemic sclerosis. Autoimmun Rev 2010; 10(2): 65–73. [DOI] [PubMed] [Google Scholar]
- 5. Sakkas LI, Platsoucas CD. Is systemic sclerosis an antigen-driven T cell disease? Arthritis Rheum 2004; 50(6): 1721–1733. [DOI] [PubMed] [Google Scholar]
- 6. Giovannetti A, Rosato E, Renzi C, et al. Analyses of T cell phenotype and function reveal an altered T cell homeostasis in systemic sclerosis. Correlations with disease severity and phenotypes. Clin Immunol 2010; 137(1): 122–133. [DOI] [PubMed] [Google Scholar]
- 7. Harrington LE, Hatton RD, Mangan PR, et al. Interleukin 17-producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nat Immunol 2005; 6(11): 1123–1132. [DOI] [PubMed] [Google Scholar]
- 8. Park H, Li Z, Yang XO, et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol 2005; 6(11): 1133–1141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Radstake TR, van Bon L, Broen J, et al. The pronounced Th17 profile in systemic sclerosis (SSc) together with intracellular expression of TGFbeta and IFNgamma distinguishes SSc phenotypes. PLoS ONE 2009; 4: e5903. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Rodríguez-Reyna TS, Furuzawa-Carballeda J, Cabiedes J, et al. Th17 peripheral cells are increased in diffuse cutaneous systemic sclerosis compared with limited illness: a cross-sectional study. Rheumatol Int 2012; 32(9): 2653–2660. [DOI] [PubMed] [Google Scholar]
- 11. Truchetet ME, Brembilla NC, Montanari E, et al. Increased frequency of circulating Th22 in addition to Th17 and Th2 lymphocytes in systemic sclerosis: association with interstitial lung disease. Arthritis Res Ther 2011; 13(5): R166. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Omenetti S, Bussi C, Metidji A, et al. The intestine harbors functionally distinct homeostatic tissue-resident and inflammatory Th17 cells. Immunity 2019; 51: 77.e6–89.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13. Miossec P, Kolls JK. Targeting IL-17 and TH17 cells in chronic inflammation. Nat Rev Drug Discov 2012; 11(10): 763–776. [DOI] [PubMed] [Google Scholar]
- 14. Tabarkiewicz J, Pogoda K, Karczmarczyk A, et al. The role of IL-17 and Th17 lymphocytes in autoimmune diseases. Arch Immunol Ther Exp 2015; 63(6): 435–449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Chen K, Kolls JK. Interluekin-17A (IL17A). Gene 2017; 614: 8–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Veldhoen M. Interleukin 17 is a chief orchestrator of immunity. Nat Immunol 2017; 18: 612–621. [DOI] [PubMed] [Google Scholar]
- 17. Monin L, Gaffen SL. Interleukin 17 family cytokines: signaling mechanisms, biological activities, and therapeutic implications. Cold Spring Harb Perspect Biol 2018; 10: a028522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Brevi A, Cogrossi LL, Grazia G, et al. Much more than IL-17A: cytokines of the IL-17 family between microbiota and cancer. Front Immunol 2020; 11: 565470. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Cua DJ, Tato CM. Innate IL-17-producing cells: the sentinels of the immune system. Nat Rev Immunol 2010; 10(7): 479–489. [DOI] [PubMed] [Google Scholar]
- 20. McGeachy MJ, Cua DJ, Gaffen SL. The IL-17 family of cytokines in health and disease. Immunity 2019; 50: 892–906. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Gaffen SL, Jain R, Garg AV, et al. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol 2014; 14(9): 585–600. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Mangan PR, Su LJ, Jenny V, et al. Dual inhibition of interleukin-23 and interleukin-17 offers superior efficacy in mouse models of autoimmunity. J Pharmacol Exp Ther 2015; 354(2): 152–165. [DOI] [PubMed] [Google Scholar]
- 23. Patel DD, Kuchroo VK. Th17 cell pathway in human immunity: lessons from genetics and therapeutic interventions. Immunity 2015; 43: 1040–1051. [DOI] [PubMed] [Google Scholar]
- 24. Chizzolini C, Dufour AM, Brembilla NC. Is there a role for IL-17 in the pathogenesis of systemic sclerosis? Immunol Lett 2018; 195: 61–67. [DOI] [PubMed] [Google Scholar]
- 25. Xing X, Yang J, Yang X, et al. IL-17A induces endothelial inflammation in systemic sclerosis via the ERK signaling pathway. PLoS ONE 2013; 8(12): e85032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Fossiez F, Djossou O, Chomarat P, et al. T cell interleukin-17 induces stromal cells to produce proinflammatory and hematopoietic cytokines. J Exp Med 1996; 183: 2593–2603. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Kurasawa K, Hirose K, Sano H, et al. Increased interleukin-17 production in patients with systemic sclerosis. Arthritis Rheum 2000; 43(11): 2455–2463. [DOI] [PubMed] [Google Scholar]
- 28. Gu L, Xu Q, Cao H. 1,25(OH)2D3 protects liver fibrosis through decreasing the generation of TH17 cells. Med Sci Monit 2017; 23: 2049–2058. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Wree A, McGeough MD, Inzaugarat ME, et al. NLRP3 inflammasome driven liver injury and fibrosis: roles of IL-17 and TNF in mice. Hepatology 2018; 67(2): 736–749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Zepeda-Morales AS, Del Toro-Arreola S, García-Benavides L, et al. Liver fibrosis in bile duct-ligated rats correlates with increased hepatic IL-17 and TGF-β2 expression. Ann Hepatol 2016; 15(3): 418–426. [DOI] [PubMed] [Google Scholar]
- 31. Meng F, Wang K, Aoyama T, et al. Interleukin-17 signaling in inflammatory, Kupffer cells, and hepatic stellate cells exacerbates liver fibrosis in mice. Gastroenterology 2012; 143(3): 765.e3–776.e3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Amara S, Lopez K, Banan B, et al. Synergistic effect of pro-inflammatory TNFα and IL-17 in periostin mediated collagen deposition: potential role in liver fibrosis. Mol Immunol 2015; 64(1): 26–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Tan Z, Qian X, Jiang R, et al. IL-17A plays a critical role in the pathogenesis of liver fibrosis through hepatic stellate cell activation. J Immunol 2013; 191: 1835–1844. [DOI] [PubMed] [Google Scholar]
- 34. Chakir J, Shannon J, Molet S, et al. Airway remodeling-associated mediators in moderate to severe asthma: effect of steroids on TGF-beta, IL-11, IL-17, and type I and type III collagen expression. J Allergy Clin Immunol 2003; 111(6): 1293–1298. [DOI] [PubMed] [Google Scholar]
- 35. Jiang G, Liu CT, Zhang WD. IL-17A and GDF15 are able to induce epithelial-mesenchymal transition of lung epithelial cells in response to cigarette smoke. Exp Ther Med 2018; 16(1): 12–20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Wilson MS, Madala SK, Ramalingam TR, et al. Bleomycin and IL-1β-mediated pulmonary fibrosis is IL-17A dependent. J Exp Med 2010; 207: 535–552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Zhou X, Loomis-King H, Gurczynski SJ, et al. Bone marrow transplantation alters lung antigen-presenting cells to promote TH17 response and the development of pneumonitis and fibrosis following gammaherpesvirus infection. Mucosal Immunol 2016; 9(3): 610–620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Feng W, Li W, Liu W, et al. IL-17 induces myocardial fibrosis and enhances RANKL/OPG and MMP/TIMP signaling in isoproterenol-induced heart failure. Exp Mol Pathol 2009; 87(3): 212–218. [DOI] [PubMed] [Google Scholar]
- 39. Liu W, Wang X, Feng W, et al. Lentivirus mediated IL-17R blockade improves diastolic cardiac function in spontaneously hypertensive rats. Exp Mol Pathol 2011; 91(1): 362–367. [DOI] [PubMed] [Google Scholar]
- 40. Valente AJ, Yoshida T, Gardner JD, et al. Interleukin-17A stimulates cardiac fibroblast proliferation and migration via negative regulation of the dual-specificity phosphatase MKP-1/DUSP-1. Cell Signal 2012; 24(2): 560–568. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Liu Y, Zhu H, Su Z, et al. IL-17 contributes to cardiac fibrosis following experimental autoimmune myocarditis by a PKCβ/Erk1/2/NF-κB-dependent signaling pathway. Int Immunol 2012; 24(10): 605–612. [DOI] [PubMed] [Google Scholar]
- 42. Zhou SF, Yuan J, Liao MY, et al. IL-17A promotes ventricular remodeling after myocardial infarction. J Mol Med 2014; 92(10): 1105–1116. [DOI] [PubMed] [Google Scholar]
- 43. Chang SL, Hsiao YW, Tsai YN, et al. Interleukin-17 enhances cardiac ventricular remodeling via activating MAPK pathway in ischemic heart failure. J Mol Cell Cardiol 2018; 122: 69–79. [DOI] [PubMed] [Google Scholar]
- 44. Faust SM, Lu G, Marini BL, et al. Role of T cell TGFbeta signaling and IL-17 in allograft acceptance and fibrosis associated with chronic rejection. J Immunol 2009; 183: 7297–7306. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Needleman BW, Wigley FM, Stair RW. Interleukin-1, interleukin-2, interleukin-4, interleukin-6, tumor necrosis factor alpha, and interferon-gamma levels in sera from patients with scleroderma. Arthritis Rheum 1992; 35(1): 67–72. [DOI] [PubMed] [Google Scholar]
- 46. Scala E, Pallotta S, Frezzolini A, et al. Cytokine and chemokine levels in systemic sclerosis: relationship with cutaneous and internal organ involvement. Clin Exp Immunol 2004; 138(3): 540–546. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Koch AE, Kronfeld-Harrington LB, Szekanecz Z, et al. In situ expression of cytokines and cellular adhesion molecules in the skin of patients with systemic sclerosis. Their role in early and late disease. Pathobiology 1993; 61(5–6): 239–246. [DOI] [PubMed] [Google Scholar]
- 48. Yang X, Yang J, Xing X, et al. Increased frequency of Th17 cells in systemic sclerosis is related to disease activity and collagen overproduction. Arthritis Res Ther 2014; 16: R4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Zhou Y, Hou W, Xu K, et al. The elevated expression of Th17-related cytokines and receptors is associated with skin lesion severity in early systemic sclerosis. Hum Immunol 2015; 76(1): 22–29. [DOI] [PubMed] [Google Scholar]
- 50. Valentini G, Della Rossa A, Bombardieri S, et al. European multicentre study to define disease activity criteria for systemic sclerosis. II. Identification of disease activity variables and development of preliminary activity indexes. Ann Rheum Dis 2001; 60: 592–598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Liu M, Yang J, Xing X, et al. Interleukin-17A promotes functional activation of systemic sclerosis patient-derived dermal vascular smooth muscle cells by extracellular-regulated protein kinases signalling pathway. Arthritis Res Ther 2014; 16: 4223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Truchetet ME, Brembilla NC, Montanari E, et al. Interleukin-17A+ cell counts are increased in systemic sclerosis skin and their number is inversely correlated with the extent of skin involvement. Arthritis Rheum 2013; 65(5): 1347–1356. [DOI] [PubMed] [Google Scholar]
- 53. Lonati PA, Brembilla NC, Montanari E, et al. High IL-17E and low IL-17C dermal expression identifies a fibrosis-specific motif common to morphea and systemic sclerosis. PLoS ONE 2014; 9(8): e105008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Korn T, Bettelli E, Oukka M, et al. IL-17 and Th17 cells. Annu Rev Immunol 2009; 27: 485–517. [DOI] [PubMed] [Google Scholar]
- 55. Ogura H, Murakami M, Okuyama Y, et al. Interleukin-17 promotes autoimmunity by triggering a positive-feedback loop via interleukin-6 induction. Immunity 2008; 29: 628–636. [DOI] [PubMed] [Google Scholar]
- 56. Koenders MI, Kolls JK, Oppers-Walgreen B, et al. Interleukin-17 receptor deficiency results in impaired synovial expression of interleukin-1 and matrix metalloproteinases 3, 9, and 13 and prevents cartilage destruction during chronic reactivated streptococcal cell wall-induced arthritis. Arthritis Rheum 2005; 52(10): 3239–3247. [DOI] [PubMed] [Google Scholar]
- 57. Simonian PL, Roark CL, Wehrmann F, et al. Th17-polarized immune response in a murine model of hypersensitivity pneumonitis and lung fibrosis. J Immunol 2009; 182: 657–665. [PMC free article] [PubMed] [Google Scholar]
- 58. Park MJ, Moon SJ, Lee EJ, et al. IL-1-IL-17 signaling axis contributes to fibrosis and inflammation in two different murine models of systemic sclerosis. Front Immunol 2018; 9: 1611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Lei L, Zhong XN, He ZY, et al. IL-21 induction of CD4+ T cell differentiation into Th17 cells contributes to bleomycin-induced fibrosis in mice. Cell Biol Int 2015; 39(4): 388–399. [DOI] [PubMed] [Google Scholar]
- 60. François A, Gombault A, Villeret B, et al. B cell activating factor is central to bleomycin- and IL-17-mediated experimental pulmonary fibrosis. J Autoimmun 2015; 56: 1–11. [DOI] [PubMed] [Google Scholar]
- 61. Mi S, Li Z, Yang HZ, et al. Blocking IL-17A promotes the resolution of pulmonary inflammation and fibrosis via TGF-beta1-dependent and -independent mechanisms. J Immunol 2011; 187: 3003–3014. [DOI] [PubMed] [Google Scholar]
- 62. Okamoto Y, Hasegawa M, Matsushita T, et al. Potential roles of interleukin-17A in the development of skin fibrosis in mice. Arthritis Rheum 2012; 64(11): 3726–3735. [DOI] [PubMed] [Google Scholar]
- 63. Hsu HC, Yang P, Wang J, et al. Interleukin 17-producing T helper cells and interleukin 17 orchestrate autoreactive germinal center development in autoimmune BXD2 mice. Nat Immunol 2008; 9(2): 166–175. [DOI] [PubMed] [Google Scholar]
- 64. Berger M, Steen VD. Role of anti-receptor autoantibodies in pathophysiology of scleroderma. Autoimmun Rev 2017; 16(10): 1029–1035. [DOI] [PubMed] [Google Scholar]
- 65. Nakashima T, Jinnin M, Yamane K, et al. Impaired IL-17 signaling pathway contributes to the increased collagen expression in scleroderma fibroblasts. J Immunol 2012; 188: 3573–3583. [DOI] [PubMed] [Google Scholar]
- 66. Murata M, Fujimoto M, Matsushita T, et al. Clinical association of serum interleukin-17 levels in systemic sclerosis: is systemic sclerosis a Th17 disease? J Dermatol Sci 2008; 50(3): 240–242. [DOI] [PubMed] [Google Scholar]
- 67. Lewis BP, Burge CB, Bartel DP. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005; 120: 15–20. [DOI] [PubMed] [Google Scholar]
- 68. Agarwal S, Misra R, Aggarwal A. Interleukin 17 levels are increased in juvenile idiopathic arthritis synovial fluid and induce synovial fibroblasts to produce proinflammatory cytokines and matrix metalloproteinases. J Rheumatol 2008; 35(3): 515–519. [PubMed] [Google Scholar]
- 69. Wei J, Bhattacharyya S, Tourtellotte WG, et al. Fibrosis in systemic sclerosis: emerging concepts and implications for targeted therapy. Autoimmun Rev 2011; 10(5): 267–275. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Brembilla NC, Montanari E, Truchetet ME, et al. Th17 cells favor inflammatory responses while inhibiting type I collagen deposition by dermal fibroblasts: differential effects in healthy and systemic sclerosis fibroblasts. Arthritis Res Ther 2013; 15: R151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Boukamp P, Breitkreutz D, Stark HJ, et al. Mesenchyme-mediated and endogenous regulation of growth and differentiation of human skin keratinocytes derived from different body sites. Differentiation 1990; 44(2): 150–161. [DOI] [PubMed] [Google Scholar]
- 72. Dufour AM, Borowczyk-Michalowska J, Alvarez M, et al. IL-17A dissociates inflammation from fibrogenesis in systemic sclerosis. J Invest Dermatol 2020; 140(1): 103.e8–112.e8. [DOI] [PubMed] [Google Scholar]
- 73. Gonçalves RSG, Pereira MC, Dantas AT, et al. IL-17 and related cytokines involved in systemic sclerosis: perspectives. Autoimmunity 2018; 51(1): 1–9. [DOI] [PubMed] [Google Scholar]
- 74. Zeng C, Kahlenberg JM, Gudjonsson JE. IL-17A softens the skin: antifibrotic properties of IL-17A in systemic sclerosis. J Invest Dermatol 2020; 140(1): 13–14. [DOI] [PubMed] [Google Scholar]
- 75. Leonardi CL. Antibodies in the treatment of psoriasis: IL-12/23 p40 and IL-17a. Semin Cutan Med Surg 2016; 35: S74–S77. [DOI] [PubMed] [Google Scholar]
- 76. Lebwohl M, Leonardi C, Griffiths CE, et al. Long-term safety experience of ustekinumab in patients with moderate-to-severe psoriasis (Part I of II): results from analyses of general safety parameters from pooled Phase 2 and 3 clinical trials. J Am Acad Dermatol 2012; 66(5): 731–741. [DOI] [PubMed] [Google Scholar]
- 77. Kavanaugh A, Ritchlin C, Rahman P, et al. Ustekinumab, an anti-IL-12/23 p40 monoclonal antibody, inhibits radiographic progression in patients with active psoriatic arthritis: results of an integrated analysis of radiographic data from the phase 3, multicentre, randomised, double-blind, placebo-controlled PSUMMIT-1 and PSUMMIT-2 trials. Ann Rheum Dis 2014; 73(6): 1000–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Menter A, Cather JC, Jarratt M, et al. Efficacy of secukinumab on moderate-to-severe plaque psoriasis affecting different body regions: a pooled analysis of four phase 3 studies. Dermatol Ther 2016; 6(4): 639–647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Alzabin S, Abraham SM, Taher TE, et al. Incomplete response of inflammatory arthritis to TNFα blockade is associated with the Th17 pathway. Ann Rheum Dis 2012; 71(10): 1741–1748. [DOI] [PubMed] [Google Scholar]
- 80. Queiroz-Junior CM, Bessoni RLC, Costa VV, et al. Preventive and therapeutic anti-TNF-α therapy with pentoxifylline decreases arthritis and the associated periodontal co-morbidity in mice. Life Sci 2013; 93: 423–428. [DOI] [PubMed] [Google Scholar]
- 81. Xueyi L, Lina C, Zhenbiao W, et al. Levels of circulating Th17 cells and regulatory T cells in ankylosing spondylitis patients with an inadequate response to anti-TNF-α therapy. J Clin Immunol 2013; 33(1): 151–161. [DOI] [PubMed] [Google Scholar]
- 82. McGovern JL, Nguyen DX, Notley CA, et al. Th17 cells are restrained by Treg cells via the inhibition of interleukin-6 in patients with rheumatoid arthritis responding to anti-tumor necrosis factor antibody therapy. Arthritis Rheum 2012; 64(10): 3129–3138. [DOI] [PubMed] [Google Scholar]
- 83. Yue C, You X, Zhao L, et al. The effects of adalimumab and methotrexate treatment on peripheral Th17 cells and IL-17/IL-6 secretion in rheumatoid arthritis patients. Rheumatol Int 2010; 30(12): 1553–1557. [DOI] [PubMed] [Google Scholar]
- 84. Van de Veerdonk FL, Lauwerys B, Marijnissen RJ, et al. The anti-CD20 antibody rituximab reduces the Th17 cell response. Arthritis Rheum 2011; 63(6): 1507–1516. [DOI] [PubMed] [Google Scholar]
- 85. Lai K, Zeng K, Zeng F, et al. Allogeneic adipose-derived stem cells suppress Th17 lymphocytes in patients with active lupus in vitro. Acta Biochim Biophys Sin 2011; 43(10): 805–812. [DOI] [PubMed] [Google Scholar]
- 86. Li Y, Jiang L, Zhang S, et al. Methotrexate attenuates the Th17/IL-17 levels in peripheral blood mononuclear cells from healthy individuals and RA patients. Rheumatol Int 2012; 32(8): 2415–2422. [DOI] [PubMed] [Google Scholar]
- 87. Truchetet ME, Allanore Y, Montanari E, et al. Prostaglandin I(2) analogues enhance already exuberant Th17 cell responses in systemic sclerosis. Ann Rheum Dis 2012; 71(12): 2044–2050. [DOI] [PubMed] [Google Scholar]
- 88. Roofeh D, Lin CJF, Goldin J, et al. Tocilizumab prevents progression of early systemic sclerosis-associated interstitial lung disease. Arthritis Rheumatol 2021; 73(7): 1301–1310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Khanna D, Lin CJF, Furst DE, et al. Tocilizumab in systemic sclerosis: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med 2020; 8(10): 963–974. [DOI] [PubMed] [Google Scholar]
- 90. Zacay G, Levy Y. Outcomes of patients with systemic sclerosis treated with tocilizumab: case series and review of the literature. Best Pract Res Clin Rheumatol 2018; 32(4): 563–571. [DOI] [PubMed] [Google Scholar]
- 91. Huang Y, Fan Y, Liu Y, et al. Efficacy and safety of secukinumab in active rheumatoid arthritis with an inadequate response to tumor necrosis factor inhibitors: a meta-analysis of phase III randomized controlled trials. Clin Rheumatol 2019; 38(10): 2765–2776. [DOI] [PubMed] [Google Scholar]
- 92. Mease PJ, Helliwell PS, Hjuler KF, et al. Brodalumab in psoriatic arthritis: results from the randomised phase III AMVISION-1 and AMVISION-2 trials. Ann Rheum Dis 2021; 80(2): 185–193. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Madureira P, Pimenta SS, Bernardo A, et al. Off-label use of tocilizumab in psoriatic arthritis: case series and review of the literature. Acta Reumatol Port 2016; 41(3): 251–255. [PubMed] [Google Scholar]
- 94. Lebwohl M, Strober B, Menter A, et al. Phase 3 studies comparing brodalumab with ustekinumab in psoriasis. N Engl J Med 2015; 373(14): 1318–1328. [DOI] [PubMed] [Google Scholar]
- 95. Papp KA, Reich K, Paul C, et al. A prospective phase III, randomized, double-blind, placebo-controlled study of brodalumab in patients with moderate-to-severe plaque psoriasis. Br J Dermatol 2016; 175(2): 273–286. [DOI] [PubMed] [Google Scholar]
- 96. Marighela TF, Arismendi MI, Marvulle V, et al. Effect of probiotics on gastrointestinal symptoms and immune parameters in systemic sclerosis: a randomized placebo-controlled trial. Rheumatology 2019; 58: 1985–1990. [DOI] [PubMed] [Google Scholar]