Skip to main content

REVIEW article

Front. Immunol., 22 February 2021
Sec. Cytokines and Soluble Mediators in Immunity
This article is part of the Research Topic Role of the IL-23/IL-17 Pathway in Chronic Immune-Mediated Inflammatory Diseases: Mechanisms and Targeted Therapies View all 21 articles

Role of the IL-23/IL-17 Pathway in Rheumatic Diseases: An Overview

\nClaudia SchinoccaClaudia Schinocca1Chiara RizzoChiara Rizzo1Serena FasanoSerena Fasano2Giulia GrassoGiulia Grasso1Lidia La BarberaLidia La Barbera1Francesco CicciaFrancesco Ciccia2Giuliana Guggino
Giuliana Guggino1*
  • 1Rheumatology Section, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University Hospital “P. Giaccone”, Palermo, Italy
  • 2Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, Naples, Italy

Interleukin-23 (IL-23) is a pro-inflammatory cytokine composed of two subunits, IL-23A (p19) and IL-12/23B (p40), the latter shared with Interleukin-12 (IL-12). IL-23 is mainly produced by macrophages and dendritic cells, in response to exogenous or endogenous signals, and drives the differentiation and activation of T helper 17 (Th17) cells with subsequent production of IL-17A, IL-17F, IL-6, IL-22, and tumor necrosis factor α (TNF-α). Although IL-23 plays a pivotal role in the protective immune response to bacterial and fungal infections, its dysregulation has been shown to exacerbate chronic immune-mediated inflammation. Well-established experimental data support the concept that IL-23/IL-17 axis activation contributes to the development of several inflammatory diseases, such as PsA, Psoriasis, Psoriatic Arthritis; AS, Ankylosing Spondylitis; IBD, Inflammatory Bowel Disease; RA, Rheumatoid Arthritis; SS, Sjogren Syndrome; MS, Multiple Sclerosis. As a result, emerging clinical studies have focused on the blockade of this pathogenic axis as a promising therapeutic target in several autoimmune disorders; nevertheless, a greater understanding of its contribution still requires further investigation. This review aims to elucidate the most recent studies and literature data on the pathogenetic role of IL-23 and Th17 cells in inflammatory rheumatic diseases.

Review

Interleukin-23

Interleukin-23 (IL-23) is a member of the IL-12 cytokine family composed of the IL-23p19 subunit and the IL-12/23p40 subunit, the latter shared with IL-12, encoded by genes located on chromosomes 12q13.2 and 11q1.3, respectively (13).

IL- 23 is mainly secreted by activated macrophages and dendritic cells (DCs) located in peripheral tissues, such as skin, intestinal mucosa, joints and lungs (46).

Despite the protective role played by the IL-23/IL-17 axis against bacterial and fungal infections, extensive knowledge supports the contribution of its dysregulation in triggering chronic inflammation and autoimmunity, providing a solid substrate for the development of several autoimmune diseases like PsA, Psoriasis, Psoriatic Arthritis; AS, Ankylosing Spondylitis; IBD, Inflammatory Bowel Disease; RA, Rheumatoid Arthritis; SS, Sjogren Syndrome; MS, Multiple Sclerosis (3, 710) (Table 1).

TABLE 1
www.frontiersin.org

Table 1. Therapeutic agents in rheumatic diseases.

The main role of IL-23 is to induce the differentiation of αβ T CD4+ naïve cells (Th0 cells) in T helper type 17 (Th17 cells) (11, 12), which are considered pivotal players in autoimmunity (1, 9).

Although IL-12 and IL-23 are both members of the IL-12 family and have a similar structure, the role of these two cytokines in Th0 differentiation is totally different (13, 14); indeed, unlike IL-23, IL-12 induces differentiation of Th0 cells into T helper type 1 (Th1 cells) rather than into Th17 (Figure 1) (1519). Both cytokines are produced by DCs and the balance of IL-12 and IL-23 production is controlled by prostaglandin E2 (PGE2), which promotes inflammatory responses (20, 21).

FIGURE 1
www.frontiersin.org

Figure 1. Schematic image of the cascade of cytokines and transcription factors involved in the differentiation of Th1 and Th17 cells. IFN-γ, interferon-γ; IL, interleukin; RORγt, retinoid-related orphan receptor γt, STAT, signal transducer and activator of transcription; GATA, GATA transcription factor; TGF-β, transforming growth factor β, Th, T helper.

IL-12 and IL-23 act as a bridge between the innate and adaptive arms of the immune response (22). IL-12, produced by antigen presenting cells (APCs), is essential for the optimal proliferation and production of cytokines by Th1 cells in response to antigens. Overall, IL-12-induced IFN-γ is an effective activator of the antimicrobial functions of phagocytes and plays a critical role in resistance to many pathogenic bacteria, fungi and intracellular parasites (23).

Regarding the IL-23/IL-17 axis, γδ T cells and innate lymphoid cells (ILCs) constitutively express the IL-23 receptor (IL-23R), suggesting their prompt first-line response to IL-23, followed by cytokine secretion and subsequent activation of the adaptive immune response. Moreover, since Th0 cells do not express IL-23R, they require prior stimulation with transforming growth factor β (TGF-β), IL-6 and IL-21 to become responsive to IL-23 (2429).

In response to IL-23-mediated activation, αβ T cells, γδ T cells and ILCs produce IL-17, IL-22, TNF-α, and interferon-gamma (IFN-γ); in addition, IL-23-activated γδ T cells make αβ T cells refractory to the suppressive activity of regulatory T cells (Treg) and they also prevent the conversion of conventional T cells into FOXP3+ Treg cells in vivo (30).

IL-23 Receptor

The IL-23 receptor (IL-23R) is a heterodimeric receptor composed of 2 subunits: IL-12Rβ1, in common with the IL-12 receptor (IL-12R) and IL-23Rα, specific to IL-23 signaling (31).

Therefore, T cells lacking IL-12Rβ1 cannot respond to IL-12 nor IL-23. Conversely, IL-23Rα-deficient T cells cannot respond to IL-23, while maintaining IL-12 signaling capability (32). IL-23Rα and IL-12Rβ1 chains are expressed on T cells, natural killer (NK) T cells, monocytes/macrophage and DCs (33).

The intracellular pathways require different signaling proteins: JAK2, Janus kinase 2; TYK2, tyrosine kinase 2; STAT3, STAT4, signal transducer and activator of transcription 3 and 4 (34, 35).

Specifically, IL-12Rβ1 binds to TYK2 inducing STAT4 phosphorylation which is essential for increasing IFN-γ production and subsequent Th1 cells differentiation. Instead, IL-23Rα interacts with JAK2, inducing STAT3 phosphorylation and leading to the upregulation of retinoid-related orphan receptor gamma tau (RORγt), crucial for the development of Th17 cells (19, 31, 36, 37).

Once activated, STATs homodimers translocate into the nucleus, where they bind to DNA in the promoter region of target genes, acting as downstream effectors in the IL-23/IL-12 signaling pathway (38, 39).

Finally, the evaluation of IL-23 functions in vivo in different mouse models supports the hypothesis that IL-23 may act in both lymph nodes and peripheral tissues to drive terminal differentiation of effector Th17 cells in vivo, promoting Th-17-mediated inflammation. Confirming these observations, in the absence of Il-23, Th17 cells experience “arrested development” leading to impaired function (40).

Interleukin-17A

Interleukin-17A (IL-17A) is the first described member of the IL-17 cytokine family, which includes six members, IL-17A to IL-17F (41, 42).

Many IL-17A-producing cells have been reported, including T CD8+ cells (43), γδ T cells (44), and NK T cells (45); however, according to current knowledge, T CD4+ cells (Th17) are the major source of IL-17.

Although IL-23 has mainly been identified as the initiating factor for IL-17 expression from T cells (9, 46), Th0 cells do not constitutively express IL-23R, but they are still sensitive to IL-23 (47); in this context, it is reasonable to assume that IL-23R expression on these cells can be induced in the presence of other pro-inflammatory cytokines (48, 49).

Several findings clearly demonstrated that TGF-β and IL-6 are sufficient for Th17 differentiation in vitro and in vivo, in the absence of IL-23 (5053).

Therefore, TGF-β, IL-6, and IL-21 seem to activate T lymphocytes and promote the initial differentiation of Th0 into Th17 cells, conferring responsiveness to IL-23 (50, 51, 5460), which is a crucial step for Th17 cells stabilization and expansion (61).

Conversely, increased TGF-β levels coupled with the absence of inflammatory cytokines inhibit Th17 differentiation (62), as well as the common inhibitors of Th17 commitment (IFN-γ, IL-4, IL-25, IL-27) (5456, 6367).

Finally, IL-17-producing cells have been shown to express a wide range of heterogeneous cytokines such as IL-17A, IL-17F, IL-26 (62), and other proinflammatory mediators including IL-22, IL-21,IL-6, TNF-α, granulocyte colony-stimulating factors (GM-CSF), and chemokines (e.g., CCL20, CXCL8, CXCL1, CXCL10) (9).

IL-17 Receptor

IL-17 receptor (IL-17R) is expressed on many cell types, including epithelial cells, B and T cells, fibroblasts, monocytic cells, and bone marrow stroma (68).

IL-17 signaling activates nuclear factor κB (NFκB) activator adaptor protein (ACT1), which in turn acts on mitogen-activated protein kinases (MAPKs), including p38MAK (69), c-jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK), Janus kinase (JAK), signal transducer and activator of transcription (STAT), phosphoinositol 3 kinase (PI3K) and induces several pro-inflammatory cytokines (IL-1β, IL-6, TNF-α, CCL2), antimicrobial peptides (β-defensin), and matrix metalloproteinases (6971).

In health conditions, IL-17 is one of the main contributors to the host defense against microbial infections (68, 7274). Of note, the IL-17 pathway regulates antifungal immunity, in human and mice, inducing upregulation of proinflammatory cytokines, antimicrobial peptides and neutrophil-recruiting chemokines, which lead to limit fungal overgrowth (75).

Historically, CD4+ T cells have been involved in protecting against Candida albicans infection were found in Human Immunodeficiency Virus (HIV) positive patients (76, 77).

Subsequently, Th17 subset was identified as CD4+ T cells with reactivity to Candida albicans (37, 78).

Moreover, ex vivo studies on human T cells demonstrated that Candida albicans triggers Th17 cells which produce IL-17 and IFN-γ, but not IL-10. On the contrary, Staphylococcus aureus–activated Th17 cells produce IL-10, which can limit the immune system responses. These different responses could derive from the presence, in the priming phase, of different cytokine environments induced by each microbe (79).

The specific role of IL-17 in protection against the fungus Candida albicans was confirmed by evidence that IL-17RA deficient mice or mice and humans with defects along the IL-17 signaling pathway, were susceptible to systemic Candida albicans infection (8083).

Major roles of IL-17 include the promotion and initiation of chemotaxis and the recruitment and activation of neutrophils in inflamed tissues (71, 84, 85). Among its pleiotropic effects, the enhancement of angiogenesis (86) and the tissue remodeling through the production of angiogenic factors and matrix metalloproteases are worthy of note (87). IL-17 works in synergy with TNF-α causing release of the IL-6, TNF-α, and IL-1β, in order to amplify the multifaceted and complex inflammatory process (88).

Consistently, increased serum and tissue levels of IL-17 have been widely reported in inflammatory condition such as IBD, MS, and arthritis, compared to a non-pathological setting where IL-17A levels are extremely low or undetectable in human sera (Figure 2) (3, 10, 8992).

FIGURE 2
www.frontiersin.org

Figure 2. Pathogenesis of IL-17-correlated disease and different targets of therapy. IFN-γ, interferon-γ; IL, interleukin; Th, T helper; TNF-α, Tumor necrosis factor.

Ankylosing Spondylitis (AS)

Ankylosing Spondylitis (AS) is the prototypical subset of SpA characterized by a predominant axial involvement (93). As a result, sacroiliitis is the clinical hallmark of disease and its identification through the most sophisticated imaging techniques, such as high-field magnetic resonance imaging (MRI), is extremely important in order to achieve an early diagnosis that can prompt a rapid treatment administration. The new classification criteria published by the Assessment in Spondylo-Arthritis international Society (ASAS) in 2009 have included MRI as the gold standard technique to identify active sacroiliitis, consisting in bone marrow oedema and osteitis, even in patients that have not developed radiographical signs of disease (94). According to these criteria, the presence of alterations (both in standard radiography or MRI) coupled with clinical, genetic and laboratory data, allow the classification of AS patients into radiological axial SpA (r-axSpA) or non-radiological axial SpA (nr-axSpA) (95).

New bone formation, determined by chronic inflammation involving the spine, leads to vertebral ankylosis, severe chronic pain and disability as major consequences of disease progression.

The beginning of the inflammatory process in AS relies on a complex, multifactorial interplay between genetic, epigenetic and environmental factors associated with a dysregulated immune response. The current understanding of AS pathogenesis suggests that the IL-23/IL-17 axis acts as the major driver in disease development, even if type 17 response could not entirely elucidate the mechanisms behind this rheumatic disease (96).

Genetics and epigenetics play a pivotal role in the pathogenesis of AS; siblings of AS patients have a higher risk of developing the disease and a high degree of concordance in twins is observed, 50–63% in monozygotic and 13–20% in dizygotic twins, respectively (97). The strongest genetic association is with the allele human leukocyte antigen B27 (HLA-B27) of the Major Histocompatibility Complex-I (MHC-I) gene, located on chromosome 6 (98).

Several theories have emerged to explain the possible pathomechanism related to HLA-B27, AS starting, and subsequent dysregulated activation of the IL-23/IL-17 axis via Th17 cells (99). Modifications in the shape of HLA-B27 affect the protein binding domain impairing both the antigen presentation process and the correct folding of the HLA-B27 molecule (100). The altered binding domain may lead to the presentation of self-peptides to cytotoxic CD8+ T cells that in turn give rise to a pathological autoimmune response (101). On the other hand, the unfolded protein response (UPR) theory postulates that unconventional HLA-B27 variants homodimerize instead of heterodimerize; the misfolded proteins accumulate in the intracellular compartment triggering endoplasmic reticulum stress and increasing IL-23 production (102). In fact, heterodimers expressed on APC surface directly interact with cell receptors on a wide range of immune cells such as NK, monocytes, B cells and promote a significant impact on Th17 stimulation (103). In particular, the aberrant expression of the allele of the HLA B 27 in spondylitis, could act by binding cells that contain a natural killer receptor for HLA B 27 homodimer, named killer cell immunoglobulin-like receptor 3DL2 (KIR3DL2), determining IL-17 production (104).

HLA-B27 alone accounts for almost the 25% of AS heritability and genetic-wide association studies (GWAS) have identified multiple genetic loci linked to disease pathogenesis (105, 106). In particular, single nucleotide polymorphisms (SNPs) in genes coding for aminopeptidases expressed in the endoplasmic reticulum (ER), such as ERAP1 and ERAP2, were identified in the past decade. These proteins trim peptides in the ER so that these molecules get to the right length, usually between 8 and 10 amino acids, to be presented by MHC-I molecules (107). Mutations in ERAP enzymes are supposed to lead to the formation of the so called “arthritogenic peptide” which, through mechanisms related to molecular mimicry, triggers immune cells to react against self-antigens located at joint and enthesal sites (108). ERAP1 and HLA-B27 effects are linked in an epistatic way, meaning that ERAP1 mutation effects are only observed in HLA-B27 positive patients (109).

SNPs directly affecting the IL-23/IL-17 axis were described in AS and further stress the importance of this pathway. The most relevant are located in the genes coding for IL-23R and STAT3 and TYK2, which are downstream targets of IL-23 signaling (110, 111).

However, it is not entirely clear how this polymorphism is responsible for pathogenesis, but this probably alters the amino acid sequence of the intracellular portion of IL-23R, and it could have a secondary regulatory function; indeed, protective alleles have also been identified in IL-23R, and some studies have detected polymorphisms in the IL-23R as a strong protective genetic factor (112).

The fundamental role played by the IL-23/IL-17 axis comes from several lines of evidence depicting a clear increase in IL-23 and IL-17 levels in the sera of AS patients (113). This observation is coupled with the increased number of Th17 cells in peripheral blood from AS patients (114). In this regard, the most intriguing and recent theories suggest that the interface between environment and immune system in AS can be represented by the gut epithelial barrier, where IL-17A exerts its functions in maintaining mucosal immunity and barrier functions (115). The intimate relationship between the articular disease and the gut is underlined by the detection of subclinical inflammation in up to 70% of AS patients on endoscopic examination, as well as on histological samples (116) and in about 10% of these occurs a clinical IBD suggesting a pre-clinical stage of IBD (115). Even bone marrow oedema in sacroiliac joints was correlated to gut inflammation, as emerged from the analysis of the Ghent cohort (117).

At gut level the huge number of adherent and invading bacteria, known as human microbiota, may be perturbed leading to both quantitative and qualitative alterations that affect the integrity of the gut-epithelial and vascular barriers in accordance with the “joint-gut axis theory” (118). The “leaky gut” allows the translocation of bacteria-derived peptides and primed immune cells to the interstitium and then to the bloodstream, eliciting a systemic abnormal inflammatory response. The derangement of the gut interface has been related to the alteration of the tight junction system and an increase in zonulin level was retrieved both in gut epithelium and peripheral blood (119). In AS patients dysbiosis was evidenced in comparison to healthy individuals and increased level of IL-23 are found in patients' gut and in particular in the terminal ileum (120). Several cell populations involved in epithelial immunity and joint/enthesal inflammation, such as Th17, ILC, γδ T cells, and mucosa-associate invariant T (MAIT) cells, as well as cells involved in mucosal homeostasis, such as Paneth cells, produce IL-23 at intestinal level. ILC3 are expanded in gut and differentiate upon IL-17 and TNF-α stimulation becoming an important source of IL-23 and IL-17 (121). Among the intraepithelial lymphocyte (IEL) compartment γδ T cells are the most represented population, accounting for approximately the 50% of IEL; on the other hand, they represent only the 3–5% of circulating T cells. Once activated with IL-23 these cells produce IL-17 (44). Their number was found increased in gut and γδ T cells obtained from AS patients show hyper-responsiveness to IL-23, due to IL-23R hyper-expression, with consequent discharge of higher amount of IL-17 when stimulated (122).

According to the gut-joint axis theory the intestinal activation of different immune cell subsets, among the ones above described, followed by their recirculation in blood may lead to their final localization in joint and enthesis where the inflammatory process is carried out. Intestine seems to be the major site of IL-23 production and is also the site where it acts most. Even gut- derived IL-17 producing ILC3 were found expanded at bone marrow, joint and peripheral blood level. ILC3 are also characterized by a significant expression of an integrin that regulates intestinal T cells homing, called α4β7. Moreover, the receptor of this integrin (named MADCAM1) was found upregulated in the gut and in bone marrow of AS patients, suggesting a chemoattraction process of ILC3 at inflammation site (121). Cuthbert et al. described resident γδ T cells at spinal enthesis where they contribute to IL-17 production in both IL-23 dependent and independent ways. In fact, a subpopulation of these cells lacking IL-23R was proven to produce IL-17 (123). In addition, IL-17 producing MAIT cells were found elevated in AS patients both in blood and synovial fluid (124, 125). To add more pieces to the already complex puzzle of IL-17 producing cells, a new population of CD8+ T cells able to produce IL-17, named Tc17 cells, was described and was found increased in both peripheral blood and synovial fluid of AS patients (126). Other possible sources of IL-17 that deserve deeper investigation are tissue-resident memory T cells (TRM), mast cells and CD3CD56+NK cells (127, 128). In axial tissues IL-23 producing cells are macrophages and DC while IL-17 sources are myeloid cells as neutrophils (129, 130).

Animal models have supported the actual knowledge on the role of the IL-23/IL-17 axis, even if human disease appears far more complex and no animal model can comprehensively elucidate it. Transgenic HLA-B27 rat model was used to demonstrate the importance of the HLA-B27, gut microbiota and Th17 cells in the pathogenesis of SpA (131). These rats, grown in germ free condition do not develop SpA (132). Several mice models even exist and have contributed to the unraveling of the pivotal role played by IL-23 and IL-17 in activating T cells and driving disease development via type 17 immunity (133).

Taken together, the above resumed evidence underlines the importance of this axis in inducing and sustaining the multifaceted inflammatory process depicted in SpA, making it a central therapeutic target.

Advances in understanding the pathogenesis of SpA have prompted the development of biologic drugs designed to inhibit the IL-23/IL-17 axis. In fact, for more than 15 years, TNF-α inhibitors were the only biologic treatment available and, despite an initial great success in SpA management, it came out clearly that almost 40% of patients failed to reach a significative response. The new therapeutic agents interfering with the IL-23/IL-17 axis can be divided into monoclonal antibodies directly targeting IL-17, IL-23 or their receptors and small molecules inhibiting the intracellular pathways triggered by these cytokines (134).

Among monoclonal antibodies targeting IL-17, Secukinumab and Ixekizumab, respectively a fully human monoclonal IgG1/k antibody and a humanized IgG4 monoclonal antibody, are the only two molecules already marketed for SpA.

The MEASURE trials demonstrated the superiority of Secukinumab against placebo in providing sustained efficacy in relieving signs and symptoms of AS as well as in granting a good retention rate, as demonstrated in the 5-years extension study (135, 136). In the 2-years follow up in the MEASURE 1 trial no radiographic progression was evidenced in the 80% of patients included (137).

Ixekizumab was licensed for r-axSpA treatment in 2019 and for nr-axSpA in 2020, the COAST-V trial demonstrated a superior response rate in ASAS40 score at week 16 over placebo (138).

Several trials aimed to assess the therapeutic value of other IL-17 inhibitors, such as Netakimab and Bimekizumab, are currently ongoing (139, 140). The randomized controlled trial on Brodalumab, a humanized IgG2 monoclonal antibodies that binds IL-17R was discontinued because of the occurrence of high suicide ideation in the active group (141).

Up to date, no IL-23 targeting drug has been proven effective for AS (142, 143).

New treatments for AS are small molecules inhibitors that target intracellular proinflammatory pathways, as those triggered by cytokine stimulation. Among targeted synthetic DMARDs (tsDMARDs), JAK-inhibitors stand out as the most promising treatment (144). In particular, Tofacitinib, a pan-JAK inhibitor, Filgotinib and Upadacitinib, both JAK1 inhibitors, were shown to be superior to placebo in axSpA (145147).

The inhibition of PDE4 through the small molecule Apremilast failed to achieve the primary end-point in the specifically designed phase 2 trial including 490 AS patients (148).

Future perspectives to implement the therapeutic options for AS patients include broader anti-inflammatory approaches with multi-cytokine blockade to overcome the inhibition of a single pathway, for example targeting simultaneously TNF-α and IL-17, as in an ongoing study in PsA (149).

Psoriatic Arthritis

Psoriatic arthritis (PsA) is a chronic, immune-mediated, inflammatory disease dominated by a heterogeneous phenotype that mainly affects peripheral and axial joints, entheses, skin, and nails, leading to juxta-articular new bone formation, bone erosions and abnormal keratinocyte proliferation (31, 150).

Interactions between genes and environmental triggers, including infections, trauma, stress, obesity and smoking are recognized as crucial for the onset of the autoimmune process in PsA. Furthermore, the disruption of the gut microbiota composition in PsA patients is supported by consolidate evidence (151, 152).

The main effector cells of the inflammatory cascade, both in joints and in plaques of patients with PsA, are DCs, macrophages, NK cells (153, 154), mast cells, neutrophils (155, 156), γδ T cells (157), T CD4+ and T CD8 + cells.

All the ones above described have a predominant IL-17 secretory phenotype (41, 158), defined by the production of cytokines such us IL-17, IL-22, and TNFa; however, T CD4+ cells, as the major source of IL-17, are considered the cornerstone in the pathogenesis of psoriasis (157, 159).

Specifically, DC-derived cytokines, IL-23 and IL-12, drive the differentiation of distinct Th17 and Th1 cells, which are known to be implicated in the pathogenesis of PsA. Activated T cells move from the circulation to the target organs, and chronic inflammation occurs in the skin and joints.

Accordingly, increased numbers of Th17 cells were detected in the blood and affected skin of patients with psoriasis and in the blood and synovial fluid of patients with PsA. Furthermore, the assessment of the expression of IL-23, IL-17, and their related receptors in psoriatic skin lesions and inflamed synovium supports the concept of IL-23/IL-17 axis as a driving force of immune inflammation in psoriasis (160164) PsA synovitis is characterized by significant infiltration of mononuclear cells, T and B cells, vascular proliferation and hyperplasia of synovial lining cells, similar to the pathological changes observed in RA (165). Additionally, ectopic lymphoid tissues were frequently found in PsA synovial membrane with microanatomical features for germinal center formation, capable of antibody production (166). The role of B cells in PsA is still elusive; a recent study reported that autoantibodies against a peptide sharing sequence homology with skin and entheseal autoantigens were detected in 85% of patients with PsA (167).

Several findings have clearly demonstrated the influence of IL-17 on bone metabolism. Despite the direct effects of IL-17 on osteoclasts, induction of osteoclastogenesis is mediated by the production of matrix metalloproteinases by macrophages and the activator of the NF-κB ligand receptor (RANKL) presented by osteoblasts (168170).

Skin lesions from patients with psoriasis exhibit epidermal hyperplasia and infiltration with neutrophils, T CD4 + and T CD8 + cells, B cells, dendritic cells and mast cells, type 3 innate lymphoid cells (ILC3) and γδ T cells (171174).

In contrast, IL-22, totally absent in synovial tissue, is highly expressed in entheses, where it promotes entheseal and periosteal bone formation through STAT3 activation, explaining the formation of enthesophyte and juxta-articular bone, hallmarks of PsA (175, 176). In skin lesions IL-22 drives keratinocyte hyperproliferation via STAT3 signaling (177) and prompts epithelial cells to release chemokines, such as IL-8 (178, 179), a key factor in neutrophil recruitment in psoriatic lesions and stimulates keratinocytes to secrete antimicrobial peptides (180, 181), preventing skin lesions from becoming infected (182).

In inflammation, Th17 cells also produce interleukin 9 (IL-9), which in turn induces the differentiation of Th17 cells and potentiates the suppressive effect of regulatory T lymphocytes via activation of STAT3 and STAT5. Our group demonstrated that IL-9 overexpression and T helper type 9 (Th9) polarization occur in the synovial tissue and peripheral blood of PsA patients. Furthermore, clinical improvement after treatment with TNFi and ustekinumab was associated with a significant reduction in circulating Th9 cells (183, 184).

Up to date, a wide range of therapeutic approaches have been proposed for PsA, depending on disease severity, including disease-modifying anti-rheumatic drugs (DMARDs) such as methotrexate; anti-tumor necrosis factor α (anti-TNF-α) agents, phosphodiesterase 4 (PDE4) inhibitors, IL-17 and IL-12/IL-23 inhibitors (185, 186).

The central contribution of IL-23/IL-17 axis in both PsA and psoriasis is confirmed by the efficacy of biologics neutralizing IL-17 or IL-23/IL-12 (187) as well as the effectiveness of TNF-α inhibition dependent on down-regulation of IL-17 pathway genes (188190).

To date, several monoclonal antibodies have been approved for the treatment of PsA and can be introduce following the failure of non-steroidal anti-inflammatory drugs (NSAIDs) and/or conventional DMARDs, or an anti-TNF-α agent.

Ustekinumab is a human IgG1 monoclonal antibody that binds to the p40 subunit, shared with IL-12 and IL-23, and blocks downstream events of both the IL-12 and IL-23 signaling cascade through inhibition of IL-12Rβ1 binding (191). Ustekinumab has produced consistent and sustained clinical efficacy in two phase three clinical trials in PsA, PSUMMIT-1 and PSUMMIT-2, with data out to 52 weeks, and no new safety signals. PSUMMIT-1 included patients with active PsA despite conventional therapy who were all naïve to anti-TNF-α agents, whereas PSUMMIT-2 also included anti-TNF-α experienced patients (192).

Secukinumab, is a human IgG1κ monoclonal antibody that binds to IL-17A neutralizing its interaction with IL-17 receptors. It is currently approved in several countries for the treatment of PsA (193) and, in two phase 3 trials FUTURE1 and FUTURE 2, secukinumab has provided sustained improvements in disease signs and symptoms, assessing reduced radiographic progression in patients with active PsA through 2 years of therapy (194).

Ixekizumab is a humanized IgG4 monoclonal antibody targeting IL-17A, approved for the treatment of moderate-to-severe plaque psoriasis, active PsA, and active AS. Two phase three trials (SPIRIT-P1 and SPIRIT-P2) demonstrated that treatment with ixekizumab improved joint and skin disease compared to placebo (195, 196).

In addition, the SPIRIT-H2H study confirmed the superiority of ixekizumab over adalimumab in patients with Psa and inadequate response to csDMaRDs (197).

Over the past decade, the development of Janus kinase inhibitors (JAK inhibitors) has emerged as a new therapeutic option in autoimmune diseases, including PsA. The rationale of their use is suggested by the observation that the blockade of JAK receptor downregulates the production of the cytokines (TNF-α, IL-17, IL-6, IL-23) involved in the pathogenesis of PsA.

Tofacitinib is an orally administered inhibitor of predominantly JAK1 and JAK3, with functional selectivity to JAK2, used for the treatment of RA. Interestingly, its efficacy in managing treatment-resistant disease and ameliorating enthesitis, dactylitis, and radiographic progression has been reported. Consistently with previous observations, Tofacitinib could provide an alternative approach for PsA patients with inadequate response to DMARDs (198, 199).

Finally, Guselkumab, a monoclonal antibody targeting IL-23 via IL-23 p19 subunit, was recently approved for the treatment of PsA in adults with an inadequate response or intolerance to DMARDs therapy. Results for the use of Guselkumab derive from phase three clinical trials, DISCOVER-1 and DISCOVER-2, which demonstrated significantly better clinical and radiographic outcomes among PsA patients treated with the IL-23 inhibitor compared with placebo group.

Participants treated with Guselkumab achieved 20% improvement in American College of Rheumatology (ACR) response criteria at week 24 at rates of 52% in DISCOVER-1 and 64% in DISCOVER-2, whereas placebo-treated patients had rates of 22 and 33%, respectively (200, 201).

Rheumatoid Arthritis (RA)

Rheumatoid arthritis (RA) is a systemic autoimmune disease that primarily affects synovial joints, accompanied by systemic inflammation and production of autoantibodies (202).

The synovitis in RA is characterized by an inflammatory infiltrate, consisting of leukocytes such as T and B cells, macrophages, granulocytes and dendritic cells, together with a synovial milieu dominated by proinflammatory cytokines and chemokines (203).

For a long time, RA was considered a Th1 dependent disease, until a significant amount of research in RA patients and experimental mouse models suggested that Th17 cells may play a central role in the pathogenesis of RA.

IL-17 is involved in both early and established RA disease, promoting activation of fibroblast-like synoviocytes (FLS), osteoclastogenesis, recruitment and activation of neutrophils, macrophages and B cells (204).

Synergism between IL-17 and TNF-α has been shown to activate the production of pro-inflammatory mediators, such as IL-1β, IL-6, IL-8, PGE2, and matrix metalloproteinases (MMPs), promoting progression of early inflammation toward a chronic arthritis (205, 206).

The differentiation of osteoclasts is induced significantly in the presence of IL-17 either directly (207), or indirectly, through upregulation of RANKL.

By the late 1990s studies had already shown that IL-17 expression had increased in the joint of RA patients compared to healthy individuals or osteoarthritis (OA) patients (89).

More recently, an increased proportion of chemokine receptor CCR6+ Th17 cells has been described in the peripheral blood of treatment-naive patients with early RA (208), and higher frequencies of Th17 cells have been detected in the synovial compartment of RA patients, compared to OA patients (209).

Moreover, Th17 cells were associated with clinical parameters, such as disease activity score 28 (DAS28), C-reactive protein (CRP) levels and presence of anti-citrullinated protein antibodies (ACPAs), highly specific for RA (210, 211).

Direct evidence obtained from experimental mouse models confirms the critical role of the IL-23/IL-17 axis in the pathogenesis of arthritis. IL-23p19-deficient (Il23a−/−) mice were protected against the development of collagen-induced arthritis (CIA), a mouse model of RA. IL-17-producing CD4+ T cells were absent in the Il23a−/− mice despite normal induction of IFN-γ-producing CD4+ Th1 cells (212).

Interplay between IL-23 and IL-17 production may be a critical immune pathway and a potential therapeutic target for a range of inflammatory arthritis.

Antibodies against IL-17 (Ixekizumab and Secukinumab) or IL-17R (Brodalumab) have been tested in RA patients (213216). In a phase I RCT of RA patients treated with oral DMARDs, the addition of Ixekizumab improved RA signs and symptoms and disease activity scores such as DAS28, compared to placebo (213). This improvement was confirmed in a phase II study in which Ixekizumab was administered to patients who were naive to biological therapy or resistant to TNF-α inhibitors (217).

In a phase II study enrolling RA patients with inadequate response to methotrexate, greater decreases in DAS28 were observed with Secukinumab than with placebo (214).

Furthermore, patients with active RA who did not respond to DMARDs showed improvements after long-term treatment (52 weeks) with 150 mg of Secukinumab, with ACR50 rates increased from 45% to 16 to 55% at week 16 at week 52 (215). Conversely, Brodalumab showed no evidence of clinical benefit in RA patients in a phase Ib study (213).

In addition, the efficacy of Ustekinumab, a human anti-IL-12/23 p40 monoclonal antibody and human anti-IL-23 monoclonal antibody, Guselkumab, has been evaluated in patients with active RA not responsive to methotrexate therapy. However, no significant clinical improvement was recorded compared to the control group (218).

Recently, the double blockade of IL-17 and TNF-α has been studied using ABT-122, a variable double domain Ig that targets human TNF-α and IL-17 (219). A phase II study showed that there was no significant difference in the ACR20 response at week 12 by the double inhibition of IL-17 and TNF-α compared to treatment with only anti-TNF-α (220). Lastly, double blockade of IL-17A and IL-17F using Bimekizumab in RA patients with inadequate TNF-α response resulted in a greater reduction in DAS28-CRP at week 20 compared to anti-TNF-α inadequate response plus placebo group (221).

Sjögren's Syndrome (SS)

Sjögren's syndrome (SS) is a systemic autoimmune disease characterized by the lymphocytic infiltration into the exocrine glands, mainly salivary and lacrimal glands, and other tissues (222).

The role of the adaptive immune system in the pathogenesis of SS is supported by the presence of ectopic germinal centers in almost 25% of the patients, which promote local expansion of antigen-specific B cells, production of autoantibodies and hypergammaglobulinemia, as well as increased risk of developing non-Hodgkin lymphoma (NHL) (223, 224).

In this regard, the interaction between CD4+ T cells and B cells appears to be a key step in the development of the disease (222).

Although SS has historically been considered a Th1-driven disease, subsequent studies have revealed that, in addition to Th1 cells, several subgroups of CD4+ T cells are involved, follicular T helper cells (Tfh) and Th17. In healthy subjects, Th17 cells play an important role at mucosal barriers and are involved in immune responses. In SS, Th17 cells may be activated by dendritic cells in lymph nodes draining the salivary and lacrimal glands by the production of cytokines as TGF-β and IL-23. In the later phases of the disease, naïve T cells can also be polarized locally in Th17 cells by APC and cytokines as IL-6 and TGF-β (225). IL-23 production by macrophages is also important for maintenance and expansion of Th17 cells by STAT3 activation (226). The main effector cytokines of Th17 cells are IL-17 and IL-22. Recent studies reported that IL-17 protein and mRNA are present within lymphocytic infiltrates of minor salivary gland (MSG) tissue of SS patients.

Furthermore, IL-17 mRNA levels in MSG biopsies were related to the degree of inflammation (226228).

Along with IL-17, the expression of IL-23 and IL-22 was also increased in the inflamed salivary glands of SS patients. IL-17 and IL-22 secretion in the exocrine glands promote inflammation and the induction of matrix metalloproteinases which may cause acinar damage (229).

Moreover, the presence of Th17 cells in SS infiltrate has been hypothesized to be crucial in B cell activation and formation of germinal centers within glands (230).

The IL-23/IL-17 axis also plays a role in the symptoms development, as confirmed by the increased levels of IL-17 and IL-6 protein and mRNA in tears and saliva from SS patients compared to non-SS controls. Additionally, the expression of Th17-associated cytokines correlated with ocular surface parameters, such as Schirmer I Test, break up time (BUT) and corneal fluorescent staining (CFS) (231).

A recent study showed that serum IL-17F levels were significantly increased in SS patients and were associated with high levels of autoantibodies and increased EULAR SS disease activity index (ESSDAI), compared to IL-17A, suggesting the possibility of several pathogenetic roles played by different IL-17 family members (232).

The role of the IL-23/IL-17 pathway in the pathogenesis of SS has been also supported by data from animal models. In C57BL/6.NOD-Aec1Aec2 mouse, a model of spontaneous SS, genetic ablation of IL-17 reduced lymphocytic infiltration and restored glandular function, especially in female animals (233).

In addition, adoptive transfer of Th17 cells induced the development of experimental SS (ESS) in immunized IL-17 knockout mice (234).

Even if Th17 cells are the main source of IL-17 in SS, γδ T cells, NK cells, ILCs and CD8+ T lymphocytes can also produce IL-17 (68). However, recent evidence has shown that CD4 CD8 (double negative, DN) T cells and mast cells can also participate in local IL-17 production in SS (235).

Rituximab treatment demonstrated a significant reduction in IL-17 expression in the salivary glands of SS patients, while factors that are important for maintenance of Th17 cells, as STAT3 and IL-23, are likely not affected (228).

Despite the increase in Th17 cells in SS is well-established, data on the amount of T reg cells are still controversial. Th17 and Treg cells are counter-regulated. IL-6 promotes Th17 differentiation by inhibiting T reg generation (51), while IL-2 acts in the opposite way (236). Since reduced IL-2 levels could underline Th17 upregulation (237), the effects of administration of recombinant IL-2 have been studied in SS. Reduction of glucocorticoid and Hydroxychloroquine (HCQ) in patients treated with IL-2 and restored Th17/Treg balance have been reported (238).

Regarding IL-6 inhibition, as the IL-6 signaling is important for the IL-17 cells differentiation, an effect of tocilizumab on Th17 cells is probable. However, the results of a randomized placebo-controlled study showed that Tocilizumab had no impact on the main symptoms of SS, although improvements in joint involvement were observed (239).

Long-term efficacy of Ustekinumab, a human monoclonal antibody directed against the p40 protein subunit shared by IL-12 and IL-23, in a patient suffering from psoriasis and SS has been recorded on both the cutaneous and joint component (240). However, the inhibition of IL-17 achieved by systemic administration of Secukinumab did not affect the severity of the dry eye (241). Given the pilot role of the IL-23/IL-17 axis in SS pathogenesis, further investigations are needed to confirm the selective blockade of Th17-associated cytokines as a potential therapeutic target.

Systemic Lupus Erythematosus (SLE)

Systemic lupus erythematosus (SLE) is a prototypic systemic autoimmune disease with multiple immunological abnormalities including dysregulation of both T and B lymphocytes and production of autoreactive antibodies directed toward nuclear self-antigens, with immune complex formation and tissue damage (242). A wide number of cytokines is involved in disease pathogenesis. Recently, the role of the IL-23/IL-17 axis has emerged in SLE and has been investigated either in humans or mice. Higher serum levels of IL-17 and IL-23 as well as increased number of Th17 cells has been demonstrated in patients with SLE compared with healthy controls (243, 244). Moreover, T cells that express IL-17 were found within infiltrates in kidney biopsies of patients with active lupus nephritis and significant proportion of these cells are double negative (DN) T cells, which are also expanded in the peripheral blood of patients with SLE, produce significant amounts of IL-17 and contribute to the disease pathogenesis (245). In a murine model of lupus, Zhang et al. detected DN T cells expressing high levels of IL-17A in the kidneys of mice with active nephritis. Interestingly, the lymphocytes isolated from these lupus-prone mice progressively express higher levels of IL-23 receptor as their disease worsens. Treating these lymphocytes in vitro with IL-23, they induce nephritis when transferred to non-autoimmune, control mice. Additionally, the kidneys of these recipient affected mice showed significant Ig and complement deposition in the glomeruli. This finding suggests that IL-23 also promoted an autoimmune B cell response. These data added to previous evidence indicate that DN T cells provide excessive help to B cells, resulting in abnormal production of pathogenic autoantibodies in SLE (246).

Consistently with previous findings, Chen et al. found that circulating Th17 frequency correlated with SLE activity, in particular with Systemic Lupus Erythematosus Disease Activity Index (SLEDAI) and histological activity index in 24 lupus nephritis (LN) patients (247).

The link between IL-23/IL-17 axis and LN is also supported by the observation that high serum levels of IL-23 and IL-17 at baseline predict an unfavorable histopathological response and British Isles Lupus Assessment Group (BILAG)-non-responders had high IL-23, indicating that a number of LN-patients has a Th-17 phenotype that may influence response to treatment.

In addition to LN, high serum levels of IL-23 and Th17 cells are closely related to other SLE manifestations, including vasculitis, serositis, lymphopenia, central nervous system and cutaneous involvement and the production of autoantibodies (antinucleosome antibodies, antiphospholipid antibodies, and anti-SS-B/La antibodies) (244, 248250).

Taken together, these findings suggest that Th17 cells are a promising therapeutic target for SLE.

Previous studies have shown that Hydroxychloroquine (HCQ), an essential drug for the treatment of SLE, can inhibit Th17 cell differentiation and production (251).

In a phase two trial of 102 adult patients with active SLE, the addition to standard-of-care treatment of Ustekinumab, a human IgG1k monoclonal antibody targeting both the IL-12 and IL-23 cytokines, resulted in better efficacy in clinical and laboratory parameters than placebo. In particular, 37 (62%) of 60 patients in the Ustekinumab group and 13 (33%) of 42 patients in the placebo group achieved a SLE disease activity index 2000 responder index-4 (SRI-4) response (difference 28%, p = 0.006) (252).

Few data are available in literature about the anti-IL-17A antibody (Secukinumab). A case of 62-year-old female who presented with psoriasis vulgaris and refractory lupus nephritis successfully treated with Secukinumab was reported. Further studies are needed to test the efficacy of drugs targeting.

Conclusion

This review provides an overview of the critical role played by the IL-23/IL-17 immune axis in a wide variety of inflammatory processes, and summarizes the current knowledge on cytokine milieu that regulates IL-17-producing cells. The growing body of evidence on the relevance of this intricate pathway in autoimmunity has allowed the implementation of treatment options in pathological conditions.

However, further studies are needed to clarify the complexity of IL-17 signaling, in order to allow the discovery of new potential therapeutic targets of inflammatory processes and the availability of cutting edge therapies designed on patients not responsive to standard treatments.

Author Contributions

CS and SF: conceptualization, methodology, formal analysis, investigation, resources, data curation, writing—original draft preparation, writing—review & editing, and visualization. GGr and FC: methodology, formal analysis, data curation, and writing—review & editing. CR: software, methodology, formal analysis, data curation, and writing—review & editing. LL: formal analysis, data curation, and writing—review & editing. GGu: conceptualization, methodology, formal analysis, investigation, resources, data curation, writing—original draft preparation, writing—review & editing, visualization, supervision, and project administration. All authors had access to the study data and reviewed and approved the final manuscript.

Conflict of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

References

1. Tang C, Chen S, Qian H, Huang W. Interleukin-23: as a drug target for autoimmune inflammatory diseases. Immunology. (2012) 135:112–24. doi: 10.1111/j.1365-2567.2011.03522.x

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Kleinschek MA, Muller U, Brodie SJ, Stenzel W, Kohler G, Blumenschein WM, et al. IL-23 enhances the inflammatory cell response in Cryptococcus neoformans infection and induces a cytokine pattern distinct from IL-12. J Immunol. (2006) 176:1098–6. doi: 10.4049/jimmunol.176.2.1098

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Oppmann B, Lesley R, Blom B, Timans JC, Xu Y, Hunte B, et al. Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity. (2000) 13:715–25. doi: 10.1016/S1074-7613(00)00070-4

PubMed Abstract | CrossRef Full Text | Google Scholar

4. McKenzie BS, Kastelein RA, Cua DJ. Understanding the IL-23/IL-17 immune pathway. Trends Immunol. (2006) 27:17–23. doi: 10.1016/j.it.2005.10.003

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Goldberg M, Nadiv O, Luknar-Gabor N, Agar G, Beer Y, Katz Y. Synergism between tumor necrosis factor a and interleukin-17 to induce IL-23 p19 expression in fibroblast- like synoviocytes. Mol Immunol. (2009) 46:1854–9. doi: 10.1016/j.molimm.2009.01.004

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Brentano F, Ospelt C, Stanczyk J, Gay RE, Gay S, Kyburz D. Abundant expression of the interleukin (IL)23 subunit p19, but low levels of bioactive IL23 in the rheumatoid synovium: differential expression and Toll-like receptor-(TLR) dependent regulation of the IL23 subunits, p19 and p40, in rheumatoid arthritis. Ann Rheum Dis. (2009) 68:143–50. doi: 10.1136/ard.2007.082081

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Curtis MM, Way SS. Interleukin-17 in host defence against bacterial, mycobacterial and fungal pathogens. Immunology. (2009) 126:177–85. doi: 10.1111/j.1365-2567.2008.03017.x

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Cua DJ, Sherlock J, Chen Y, Murphy CA, Joyce B, Seymour B, et al. Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature. (2003) 421:744–8. doi: 10.1038/nature01355

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. J Exp Med. (2005) 201:233–40. doi: 10.1084/jem.20041257

PubMed Abstract | CrossRef Full Text | Google Scholar

10. Kopp T, Lenz P, Bello-Fernandez C, Kastelein RA, Kupper TS, Stingl G. IL-23 production by cosecretion of endogenous p19 and transgenic p40 in keratin 14/p40 transgenic mice: evidence for enhanced cutaneous immunity. J Immunol. (2003) 170:5438–44. doi: 10.4049/jimmunol.170.11.5438

PubMed Abstract | CrossRef Full Text | Google Scholar

11. Belladonna ML, Renauld JC, Bianchi R, Vacca C, Fallarino F, Orabona C, et al. IL-23 and IL-12 have overlapping, but distinct, effects on murine dendritic cells. J Immunol. (2002) 168:5448–54. doi: 10.4049/jimmunol.168.11.5448

PubMed Abstract | CrossRef Full Text | Google Scholar

12. Bastos KR, de Deus Vieira de Moraes L, Zago CA, Marinho CR, Russo M, Alvarez JM, et al. Analysis of the activation profile of dendritic cells derived from the bone marrow of interleukin-12/interleukin-23-deficient mice. Immunology. (2005) 114:499–506. doi: 10.1111/j.1365-2567.2005.02118.x

PubMed Abstract | CrossRef Full Text | Google Scholar

13. Goriely S, Goldman M. The interleukin-12 family: new players in transplantation immunity? Am J Transplant. (2007) 7:278–84. doi: 10.1111/j.1600-6143.2006.01651.x

PubMed Abstract | CrossRef Full Text | Google Scholar

14. Goriely S, Neurath MF, Goldman M. How microorganisms tip the balance between interleukin-12 family members. Nat Rev Immunol. (2008) 8:81–6. doi: 10.1038/nri2225

PubMed Abstract | CrossRef Full Text | Google Scholar

15. Ferber IA, Brocke S, Taylor-Edwards C, Ridgway W, Dinisco C, Steinman L, Dalton D, et al. Mice with a disrupted IFN-c gene are susceptible to the induction of experimental autoimmune encephalomyelitis (EAE). J Immunol. (1996) 156:5–7.

Google Scholar

16. Vermeire K, Heremans H, Vandeputte M, Huang S, Billiau A, Matthys P. Accelerated collagen-induced arthritis in IFN-c receptor-deficient mice. J Immunol. (1997) 158:5507–13.

Google Scholar

17. Manoury-Schwartz B, Chiocchia G, Bessis N, Abehsira-Amar O, Batteux F, Muller S, et al. High susceptibility to collagen induced arthritis in mice lacking IFN-c receptors. J Immunol. (1997) 158:5501–6.

Google Scholar

18. Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-c: an overview of signals,mechanisms and functions. J Leukoc Biol. (2004) 75:163–89. doi: 10.1189/jlb.0603252

PubMed Abstract | CrossRef Full Text | Google Scholar

19. Teng MWL, Bowman EP, McElwee JJ, Smyth MJ, Casanova JL, Cooper AM, et al. IL-12 and IL-23 cytokines: from discovery to targeted therapies for immune-mediated inflammatory diseases. Nat Med. (2015) 21:719–29. doi: 10.1038/nm.3895

PubMed Abstract | CrossRef Full Text | Google Scholar

20. Hayashi F, Yanagawa Y, Onoe K, Iwabuchi K. Dendritic cell differentiation with prostaglandin E results in selective attenuation of the extracellular signal-related kinase pathway and decreased interleukin-23 production. Immunology. (2010) 131:67–76. doi: 10.1111/j.1365-2567.2010.03275.x

PubMed Abstract | CrossRef Full Text | Google Scholar

21. Chizzolini C, Chicheportiche R, Alvarez M, de Rham C, Roux-Lombard P, Ferrari-Lacraz S, et al. Prostaglandin E2 synergistically with interleukin-23 favors human Th17 expansion. Blood. (2008) 112:3696–703. doi: 10.1182/blood-2008-05-155408

PubMed Abstract | CrossRef Full Text | Google Scholar

22. Langrish CL, McKenzie BS, Wilson NJ, de Waal Malefyt R, Kastelein RA, Cua DJ. IL-12 and IL-23: master regulators of innate and adaptive immunity. Immunol Rev. (2004) 202:96–105 doi: 10.1111/j.0105-2896.2004.00214.x

PubMed Abstract | CrossRef Full Text | Google Scholar

23. Trinchieri G. Interleukin-12 and the regulation of innate resistance and adaptive immunity. Nat Rev Immunol. (2003) 3:133–46. doi: 10.1038/nri1001

PubMed Abstract | CrossRef Full Text | Google Scholar

24. Kreymborg K, Etzensperger R, Dumoutier L, Haak S, Rebollo A, Buch T, et al. IL-22 is expressed by Th17 cells in an IL-23-dependent fashion, but not required for the development of autoimmune encephalomyelitis. J. Immunol. (2007) 179:8098–104. doi: 10.4049/jimmunol.179.12.8098

PubMed Abstract | CrossRef Full Text | Google Scholar

25. Meeks KD, Sieve AN, Kolls JK, Ghilardi N, Berg RE. IL-23 is required for protection against systemic infection with Listeria monocytogenes. J. Immunol. (2009) 183:8026–34. doi: 10.4049/jimmunol.0901588

PubMed Abstract | CrossRef Full Text | Google Scholar

26. Riol-Blanco L, Lazarevic V, Awasthi A, Mitsdoerffer M, Wilson BS, Croxford A, et al. IL-23 receptor regulates unconventional IL-17-producing T cells that control bacterial infections. J. Immunol. (2010) 184:1710–20. doi: 10.4049/jimmunol.0902796

PubMed Abstract | CrossRef Full Text | Google Scholar

27. Van Belle AB, de Heusch M, Lemaire MM, Hendrickx E, Warnier G, Dunussi-Joannopoulos K, et al. IL-22 is required for imiquimod-induced psoriasiform skin inflammation in mice. J. Immunol. (2012) 188:462–9. doi: 10.4049/jimmunol.1102224

PubMed Abstract | CrossRef Full Text | Google Scholar

28. Wakita D, Sumida K, Iwakura Y, Nishikawa H, Ohkuri T, Chamoto K, et al. Tumor-infiltrating IL-17-producing gammadelta T cells support the progression of tumor by promoting angiogenesis. Eur J Immunol. (2010) 40:1927–37. doi: 10.1002/eji.200940157

PubMed Abstract | CrossRef Full Text | Google Scholar

29. Sutton CE, Lalor SJ, Sweeney CM, Brereton CF, Lavelle EC, Mills KH. Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. Immunity. (2009) 31:331–41. doi: 10.1016/j.immuni.2009.08.001

PubMed Abstract | CrossRef Full Text | Google Scholar

30. Petermann F, Rothhammer V, Claussen MC, Haas JD, Blanco LR, Heink S, Prinz I, et al. Gammadelta T cells enhance autoimmunity by restraining regulatory T cell responses via an interleukin-23-dependent mechanism. Immunity. (2010) 33:351–63. doi: 10.1016/j.immuni.2010.08.013

PubMed Abstract | CrossRef Full Text | Google Scholar

31. Parham C, Chirica M, Timans J, Vaisberg E, Travis M, Cheung J, et al. A receptor for the heterodimeric cytokine IL-23 is composed of IL-12Rbeta1 a novel cytokine receptor subunit, IL-23RJ. Immunol. (2002) 168:5699–708. doi: 10.4049/jimmunol.168.11.5699

PubMed Abstract | CrossRef Full Text | Google Scholar

32. Zhang GX, Yu S, Gran B, Li J, Siglienti I, Chen X, et al. Role of IL-12 receptor beta 1 in regulation of T cell response by APC in experimental autoimmune encephalomyelitis. J. Immunol. (2003) 171:4485–92. doi: 10.4049/jimmunol.171.9.4485

PubMed Abstract | CrossRef Full Text | Google Scholar

33. Pahan K, Jana M. Induction of lymphotoxin-a by interleukin-12 p40 homodimer, the so-called biologically inactive molecule, but not IL-12 p70. Immunology. (2009) 127:312–25. doi: 10.1111/j.1365-2567.2008.02985.x

CrossRef Full Text | Google Scholar

34. Tato CM, Cua DJ. Reconciling id, ego, and superego within interleukin-23. Immunol Rev. (2008) 226:103–11. doi: 10.1111/j.1600-065X.2008.00715.x

PubMed Abstract | CrossRef Full Text | Google Scholar

35. Kastelein RA, Hunter CA, Cua DJ. Discovery and biology of IL-23 and IL-27: related but functionally distinct regulators of inflammation. Annu Rev Immunol. (2007) 25:221–42. doi: 10.1146/annurev.immunol.22.012703.104758

PubMed Abstract | CrossRef Full Text | Google Scholar

36. Zou J, Presky DH, Wu CY, Gubler U. Differential associations between the cytoplasmic regions of the interleukin-12 receptor subunits beta1 and beta2 and JAK kinases. J Biol Chem. (1997) 272:6073–7. doi: 10.1074/jbc.272.9.6073

PubMed Abstract | CrossRef Full Text | Google Scholar

37. Singh RP, Hasan S, Sharma S, Nagra S, Yamaguchi DT, Wong DT, et al. Th17 cells in inflammation and autoimmunity. Autoimmun Rev. (2014) 13:1174–81. doi: 10.1016/j.autrev.2014.08.019

PubMed Abstract | CrossRef Full Text | Google Scholar

38. Ghilardi N, Kljavin N, Chen Q, Lucas S, Gurney AL, De Sauvage FJ. Compromised humoral and delayed-type hypersensitivity responses in IL-23-deficient mice. J Immunol. (2004) 172:2827–33. doi: 10.4049/jimmunol.172.5.2827

PubMed Abstract | CrossRef Full Text | Google Scholar

39. Kikly K, Liu L, Na S, Sedgwick JD. The IL-23/Th(17) axis: therapeutic targets for autoimmune inflammation. Curr Opin Immunol. (2006) 18:670–5. doi: 10.1016/j.coi.2006.09.008

PubMed Abstract | CrossRef Full Text | Google Scholar

40. McGeachy MJ, Chen Y, Tato C M, Laurence A, Joyce-Shaikh B, et al. Interleukin 23 receptor is essential for terminal differentiation of effector T helper type 17 cells in vivo. Nat Immunol. (2009) 10:314–24. doi: 10.1038/ni.1698

CrossRef Full Text | Google Scholar

41. Weaver CT, Hatton RD, Mangan PR, Harrington LE. IL-17 family cytokines and the expanding diversity of effector T cell lineages. Annu Rev Immunol. (2007) 25:821–52. doi: 10.1146/annurev.immunol.25.022106.141557

PubMed Abstract | CrossRef Full Text | Google Scholar

42. Gu C, Wu L, Li X. IL-17 family: cytokines, receptors and signaling. Cytokine. (2013) 64:477–85. doi: 10.1016/j.cyto.2013.07.022

PubMed Abstract | CrossRef Full Text | Google Scholar

43. Shin HC, Benbernou N, Esnault S, Guenounou M. Expression of IL-17 in human memory CD45RO+ T lymphocytes and its regulation by protein kinase A pathway. Cytokine. (1999) 11:257–66. doi: 10.1006/cyto.1998.0433

PubMed Abstract | CrossRef Full Text | Google Scholar

44. Roark CL, Simonian PL, Fontenot AP, Born WK, O'Brien RL. gammadelta T cells: an important source of IL-17. Curr Opin Immunol. (2008) 20:353–7. doi: 10.1016/j.coi.2008.03.006

PubMed Abstract | CrossRef Full Text | Google Scholar

45. Rachitskaya AV, Hansen AM, Horai R, Li Z, Villasmil R, Luger D, et al. Cutting edge: NKT cells constitutively express IL-23 receptor and RORgammat and rapidly produce IL-17 upon receptor ligation in an IL-6-independent fashion. J Immunol. (2008) 180:5167–71. doi: 10.4049/jimmunol.180.8.5167

PubMed Abstract | CrossRef Full Text | Google Scholar

46. Aggarwal S, Ghilardi N, Xie MH, de Sauvage FJ, Gurney AL. Interleukin-23 promotes a distinct CD4 T cell activation state characterized by the production of interleukin-17. J. Biol. Chem. (2003) 278:1910–14. doi: 10.1074/jbc.M207577200

PubMed Abstract | CrossRef Full Text | Google Scholar

47. Awasthi A, Riol-Blanco L, Jager A, Korn T, Pot C, Galileos G, et al. Cutting edge: IL-23 receptor gfp reporter mice reveal distinct populations of IL-17-producing cells. J. Immunol. (2009) 182:5904–8. doi: 10.4049/jimmunol.0900732

PubMed Abstract | CrossRef Full Text | Google Scholar

48. Nurieva R, Yang XO, Martinez G, Zhang Y, Panopoulos AD, Ma L, et al. Essential autocrine regulation by IL-21 in the generation of inflammatory T cells. Nature. (2007) 448:480–3. doi: 10.1038/nature05969

PubMed Abstract | CrossRef Full Text | Google Scholar

49. Zhou L, Ivanov II, Spolski R, Min R, Shenderov K, Egawa T, et al. IL-6 programs T(H)-17 cell differentiation by promoting sequential engagement of the IL-21 and IL-23 pathways. Nat. Immunol. (2007) 8:967–74. doi: 10.1038/ni1488

PubMed Abstract | CrossRef Full Text | Google Scholar

50. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, Weiner HL, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. (2006) 441:235–8. doi: 10.1038/nature04753

PubMed Abstract | CrossRef Full Text | Google Scholar

51. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. (2006) 24:179–89. doi: 10.1016/j.immuni.2006.01.001

PubMed Abstract | CrossRef Full Text | Google Scholar

52. McGeachy MJ, Bak-Jensen KS, Chen Y, Tato CM, Blumenschein W, McClanahan T, et al. TGF-beta and IL-6 drive the production of IL-17 and IL-10 by T cells and restrain T(H)-17 cell-mediated pathology. Nat Immunol. (2007) 8:1390–7. doi: 10.1038/ni1539

PubMed Abstract | CrossRef Full Text | Google Scholar

53. Mangan PR, Harrington LE, O'Quinn DB, Helms WS, Bullard DC, Elson CO, Hatton RD, et al. Transforming growth factor-beta induces development of the T(H)17 lineage. Nature. (2006) 441:231–4. doi: 10.1038/nature04754

PubMed Abstract | CrossRef Full Text | Google Scholar

54. Chen Z, Tato CM, Muul L, Laurence A, O'Shea JJ. Distinct regulation of interleukin-17 in human T helper lymphocytes. Arthritis Rheum. (2007) 56:2936–46. doi: 10.1002/art.22866

PubMed Abstract | CrossRef Full Text | Google Scholar

55. Wilson NJ, Boniface K, Chan JR, McKenzie BS, Blumenschein WM, Mattson JD, et al. Development, cytokine profile and function of human interleukin 17-producing helper T cells. Nat Immunol. (2007) 8:950–7. doi: 10.1038/ni1497

PubMed Abstract | CrossRef Full Text | Google Scholar

56. 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. Nat Immunol. (2007) 8:942–9. doi: 10.1038/ni1496

PubMed Abstract | CrossRef Full Text | Google Scholar

57. Morishima N, Mizoguchi I, Takeda K, Mizuguchi J, Yoshimoto T. TGF-beta is necessary for induction of IL-23R and Th17 differentiation by IL-6 and IL-23. Biochem Biophys Res Commun. (2009) 386:105–10. doi: 10.1016/j.bbrc.2009.05.140

PubMed Abstract | CrossRef Full Text | Google Scholar

58. Korn T, Bettelli E, Gao W, Awasthi A, Jager A, Strom TB, et al. IL-21 initiates an alternative pathway to induce proinflammatory T(H)17 cells. Nature. (2007) 448:484–7. doi: 10.1038/nature05970

PubMed Abstract | CrossRef Full Text | Google Scholar

59. Huber M, Brustle A, Reinhard K, Guralnik A, Walter G, Mahiny A, von Low E, et al. IRF4 is essential for IL-21-mediated induction, amplification, and stabilization of the Th17 phenotype. Proc Natl Acad Sci USA. (2008) 105:20846–51. doi: 10.1073/pnas.0809077106

PubMed Abstract | CrossRef Full Text | Google Scholar

60. Wei L, Laurence A, Elias KM, O'Shea JJ. IL-21 is produced by Th17 cells and drives IL-17 production in a STAT3-dependent manner. J. Biol. Chem. (2007) 282:34605–10. doi: 10.1074/jbc.M705100200

PubMed Abstract | CrossRef Full Text | Google Scholar

61. Ivanov II, Zhou L, Littman DR. Transcriptional regulation of Th17 cell differentiation. Semin Immunol. (2007) 19:409–17. doi: 10.1016/j.smim.2007.10.011

PubMed Abstract | CrossRef Full Text | Google Scholar

62. Manel N, Unutmaz D, Littman DR. The differentiation of human T(H)- 17 cells requires transforming growth factor-beta and induction of the nuclear receptor RORgammat. Nat Immunol. (2008) 9:641–9. doi: 10.1038/ni.1610

PubMed Abstract | CrossRef Full Text | Google Scholar

63. Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, 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) 6:1123–32. doi: 10.1038/ni1254

PubMed Abstract | CrossRef Full Text | Google Scholar

64. Park H, Li Z, Yang XO, Chang SH, Nurieva R, Wang YH, et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat Immunol. (2005) 6:1133–4. doi: 10.1038/ni1261

PubMed Abstract | CrossRef Full Text | Google Scholar

65. Angkasekwinai P, Park H, Wang YH, Chang SH, Corry DB, Liu YJ, et al. Interleukin 25 promotes the initiation of proallergic type 2 responses. J Exp Med. (2007) 204:1509–17. doi: 10.1084/jem.20061675

PubMed Abstract | CrossRef Full Text | Google Scholar

66. Batten M, Li J, Yi S, Kljavin NM, Danilenko DM, Lucas S, et al. Interleukin 27 limits autoimmune encephalomyelitis by suppressing the development of interleukin 17-producing T cells. Nat Immunol. (2006) 7:929–36. doi: 10.1038/ni1375

PubMed Abstract | CrossRef Full Text | Google Scholar

67. Stumhofer JS, Laurence A, Wilson EH, Huang E, Tato CM, Johnson LM, et al. Interleukin 27 negatively regulates the development of interleukin 17-producing T helper cells during chronic inflammation of the central nervous system. Nat Immunol. (2006) 7:937–45. doi: 10.1038/ni1376

PubMed Abstract | CrossRef Full Text | Google Scholar

68. Patel DD, Kuchroo VK. Th17 cell pathway in human immunity: lessons from genetics and therapeutic interventions. Immunity. (2015) 43:1040–51. doi: 10.1016/j.immuni.2015.12.003

PubMed Abstract | CrossRef Full Text | Google Scholar

69. Shalom-Barak T, Quach J, Lotz M. Interleukin-17-induced gene expression in articular chondrocytes is associated with activation of mitogen-activated protein kinases and NF-kappaB. J Biol Chem. (1998) 273:27467–73. doi: 10.1074/jbc.273.42.27467

PubMed Abstract | CrossRef Full Text | Google Scholar

70. Adami S, Cavani A, Rossi F, Girolomoni G. The role of interleukin-17A in psoriatic disease. BioDrugs. (2014) 28:487–97. doi: 10.1007/s40259-014-0098-x

CrossRef Full Text | Google Scholar

71. Laan M, Cui ZH, Hoshino H, Lötvall J, Sjöstrand M, Gruenert D. C, et al. Neutrophil recruitment by human IL-17 via C-X-C chemokine release in the airways. J Immunol. (1999) 162:2347–52.

PubMed Abstract | Google Scholar

72. Happel KI, Zheng M, Young E, Quinton LJ, Lockhart E, Ramsay AJ, et al. Cutting edge: roles of Toll-like receptor 4 and IL-23 in IL-17 expression in response to Klebsiella pneumoniae infection. J Immunol. (2003) 170:4432–6. doi: 10.4049/jimmunol.170.9.4432

PubMed Abstract | CrossRef Full Text | Google Scholar

73. Happel KI, Dubin PJ, Zheng M, Ghilardi N, Lockhart C, Quinton LJ, et al. Divergent roles of IL-23 and IL-12 in host defense against Klebsiella pneumoniae. J Exp Med. (2005) 202:761–9. doi: 10.1084/jem.20050193

PubMed Abstract | CrossRef Full Text | Google Scholar

74. Meller S, Di Domizio J, Voo KS, Friedrich HC, Chamilos G, Ganguly D, et al. T(H)17 cells promote microbial killing and innate immune sensing of DNA via interleukin 26. Nat Immunol. (2015) 16:970–9. doi: 10.1038/ni.3211

PubMed Abstract | CrossRef Full Text | Google Scholar

75. Conti HR, Gaffen SL. IL-17-mediated immunity to the opportunistic fungal pathogen candida albicans. J Immunol. (2015) 195:780–8. doi: 10.4049/jimmunol.1500909

PubMed Abstract | CrossRef Full Text | Google Scholar

76. Centers for Disease Control (CDC). Pneumocystis pneumonia—Los Angeles. MMWR. (1981) 30:250–2.

Google Scholar

77. Greenspan D, Greenspan JS. Oral mucosal manifestations of AIDS. Dermatol. Clin. (1987) 5:733–737. doi: 10.1016/S0733-8635(18)30717-4

CrossRef Full Text | Google Scholar

78. Acosta-Rodriguez EV, Rivino L, Geginat J, Jarrossay D, Gattorno M, Lanzavecchia A, et al. Surface phenotype and antigenic specificity of human interleukin 17-producing T helper memory cells. Nat Immunol. (2007) 8:639–46. doi: 10.1038/ni1467

PubMed Abstract | CrossRef Full Text | Google Scholar

79. Zielinski CE, Mele F, Aschenbrenner D, Jarrossay D, Ronchi F, Gattorno M, et al. Pathogen-induced human TH17 cells produce IFN-γ or IL-10 and are regulated by IL-1β. Nature. (2012) 484:514–8. doi: 10.1038/nature10957

PubMed Abstract | CrossRef Full Text | Google Scholar

80. Huang W, Na L, Fidel PL, Schwarzenberger P. Requirement of interleukin-17A for systemic anti-Candida albicans host defense in mice. J Infect Dis. (2004) 190:624–31. doi: 10.1086/422329

PubMed Abstract | CrossRef Full Text | Google Scholar

81. Conti HR, Shen F, Nayyar N, Stocum E, Sun JN, Lindemann MJ, et al. Th17 cells and IL-17 receptor signaling are essential for mucosal host defense against oral candidiasis. J Exp Med. (2009) 206:299–311. doi: 10.1084/jem.20081463

PubMed Abstract | CrossRef Full Text | Google Scholar

82. Ferreira MC, Whibley N, Mamo AJ, Siebenlist U, Chan YR, Gaffen SL. Interleukin-17-induced protein lipocalin 2 is dispensable for immunity to oral candidiasis. Infect Immun. (2014) 82:1030–5. doi: 10.1128/IAI.01389-13

PubMed Abstract | CrossRef Full Text | Google Scholar

83. Gaffen SL, Jain R, Garg AV, Cua DJ. The IL-23-IL-17 immune axis: from mechanisms to therapeutic testing. Nat Rev Immunol. (2014) 14:585–600. doi: 10.1038/nri3707

PubMed Abstract | CrossRef Full Text | Google Scholar

84. Starnes T, Broxmeyer HE, Robertson MJ, Hromas R. Cutting edge: IL−17 D, a novel member of the IL-17 family, stimulates cytokine production and inhibits hemopoiesis. J Immunol. (2002) 169:642–6. doi: 10.4049/jimmunol.169.2.642

CrossRef Full Text | Google Scholar

85. Dragon S, Saffar AS, Shan L, Gounni AS. IL-17 attenuates the anti-apoptotic effects of GM-CSF in human neutrophils. Mol Immunol. (2008) 45:160–8. doi: 10.1016/j.molimm.2007.04.027

PubMed Abstract | CrossRef Full Text | Google Scholar

86. Numasaki M, Fukushi J, Ono M, Narula SK, Zavodny PJ, Kudo T, et al. Interleukin-17 promotes angiogenesis and tumor growth. Blood. (2003) 101:2620–7 doi: 10.1182/blood-2002-05-1461

PubMed Abstract | CrossRef Full Text | Google Scholar

87. Jovanovic DV, Martel-Pelletier J, Di Battista JA, Mineau F, Jolicoeur F. C, Benderdour M, et al. Stimulation of 92-kd gelatinase (matrix metalloproteinase 9) production by interleukin-17 in human monocyte/macrophages: a possible role in rheumatoid arthritis. Arthritis Rheum. (2000) 43:1134–44. doi: 10.1002/1529-0131(200005)43:5<1134::AID-ANR24>3.0.CO;2-#

PubMed Abstract | CrossRef Full Text | Google Scholar

88. Ruddy MJ, Wong GC, Liu XK, Yamamoto H, Kasayama S, Kirkwood KL, et al. Functional cooperation between interleukin-17 and tumor necrosis factor-alpha is mediated by CCAAT/enhancer-binding protein family members. J Biol Chem. (2004) 279:2559–2567. doi: 10.1074/jbc.M308809200

PubMed Abstract | CrossRef Full Text | Google Scholar

89. Kotake S, Udagawa N, Takahashi N, Matsuzaki K, Itoh K, Ishiyama S, et al. IL-17 in synovial fluids from patients with rheumatoid arthritis is a potent stimulator of osteoclastogenesis. J Clin Invest. (1999) 103:1345–52. doi: 10.1172/JCI5703

PubMed Abstract | CrossRef Full Text | Google Scholar

90. Matusevicius D, Kivisakk P, He B, Kostulas N, Ozenci V, Fredrikson S, et al. Interleukin-17 mRNA expression in blood and CSF mononuclear cells is augmented in multiple sclerosis. Mult Scler. (1999) 5:101–4. doi: 10.1177/135245859900500206

PubMed Abstract | CrossRef Full Text | Google Scholar

91. Lock C, Hermans G, Pedotti R, Brendolan A, Schadt E, Garren H, et al. Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat Med. (2002) 8:500–8. doi: 10.1038/nm0502-500

PubMed Abstract | CrossRef Full Text | Google Scholar

92. Fujino S, Andoh A, Bamba S, Ogawa A, Hata K, Araki Y, et al. Increased expression of interleukin 17 in inflammatory bowel disease. Gut. (2003) 52:65–70. doi: 10.1136/gut.52.1.65

PubMed Abstract | CrossRef Full Text | Google Scholar

93. Taurog JD, Chhabra A, Colbert A. Ankylosing Spondylitis and Axial Spondyloarthritis. New Engl J Med. (2016) 374:2563–74. doi: 10.1056/NEJMra1406182

CrossRef Full Text | Google Scholar

94. Rudwaleit M, Van der Heijde D, Landewé R, Listing J, Akkoc N, Brandt J, et al. The development of Assessment of SpondyloArthritis international Society classification criteria for axial spondyloarthritis (part II): validation and final selection. Ann Rheum Dis. (2009) 68:777–83. doi: 10.1136/ard.2009.108233

PubMed Abstract | CrossRef Full Text | Google Scholar

95. Van der Heijde D, Ramiro S, Landewé R, Baraliakos X, Van den Bosch F, Sepriano A, et al. 2016 Update of the ASAS-EULAR management recommendations for axial spondyloarthritis. Ann Rheum Dis. (2017) 76:978–91. doi: 10.1136/annrheumdis-2016-210770

PubMed Abstract | CrossRef Full Text | Google Scholar

96. Zhu W, He X, Cheng K, Zhang L, Di Chen Wang X, et al. Ankylosing spondylitis: etiology, pathogenesis, and treatments. Bone Res. (2019) 7:22. doi: 10.1038/s41413-019-0057-8

PubMed Abstract | CrossRef Full Text | Google Scholar

97. Brown MA, Kennedy LG, MacGregor AJ, Darke C, Duncan E, Shatford JL, et al. Susceptibility to ankylosing spondylitis in twins: the role of genes, HLA, and the environment. Arthritis Rheum. (1997) 40:1823–8. doi: 10.1002/art.1780401015

PubMed Abstract | CrossRef Full Text | Google Scholar

98. Chen B, Li J, He C, Li D, Tong W, Zou Y, et al. Role of HLA-B27 in the pathogenesis of ankylosing spondylitis. Mol Med Rep. (2017) 15:1943–51. doi: 10.3892/mmr.2017.6248

PubMed Abstract | CrossRef Full Text | Google Scholar

99. Brown MA, Kenna T, Wordsworth BP. Genetics of ankylosing spondylitis-insights into pathogenesis. Nat Rev Rheumatol. (2016) 12:81–91. doi: 10.1038/nrrheum.2015.133

CrossRef Full Text | Google Scholar

100. Colbert RA, Tran MT, Layh-Schmitt G. HLA-B27 misfolding and ankylosing spondylitis. Mol Immunol. (2014) 57:44–51. doi: 10.1016/j.molimm.2013.07.013

PubMed Abstract | CrossRef Full Text | Google Scholar

101. Watad A, Bridgewood C, Russell T, Marzo-Ortega H, Cuthbert R, McGonagle D. The early phases of ankylosing spondylitis: emerging insights from clinical and basic science. Front Immunol. (2018) 9:2668. doi: 10.3389/fimmu.2018.02668

PubMed Abstract | CrossRef Full Text | Google Scholar

102. DeLay ML, Turner JM, Klenk EI, Smith JA, Sowders DP, Colbert RA. HLA-B27 misfolding and the unfolded protein response augment interleukin-23 production and are associated with Th17 activation in transgenic rats. Arthritis Rheum. (2009) 60:2633–43. doi: 10.1002/art.24763

PubMed Abstract | CrossRef Full Text | Google Scholar

103. Turner MJ, Sowders DP, DeLay ML, Mohapatra R, Bai S, Smith JA, et al. HLA-B27 misfolding in transgenic rats is associated with activation of the unfolded protein response. J Immunol. (2005) 175:2438–48. doi: 10.4049/jimmunol.175.4.2438

PubMed Abstract | CrossRef Full Text | Google Scholar

104. Chan AT, Kollnberger SD, Wedderburn LR, Bowness P. Expansion and enhanced survival of natural killer cells expressing the killer immunoglobulin-like receptor KIR3DL2 in spondylarthritis. Arthritis and Rhum. (2005) 54:3586–95. doi: 10.1002/art.21395

PubMed Abstract | CrossRef Full Text | Google Scholar

105. International Genetics of Ankylosing Spondylitis Consortium (IGAS) Australo-Anglo-American Spondyloarthritis Consortium (TASC); Groupe Française d'Etude Génétique des Spondylarthrites (GFEGS); Nord-Trøndelag Health Study (HUNT); Spondyloarthritis Research Consortium of Canada (SPARCC); Wellcome Trust Case Control Consortium 2 (WTCCC2). Identification of multiple risk variants for ankylosing spondylitis through high-density genotyping of immune-related loci. Nat Genet. (2013) 45:730–8. doi: 10.1038/ng.2667

CrossRef Full Text | Google Scholar

106. Wellcome Trust Case Control Consortium Australo-Anglo-American Spondylitis Consortium (TASC) Biologics in RA Genetics and Genomics Study Syndicate (BRAGGS) Steering Committee Association scan of 14 500 nonsynonymous SNPs in four diseases identifies autoimmunity variants. Nat Genet. (2007) 39:1329–37. doi: 10.1038/ng.2007.17

CrossRef Full Text | Google Scholar

107. York IA, Chang SC, Saric T, Keys JA, Favreau JM, Goldberg AL, et al. The ER aminopeptidase ERAP1 enhances or limits antigen presentation by trimming epitopes to 8–9 residues. Nat Immunol. (2002) 3:1177–84. doi: 10.1038/ni860

PubMed Abstract | CrossRef Full Text | Google Scholar

108. Guiliano DB, Fussell H, Lenart I, Tsao E, Nesbeth D, Fletcher AJ, et al. Endoplasmic reticulum degradation–enhancing α-mannosidase–like protein 1 targets misfolded hla–b27 dimers for endoplasmic reticulum–associated degradation. Arthritis Rheumatol. (2014) 66:2976–88. doi: 10.1002/art.38809

PubMed Abstract | CrossRef Full Text | Google Scholar

109. Cortes A, Pulit SL, Weisman MA, Pointon JJ, Robinson PC, Weisman MH, et al. Major histocompatibility complex associations of ankylosing spondylitis are complex and involve further epistasis with ERAP1. Nat Commun. (2015) 6:7146. doi: 10.1038/ncomms8146

CrossRef Full Text | Google Scholar

110. Rahman P, Inman RD, Gladman DD, Reeve JP, Peddle L, Maksymowych WP. Association of interleukin-23 receptor variants with ankylosing spondylitis. Arthritis Rheum. (2008) 58:1020–5. doi: 10.1002/art.23389

PubMed Abstract | CrossRef Full Text | Google Scholar

111. Ellinghaus D, Jostins L, Spain SL, Cortes A, Bethune J, Han B, et al. Analysis of five chronic inflammatory diseases identifies 27 new associations and highlights disease-specific patterns at shared loci. Nature Genetics. (2016) 48:510–8. doi: 10.1038/ng.3528

PubMed Abstract | CrossRef Full Text | Google Scholar

112. Di Meglio P, Di Cesare A, Laggner U, Chu CC, Napolitano L, Villanova F, et al. The IL23R R381Q gene variant protects against immune-mediated diseases by impairing IL-23-induced Th17 effector response in humans. PLoS ONE. (2011) 6:e17160. doi: 10.1371/journal.pone.0017160

PubMed Abstract | CrossRef Full Text | Google Scholar

113. Mei Y, Pan F, Gao J, Ge R, Duan Z, Zeng Z, et al. Increased serum IL-17 and IL-23 in the patient with ankylosing spondylitis. Clin Rheum. (2011) 30:269–73. doi: 10.1007/s10067-010-1647-4

PubMed Abstract | CrossRef Full Text | Google Scholar

114. Jandus C, Jilles B, Rivals B, Rivals JP, Dudler J, Speiser D. Increased numbers of circulating polyfunctional Th17 memory cells in patients with seronegative spondylarthrities. Arthr Rheuma. (2008) 58:2307–17. doi: 10.1002/art.23655

PubMed Abstract | CrossRef Full Text | Google Scholar

115. Rizzo A, Guggino G, Ferrante A, Ciccia F. Role of subclinical gut inflammation in the pathogenesis of spondyloarthritis. Front Med. (2018) 1:5:63. doi: 10.3389/fmed.2018.00063

PubMed Abstract | CrossRef Full Text | Google Scholar

116. Van Praet L, Van den Bosch FE, Jacques P, Carron P, Jans L, Colman R, et al. Microscopic gut inflammation in axial spondyloarthritis: a multiparametric predictive model. Ann Rheum Dis. (2013). 72:414–7. doi: 10.1136/annrheumdis-2012-202135

PubMed Abstract | CrossRef Full Text | Google Scholar

117. Van Praet L, Jans L, Carron P, Jacques P, Glorieus R, Colman R, et al. Degree of bone marrow oedema in sacroiliac joints of patients with axial spondyloarthritis is linked to gut inflammation and male sex: results from the GIANT cohort. Ann Rheum Dis. (2014) 73:1186–9. doi: 10.1136/annrheumdis-2013-203854

PubMed Abstract | CrossRef Full Text | Google Scholar

118. Costello M A, Ciccia F, Dana W, Warrington W, Robinson PC, et al. Brief report: intestinal dysbiosis in ankylosing spondylitis. Arthritis Rheumatol. (2015) 67:686–91. doi: 10.1002/art.38967

PubMed Abstract | CrossRef Full Text | Google Scholar

119. Ciccia F, Guggino G, Rizzo A, Riccardo A, Luchetti MM, Milling S, et al. Dysbiosis and zonulin upregulation alter gut epithelial and vascular barriers in patients with ankylosing spondylitis. Ann Rheum Dis. (2017) 76:1123–32. doi: 10.1136/annrheumdis-2016-210000

PubMed Abstract | CrossRef Full Text | Google Scholar

120. Ciccia F, Bombardieri M, Principato A, Giardina A, Tripodo C, Porcasi R, et al. Overexpression of interleukin-23, but not interleukin-17, as an immunologic signature of subclinical intestinal inflammation in ankylosing spondylitis. Arthritis Rheum. (2009) 60:955–65. doi: 10.1002/art.24389

PubMed Abstract | CrossRef Full Text | Google Scholar

121. Ciccia F, Guggino G, Rizzo A, Saieva L, Peralta S, Giardina A, et al. Type 3 innate lymphoid cells producing IL-17 and IL-22 are expanded in the gut, in the peripheral blood, synovial fluid and bone marrow of patients with ankylosing spondylitis. Ann Rheum Dis. (2015) 74:1739–47. doi: 10.1136/annrheumdis-2014-206323

PubMed Abstract | CrossRef Full Text | Google Scholar

122. Kenna TJ, Davidson SI, Duan R, Bradbury LA, McFarlane J, Smith M, et al. Enrichment of circulating interleukin-17-secreting interleukin-23 receptor-positive γ/δ T cells in patients with active ankylosing spondylitis. Arthritis Rheum. (2012) 64:1420–9. doi: 10.1002/art.33507

PubMed Abstract | CrossRef Full Text | Google Scholar

123. Cuthbert RJ, Watad A, Fragkakis EM, Dunsmuir R, Loughenbury P, Khan A, et al. Evidence that tissue resident human enthesis γδT- cells can produce IL-17A independently of IL-23R transcript expression. Ann Rheum Dis. (2019) 78:1559–65. doi: 10.1136/annrheumdis-2019-215210

PubMed Abstract | CrossRef Full Text | Google Scholar

124. Toussirot E, Laheurte C, Gaugler B, Gabriel D, Saas P. Increased IL-22- and IL-17A-producing mucosal-associated invariant t cells in the peripheral blood of patients with ankylosing spondylitis. Front Immunol. (2018); 9:1610. doi: 10.3389/fimmu.2018.01610

PubMed Abstract | CrossRef Full Text | Google Scholar

125. Hayashi E, Chiba A, Tada K, Haga K, Kitagaichi M, Nakajima S, et al. Involvement of mucosal-associated invariant t cells in ankylosing spondylitis. J Rheumatology. (2016) 43:1695–703. doi: 10.3899/jrheum.151133

PubMed Abstract | CrossRef Full Text | Google Scholar

126. Wang C, Liao Q, Hu Y, Zhong D. T lymphocyte subset imbalances in patients contribute to ankylosing spondylitis. Chenggong Exp Therapeutic Med. (2014) 21:250–6. doi: 10.3892/etm.2014.2046

PubMed Abstract | CrossRef Full Text | Google Scholar

127. Chowdhury AC, Chaurasia S, Kumar S, Amita A, Misra R. IL-17 and IFN-γ producing NK and γδ-T cells are preferentially expanded in synovial fluid of patients with reactive arthritis and undifferentiated spondyloarthritis. Clin Immunol. (2017) 183:207–12. doi: 10.1016/j.clim.2017.03.016

PubMed Abstract | CrossRef Full Text | Google Scholar

128. Noordenbos T, Yeremenko N, Gofita I, Van de Sande M. Interleukin-17–positive mast cells contribute to synovial inflammation in spondylarthritis. Arthrt Rheum. (2011)54:99–109. doi: 10.1002/art.33396

PubMed Abstract | CrossRef Full Text | Google Scholar

129. N van Tok M, Na S, Lao CR, Alvi M, Pots D, Van de Sande MGH. The initiation, but not the persistence, of experimental spondyloarthritis is dependent on interleukin-23 signaling. Front Immunol. (2018) 9:1550. doi: 10.3389/fimmu.2018.01550

PubMed Abstract | CrossRef Full Text | Google Scholar

130. Ruiz de Morales JMG, Puig L, Daudén E, Cañete JD, Pablo JL, Olveira Martíng A. Critical role of interleukin (IL)-17 in inflammatory and immune disorders: an updated review of the evidence focusing in controversies. AutoimmRev. (2020) 19:102429. doi: 10.1016/j.autrev.2019.102429

PubMed Abstract | CrossRef Full Text | Google Scholar

131. Taurog JD, Maika s.D, Simmons WA, Breban M, Hammer RE. Susceptibility to inflammatory disease in HLA-B27 transgenic rat lines correlates with the level of B27 expression. J Immunol. (1993) 150:4168–78.

PubMed Abstract | Google Scholar

132. Taurog JD, Richardson JA, Croft JT, Simmons WA, Zhou M, Fernández-Sueiro LJ, et al. The germfree state prevents development of gut and joint inflammatory disease in HLA-B27 transgenic rats. J Exp Med. (1994) 180:2359–64. doi: 10.1084/jem.180.6.2359

PubMed Abstract | CrossRef Full Text | Google Scholar

133. Taurog JD. Animal models of spondyloarthritis. Adv Exp Med Biol. (2009) 649:245–54. doi: 10.1007/978-1-4419-0298-6_18

CrossRef Full Text | Google Scholar

134. Poddubnyy D, Sieper J. treatment of axial spondyloarthritis: what does the future hold? Curr Rheumatol Rep. (2020) 22:47. doi: 10.1007/s11926-020-00924-5

PubMed Abstract | CrossRef Full Text | Google Scholar

135. Baeten D, Baraliakos X, Braun J, Sieper J, Emery P, Van der Heijde D, et al. Anti-interleukin-17A monoclonal antibody secukinumab in treatment of ankylosing spondylitis: a randomised, double-blind, placebo-controlled trial. Lancet. (2013) 382:1705–13. doi: 10.1016/S0140-6736(13)61134-4

PubMed Abstract | CrossRef Full Text | Google Scholar

136. Baeten D, Sieper J, Braun J, Baraliakos X, Dougados M, Emery P, et al. Secukinumab, an Interleukin-17A Inhibitor, in Ankylosing Spondylitis. Engl J Med. (2015) 373:2534–48. doi: 10.1056/NEJMoa1505066

PubMed Abstract | CrossRef Full Text | Google Scholar

137. Marzo-Ortega H, Sieper J, Kivitz A, Blanco R, Cohen M, Martin R, et al. Secukinumab and sustained improvement in signs and symptoms of patients with active ankylosing spondylitis through two years: results from a phase III study. Arthritis Care Res. (2017) 69:1020–9. doi: 10.1002/acr.23233

PubMed Abstract | CrossRef Full Text | Google Scholar

138. Deodhar A, Van der Heijde D, Gensler LS, Kim TH, Maksymowych W, Østergaard M, et al. Ixekizumab for patients with non-radiographic axial spondyloarthritis (COAST-X): a randomised, placebo-controlled trial. Lancet. (2020) 395:53–64. doi: 10.1016/S0140-6736(19)32971-X

PubMed Abstract | CrossRef Full Text | Google Scholar

139. Gaydukova I, Mazurov V, Erdes S, Dubinina T, Nesmeyanova O, Ilivanova E, et al. Netakimab reduces the disease activity of radiographic axial spondyloarthritis. Results of ASTERA study. Ann Rheum Dis. (2019) 78:193–4. doi: 10.1136/Annrheumdis-2019-eular.6633

CrossRef Full Text | Google Scholar

140. Van der Heijde D, Gensler LS, Deodhar A, Baraliakos X, Poddubnyy D, Kivitz A, et al. Dual neutralisation of interleukin-17A and interleukin-17F with bimekizumab in patients with active ankylosing spondylitis: results from a 48-week phase IIb, randomised, double-blind, placebo-controlled, dose-ranging study. Ann Rheum Dis. (2020) 79:595–604. doi: 10.1136/annrheumdis-2020-216980

PubMed Abstract | CrossRef Full Text | Google Scholar

141. Wendling D, Verhoeven F, Prati C. Anti-IL-17 monoclonal antibodies for the treatment of ankylosing spondylitis. Taylor Francis Online. (2018) 19:55–64. doi: 10.1080/14712598.2019.1554053

PubMed Abstract | CrossRef Full Text | Google Scholar

142. Poddubnyy D, Hermann KGA, Callhoff J, Listing J, Sieper J. Ustekinumab for the treatment of patients with active ankylosing spondylitis: results of a 28-week, prospective, open-label, proof-of-concept study (TOPAS). Ann Rheum Dis. (2013) 73:817–23. doi: 10.1136/annrheumdis-2013-204248

PubMed Abstract | CrossRef Full Text | Google Scholar

143. Baeten D, Østergaard M, Cheng-Chung Wei J, Sieper J, Järvinen P, Tam LS, et al. Risankizumab, an IL-23 inhibitor, for ankylosing spondylitis: results of a randomised, double-blind, placebo-controlled, proof-of-concept, dose-finding phase 2 study. Ann Rheum Dis. (2018) 77:1295–302. doi: 10.1136/annrheumdis-2018-213328

PubMed Abstract | CrossRef Full Text | Google Scholar

144. Veale DJ, McGonagle D, McInnes IB, Krueger JG, Ritchlin CT, Elewaut E, et al. The rationale for Janus kinase inhibitors for the treatment of spondyloarthritis. Rheumatology. (2019) 58:197–205. doi: 10.1093/rheumatology/key070

PubMed Abstract | CrossRef Full Text | Google Scholar

145. Van der Heijde D, Deodhar A, Wei JC, Drescher E, Fleishaker E, Hendrikx T, et al. Tofacitinib in patients with ankylosing spondylitis: a phase II, 16-week, randomised, placebo-controlled, dose-ranging study. Ann Rheum Dis. (2016) 76:1340–7. doi: 10.1136/annrheumdis-2016-210322

PubMed Abstract | CrossRef Full Text | Google Scholar

146. Van der Heijde D, Baraliakos X, Lianne SG, Maksymowych W, Tseluyko V. Efficacy and safety of filgotinib, a selective Janus kinase 1 inhibitor, in patients with active ankylosing spondylitis (TORTUGA): results from a randomised, placebo-controlled, phase 2 trial Lancet. (2018) 392:2378–2387. doi: 10.1016/S0140-6736(18)32463-2

CrossRef Full Text | Google Scholar

147. van der Heijde D, Song IH, Pangan AL, Deodhar A, van den Bosch F, Maksymowych WP, et al. Efficacy and safety of upadacitinib in patients with active ankylosing spondylitis (SELECT-AXIS 1): a multicentre, randomised, double-blind, placebo-controlled, phase 2/3 trial. Lancet. (2016) 394:2108–17. doi: 10.1016/S0140-6736(19)32534-6

PubMed Abstract | CrossRef Full Text | Google Scholar

148. Pathan E, Abraham S, Van Rossen E, Withrington R, Keat A, Charles PJ, et al. Efficacy and safety of Apremilast, an oral phosphodiesterase 4 inhibitor, in ankylosing spondylitis. Ann Rheum Dis. (2016) 72:1475–80. doi: 10.1136/annrheumdis-2012-201915

PubMed Abstract | CrossRef Full Text | Google Scholar

149. Mease PJ, Genovese MC, Weinblatt M, Peloso PM, Chen K, Othman AA, et al. Phase II study of ABT-122, a tumor necrosis factor– and interleukin-17a–targeted dual variable domain immunoglobulin, in patients with psoriatic arthritis with an inadequate response to methotrexate. Arthritis Rheumatol. (2018) 70:1778–89. doi: 10.1002/art.40579

PubMed Abstract | CrossRef Full Text | Google Scholar

150. Schon MP, Boehncke WH. Psoriasis. N Engl J Med. (2005) 352:1899–912. doi: 10.1056/NEJMra041320

CrossRef Full Text | Google Scholar

151. Lowes MA, Bowcock AM, Krueger JG. Pathogenesis and therapy of psoriasis. Nature. (2007) 445:866–73. doi: 10.1038/nature05663

CrossRef Full Text | Google Scholar

152. Veale DJ, Fearon U. The pathogenesis of psoriatic arthritis. Lancet. (2018) 391:2273–84. doi: 10.1016/S0140-6736(18)30830-4

CrossRef Full Text | Google Scholar

153. Michel ML, Keller AC, Paget C, Fujio M, Trottein F, Savage PB, et al. Identification of an IL-17-producing NK1.1(neg) iNKT cell population involved in airway neutrophilia. J Exp Med. (2007) 204:995–1001. doi: 10.1084/jem.20061551

PubMed Abstract | CrossRef Full Text | Google Scholar

154. Doisne JM, Becourt C, Amniai L, Duarte N, Le Luduec JB, Eberl G, et al. Skin and peripheral lymph node invariant NKT cells are mainly retinoic acid receptor-related orphan receptor (gamma)t+ and respond preferentially under inflammatory conditions. J Immunol. (2009) 183:2142–9. doi: 10.4049/jimmunol.0901059

PubMed Abstract | CrossRef Full Text | Google Scholar

155. Brembilla NC, Stalder R, Senra L, Bohencke WH. IL-17A localizes in the exocytic compartment of mast cells in psoriatic skin. Br J Dermatol. (2017) 177:1458–60. doi: 10.1111/bjd.15358

PubMed Abstract | CrossRef Full Text | Google Scholar

156. Lin AM, Rubin CJ, Khandpur R, Wang JY, Riblett MB, Yalavarthi S, et al. Mast cells and neutrophils release IL-17 through extracellular trap formation in psoriasis. J Immunol. (2011) 187:490–500. doi: 10.4049/jimmunol.1100123

PubMed Abstract | CrossRef Full Text | Google Scholar

157. Di Cesare A, Di Meglio P, Nestle FO. The IL-23/Th17 axis in the immunopathogenesis of psoriasis. J Invest Dermatol. (2009) 129:1339–50. doi: 10.1038/jid.2009.59

PubMed Abstract | CrossRef Full Text | Google Scholar

158. Lowes MA, Suarez-Farinas M, Krueger JG. Immunology of psoriasis. Annu Rev Immunol. (2014) 32:227–55. doi: 10.1146/annurev-immunol-032713-120225

CrossRef Full Text | Google Scholar

159. Blauvelt A, Chiricozzi A. The immunologic Role of IL-17 in psoriasis and psoriatic arthritis pathogenesis. Clin Rev Allergy Immunol. (2018) 55:379–90. doi: 10.1007/s12016-018-8702-3

PubMed Abstract | CrossRef Full Text | Google Scholar

160. Lee E, Trepicchio WL, Oestreicher JL, Pittman D, Wang F, Chamian F, et al. Increased expression of interleukin 23 p19 and p40 in lesional skin of patients with psoriasis vulgaris. J Exp Med. (2004) 199:125–30. doi: 10.1084/jem.20030451

PubMed Abstract | CrossRef Full Text | Google Scholar

161. Melis L, Vandooren B, Kruithof E, Jacques P, De Vos M, Mielants H, et al. Systemic levels of IL-23 are strongly associated with disease activity in rheumatoid arthritis but not spondyloarthritis. Ann Rheum Dis. (2010) 69:618–23. doi: 10.1136/ard.2009.107649

PubMed Abstract | CrossRef Full Text | Google Scholar

162. Van Baarsen LG, Lebre MC, van der Coelen D, Aarrass S, Tang MW, Ramwadhdoebe TH, et al. Heterogeneous expression pattern of interleukin 17A (IL-17A), IL-17F and their receptors in synovium of rheumatoid arthritis, psoriatic arthritis and oste-oarthritis: possible explanation for nonresponse to anti-IL-17 therapy? Arthritis Res Ther. (2014) 16:426. doi: 10.1186/s13075-014-0426-z

PubMed Abstract | CrossRef Full Text | Google Scholar

163. Tsukazaki H, Kaito T. The Role of the IL-23/IL-17 pathway in the pathogenesis of spondyloarthritis. Int J Mol Sci. (2020) 3:6401. doi: 10.3390/ijms21176401

PubMed Abstract | CrossRef Full Text | Google Scholar

164. Boutet MA, Nerviani A, Gallo Afflitto G, Pitzalis C. Role of the IL-23/IL-17 axis in psoriasis and psoriatic arthritis: the clinical importance of its divergence in skin and joints. Int J Mol Sci. (2018) 19:530. doi: 10.3390/ijms19020530

PubMed Abstract | CrossRef Full Text | Google Scholar

165. Veale D, Yanni G, Rogers S, Barnes L, Bresnihan B, FitzGerald O. Reduced synovial membrane macrophage numbers, ELAM-1 expression, and lining layer hyperplasia in psoriatic arthritis as compared with rheumatoid arthritis. Arthr Rheum. (1993) 36:893–900. doi: 10.1002/art.1780360705

PubMed Abstract | CrossRef Full Text | Google Scholar

166. Canete JD, Santiago B, Cantaert T, Sanmartí R, Palacin A, Celis R, et al. Ectopic lymphoid neogenesis in psoriatic arthritis. Ann Rheum Dis. (2007) 66:720–6. doi: 10.1136/ard.2006.062042

PubMed Abstract | CrossRef Full Text | Google Scholar

167. Dolcino M, Lunardi C, Ottria A, Tinazzi E, Patuzzo G, Puccetti A. Crossreactive autoantibodies directed against cutaneous and joint antigens are present in psoriatic arthritis. PLoS ONE. (2014) 6:e115424. doi: 10.1371/journal.pone.0115424

PubMed Abstract | CrossRef Full Text | Google Scholar

168. Mori G, Cantatore FP, Brunetti G, Oranger A, Colaianni G, Quarta L, et al. Synovial fluid fibroblasts and lymphocytes support the osteoclastogenesis in human psoriatic arthritis. Ann N Y Acad Sci. (2007) 1117:159–64. doi: 10.1196/annals.1402.013

PubMed Abstract | CrossRef Full Text | Google Scholar

169. Gottlieb A, Korman NJ, Gordon KB, Feldman SR, Lebwohl M, Koo JY.M, et al. Guidelines of care for the management of psoriasis and psoriatic arthritis. Section 2. Psoriatic arthritis: overview and guidelines of care for treatment with an emphasis on the biologics. J Am Acad Dermatol. (2008) 58:851–64. doi: 10.1016/j.jaad.2008.02.040

PubMed Abstract | CrossRef Full Text | Google Scholar

170. Pacifici R. The role of IL-17 TH17 cells in the bone catabolic activity of PTH. Front Immunol. (2016) 7:57. doi: 10.3389/fimmu.2016.00057

PubMed Abstract | CrossRef Full Text | Google Scholar

171. Zaba LC, Fuentes-Duculan J, Eungdamrong NJ, Abello MV, Novitskaya I, Pierson KC, et al. Psoriasis is characterized by accumulation of immunostimulatory and Th1/Th17 cell-polarizing myeloid dendritic cells. J Invest Dermatol. (2009) 129:79–88. doi: 10.1038/jid.2008.194

PubMed Abstract | CrossRef Full Text | Google Scholar

172. Teunissen MB, Munneke JM, Bernink JH, Spuls PI, Res PC, Te Velde A, et al. Composition of innate lymphoid cell subsets in the human skin: enrichment of NCR(+) ILC3 in lesional skin and blood of psoriasis patients. J Invest Dermatol. (2014) 134:2351–60. doi: 10.1038/jid.2014.146

PubMed Abstract | CrossRef Full Text | Google Scholar

173. Villanova F, Flutter B, Tosi I, Grys K, Sreeneebus H, Perera GK, et al. Characterization of innate lymphoid cells in human skin and blood demonstrates increase of NKp44+ ILC3 in psoriasis. J Invest Dermatol. (2014) 134:984–91. doi: 10.1038/jid.2013.477

PubMed Abstract | CrossRef Full Text | Google Scholar

174. Cai Y, Shen X, Ding C, Qi C, Li K, Li X, et al. Pivotal role of dermal IL-17-producing gammadelta T cells in skin inflammation. Immunity. (2011) 35:596–610. doi: 10.1016/j.immuni.2011.08.001

PubMed Abstract | CrossRef Full Text | Google Scholar

175. Sherlock JP, Joyce-Shaikh B, Turner SP, Chao CC, Sathe M, Grein J, et al. IL-23 induces spondyloarthropathy by acting on ROR-gammat + CD3 + CD4-CD8- entheseal resident T cells. Nat Med. (2012) 18:1069–76. doi: 10.1038/nm.2817

PubMed Abstract | CrossRef Full Text | Google Scholar

176. Benham H, Norris P, Goodall J, Wechalekar MD, FitzGerald O, Szentpetery A, et al. Th17 and Th22 cells in psoriatic arthritis and psoriasis. Arthritis Res Ther. (2013) 15:R136. doi: 10.1186/ar4317

PubMed Abstract | CrossRef Full Text | Google Scholar

177. Fitch E, Harper E, Skorcheva I, E Kurtz S, Blauvelt A. Pathophysiology of psoriasis: recent advances on IL-23 and Th17 cytokines. Curr Rheumatol Rep. (2007) 9:461–7. doi: 10.1007/s11926-007-0075-1

PubMed Abstract | CrossRef Full Text | Google Scholar

178. Liang SC, Tan XY, Luxenberg DP, Karim R, Dunussi-Joannopoulos K, Collins M, et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells and cooperatively enhance expression of antimicrobial peptides. J Exp Med. (2006) 203:2271–9. doi: 10.1084/jem.20061308

PubMed Abstract | CrossRef Full Text | Google Scholar

179. Koga C, Kabashima K, Shiraishi N, Kobayashi M, Tokura Y. Possible pathogenic role of Th17 cells for atopic dermatitis. J Invest Dermatol. (2008) 128:2625–30. doi: 10.1038/jid.2008.111

PubMed Abstract | CrossRef Full Text | Google Scholar

180. Wolk K, Kunz S, Witte E, Friedrich M, Asadullah K, Sabat R. IL-22 increases the innate immunity of tissues. Immunity. (2004) 21:241–54. doi: 10.1016/j.immuni.2004.07.007

PubMed Abstract | CrossRef Full Text | Google Scholar

181. Wolk K, Witte E, Wallace E, Döcke WD, Kunz S, Asadullah K, et al. IL-22 regulates the expression of genes responsible for antimicrobial defense, cellular differentiation, and mobility in keratinocytes: a potential role in psoriasis. Eur J Immunol. (2006) 36:1309–23. doi: 10.1002/eji.200535503

PubMed Abstract | CrossRef Full Text | Google Scholar

182. Ong PY, Ohtake T, Brandt C, Strickland I, Boguniewicz M, Ganz T, et al. Endogenous antimicrobial peptides and skin infections in atopic dermatitis. N Engl J Med. (2002) 347:1151–60. doi: 10.1056/NEJMoa021481

PubMed Abstract | CrossRef Full Text | Google Scholar

183. Ciccia F, Guggino G, Ferrante A, Raimondo S, Bignone R, Rodolico R, et al. Interleukin-9 overexpression and th9 polarization characterize the inflamed gut, the synovial tissue, and the peripheral blood of patients with psoriatic arthritis. Arthritis Rheumatol. (2016) 68:1922–31. doi: 10.1002/art.39649

PubMed Abstract | CrossRef Full Text | Google Scholar

184. Elyaman W, Bradshaw EM, Uyttenhove C, Dardalhon V, Awasthi A, Imitola J, et al. IL-9 induces differentiation of TH17 cells and enhances function of FoxP3+ natural regulatory T cells. Proc Natl Acad Sci USA. (2009) 106:12885–90. doi: 10.1073/pnas.0812530106

PubMed Abstract | CrossRef Full Text | Google Scholar

185. Mease P, McInnes IB. Secukinumab: a new treatment option for psoriatic arthritis. Rheumatol Ther. (2016) 3:5–29. https://dx.doi.org/10.1007%2Fs40744-016-0031-5

PubMed Abstract | Google Scholar

186. Kavanaugh A, Mease PJ, Gomez-Reino JJ, Adebajo AO, Wollenhaupt J, Gladman DD, et al. Treatment of psoriatic arthritis in a phase 3 randomised,placebo-controlled trial with apremilast, an oral phosphodiesterase 4 inhibitor. Ann Rheum Dis. (2014) 73:1020–6. doi: 10.1136/annrheumdis-2013-205056

PubMed Abstract | CrossRef Full Text | Google Scholar

187. Sakkas LI, Bogdanos DP. Are psoriasis and psoriatic arthritis the same disease? The IL-23/IL-17 axis data. Autoimmun Rev. (2017) 16:10–15. doi: 10.1016/j.autrev.2016.09.015

PubMed Abstract | CrossRef Full Text | Google Scholar

188. Zaba LC, Suarez-Farinas M, Fuentes-Duculan J, Nograles KE, Guttman-Yassky E, Cardinale I, et al. Effective treatment of psoriasis with etanercept is linked to suppression of IL-17 signaling, not immediate response TNF genes. J Allergy Clin Immunol. (2009) 124:1022–10.e1-395. doi: 10.1016/j.jaci.2009.08.046

CrossRef Full Text | Google Scholar

189. Chabaud M, Garnero P, Dayer JM, Guerne PA, Fossiez F, Miossec P. Contribution of interleukin 17 to synoviummatrix destruction in rheumatoid arthritis. Cytokine. (2000) 12:1092–99. doi: 10.1006/cyto.2000.0681

PubMed Abstract | CrossRef Full Text | Google Scholar

190. Yen D, Cheung J, Scheerens H, Poulet F, McClanahan T, McKenzie B, et al. IL-23 is essential for T cell-mediated colitis and promotes inflammation via IL-17 and IL-6. J. Clin. Invest. (2006) 116:1310–6. doi: 10.1172/JCI21404

PubMed Abstract | CrossRef Full Text | Google Scholar

191. Luo J, Wu SJ, Lacy ER, Orlovsky Y, Baker A, Teplyakov A, et al. Structural basis for the dual recognition of IL-12 and IL-23 by ustekinumab. J. Mol. Biol. (2010) 402:797–812. doi: 10.1016/j.jmb.2010.07.046

PubMed Abstract | CrossRef Full Text | Google Scholar

192. Dobbin-Sears I, Roberts J, O'Rielly DD, Rahman P. Ustekinumab in psoriatic arthritis and related phenotypes. Ther Adv Chronic Dis. (2018) 9:191–8. doi: 10.1177/2040622318781760

PubMed Abstract | CrossRef Full Text | Google Scholar

193. Patel NU, Vera NC, Shealy ER, Wetzel M, Feldman SR. A review of the use of secukinumab for psoriatic arthritis. Rheumatol Ther. (2017) 4:233–46. doi: 10.1007/s40744-017-0076-0

CrossRef Full Text | Google Scholar

194. McInnes IB, Mease PJ, Ritchlin CT, Rahman P, Gottlieb AB, Kirkham B, et al. Secukinumab sustains improvement in signs and symptoms of psoriatic arthritis: 2 year results from the phase 3 FUTURE 2 study. Rheumatology. (2017) 56:1993–2003. doi: 10.1093/rheumatology/kex301

PubMed Abstract | CrossRef Full Text | Google Scholar

195. Van der Heijde D, Gladman DD, Kishimoto M, Masato Okada M, Rathmann SS, Moriarty SR, et al. Efficacy and safety of ixekizumab in patients with active psoriatic arthritis: 52-week results from a Phase III study (SPIRIT-P1). J Rheumatol. (2018) 45:1608. doi: 10.3899/jrheum.170429.C1

PubMed Abstract | CrossRef Full Text | Google Scholar

196. Nash P, Kirkham B, Okada M, Rahman P, Combe B, Burmester GR, et al. SPIRIT-P2 Study GroupIxekizumab for the treatment of patients with active psoriatic arthritis and an inadequate response to tumour necrosis factor inhibitors: results from the 24-week randomised, double-blind, placebo-controlled period of the SPIRIT-P2 phase 3 trial. Lancet. (2017) 389:2317–27. doi: 10.1136/annrheumdis-2017-eular.1576

CrossRef Full Text | Google Scholar

197. Mease PJ, Smolen JS, Behrens F, Nash P, Leage LS, Li L, et al. A head-to-head comparison of the efficacy and safety of ixekizumab and adalimumab in biological-naïve patients with active psoriatic arthritis: 24-week results of a randomised, open-label, blinded-assessor trial. Ann Rheum Dis. (2020) 79:123–31. doi: 10.1136/annrheumdis-2019-215386

PubMed Abstract | CrossRef Full Text | Google Scholar

198. Ly K, Beck KM, Smith MP, Orbai AM, Liao W. Tofacitinib in the management of active psoriatic arthritis: patient selection and perspectives. Psoriasis. (2019) 9:97–107. doi: 10.2147/PTT.S161453

PubMed Abstract | CrossRef Full Text | Google Scholar

199. Chen M, Dai SM. A novel treatment for psoriatic arthritis: Janus kinase inhibitors. Chin Med J (Engl). (2020) 133:959–67. doi: 10.1097/CM9.0000000000000711

PubMed Abstract | CrossRef Full Text | Google Scholar

200. Deodhar A, Helliwell PS, Boehncke WH, Kollmeier AP, Hsia EC, Subramanian A, et al. Guselkumab in patients with active psoriatic arthritis who were biologic-naive or had previously received TNFα inhibitor treatment (DISCOVER-1): a double-blind, randomised, placebo-controlled phase 3 trial. Lancet. (2020) 395:1115–25. doi: 10.1016/S0140-6736(20)30265-8

PubMed Abstract | CrossRef Full Text | Google Scholar

201. Mease PJ, Rahman P, Gottlieb AB, Kollmeier AP, Hsia EC, Xu XL, et al. Guselkumab in biologic-naive patients with active psoriatic arthritis (DISCOVER-2): a double-blind, randomised, placebo-controlled phase. 3 trial. Lancet. (2020) 395:1126–36. doi: 10.1016/S0140-6736(20)30263-4

PubMed Abstract | CrossRef Full Text | Google Scholar

202. Scott DL, Wolfe F, Huizinga TWJ. Rheumatoid arthritis. Lancet. (2010) 376:1094–8. doi: 10.1016/S0140-6736(10)60826-4

CrossRef Full Text | Google Scholar

203. McInnes I, Schett G. Cytokines in the pathogenesis of rheumatoid arthritis. Nat Rev Immunol. (2007) 7:429–42. doi: 10.1038/nri2094

CrossRef Full Text | Google Scholar

204. Lubberts E. The IL-23–IL-17 axis in inflammatory arthritis. Nat Rev Rheumatol. (2015) 11:415–29. doi: 10.1038/nrrheum.2015.53

CrossRef Full Text | Google Scholar

205. Van Hamburg JP, Asmawidjaja PS, Davelaar N, Mus AMC, Colin EM, Hazes JMW, et al. Th17 cells, but not Th1 cells, from patients with early rheumatoid arthritis are potent inducers of matrix metalloproteinases and proinflammatory cytokines upon synovial fibroblast interaction, including autocrine interleukin-17A production. Arthritis Rheum. (2011) 63:73–83. doi: 10.1002/art.30093

PubMed Abstract | CrossRef Full Text | Google Scholar

206. Paulissen SMJ, Van Hamburg JP, Davelaar N, Asmawidjaja PS, Hazes JMW, Lubberts E. synovial fibroblasts directly induce th17 pathogenicity via the cyclooxygenase/prostaglandin E2 pathway, independent of IL-23. J Immunology. (2013) 191:1364–72. doi: 10.4049/jimmunol.1300274

PubMed Abstract | CrossRef Full Text | Google Scholar

207. Yago T, Nanke Y, Ichikawa N, Kobashigawa T, Mogi M, Kamatani N, et al. IL-17 induces osteoclastogenesis from human monocytes alone in the absence of osteoblasts, which is potently inhibited by anti-TNF-α antibody: A novel mechanism of osteoclastogenesis by IL-17. J Cell Biochem. (2009) 108:947–55. doi: 10.1002/jcb.22326

PubMed Abstract | CrossRef Full Text | Google Scholar

208. Paulissen SM, Van Hamburg JP, Dankers W, Lubberts E. The role and modulation of CCR6+ Th17 cell populations in rheumatoid arthritis. Citokyne. (2015) 74:43–53. doi: 10.1016/j.cyto.2015.02.002

PubMed Abstract | CrossRef Full Text | Google Scholar

209. Penatti A, Facciotti F, De Matteis R, Larghi P, Paroni M, Murgo A, et al. Differences in serum and synovial CD4+ T cells and cytokine profiles to stratify patients with inflammatory osteoarthritis and rheumatoid arthritis. Arthritis Res Ther. (2017) 9:103. doi: 10.1186/s13075-017-1305-1

PubMed Abstract | CrossRef Full Text | Google Scholar

210. Paulissen SMJ, van Hamburg JP, Davelaar N, Vroman H, Hazes JMW, de Jong PHP, et al. CCR6+ Th cell populations distinguish ACPA positive from ACPA negative rheumatoid arthritis. Arthritis Res Ther. (2015) 17:344. doi: 10.1186/s13075-015-0800-5

PubMed Abstract | CrossRef Full Text | Google Scholar

211. Zizzo G, De Santis M, Bosello SL, Fedele AL, Peluso G, Gremese E, et al. Synovial fluid-derived T helper 17 cells correlate with inflammatory activity in arthritis, irrespectively of diagnosis. Clin Immunol. (2011) 138:107–16. doi: 10.1016/j.clim.2010.10.002

PubMed Abstract | CrossRef Full Text | Google Scholar

212. Murphy CA, Langrish CL, Chen Y, Blumenschein W, McClanahan T, Kastelein RA, et al. Divergent pro- and antiinflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation. J Exp Med. (2003) 198:1951–7. doi: 10.1084/jem.20030896

PubMed Abstract | CrossRef Full Text | Google Scholar

213. Genovese MC, Van den Bosch F, Roberson SA, Bojin S, Biagini IM, Ryan P, et al. LY2439821, a humanized anti–interleukin-17 monoclonal antibody, in the treatment of patients with rheumatoid arthritis: A phase I randomized, double-blind, placebo-controlled, proof-of-concept study. Arthritis Rheum. (2010) 62:929–39. doi: 10.1002/art.27334

PubMed Abstract | CrossRef Full Text | Google Scholar

214. Genovese MC, Durez P, Richards HB, Supronik J, Dokoupilova E, Mazurov V, et al. Efficacy and safety of secukinumab in patients with rheumatoid arthritis: a phase II, dose-finding, double-blind, randomised, placebo controlled study. Ann Rheum Dis. (2013) 72:863–9. doi: 10.1136/annrheumdis-2012-201601

PubMed Abstract | CrossRef Full Text | Google Scholar

215. Genovese MC, Durez P, Richards HB, Supronik J, Dokoupilova E, Aelion JA, et al. One-year efficacy and safety results of secukinumab in patients with rheumatoid arthritis: phase ii, dose-finding, double-blind, randomized, placebo-controlled study. J Rheumatol. (2014) 41:414421. doi: 10.3899/jrheum.130637

PubMed Abstract | CrossRef Full Text | Google Scholar

216. Martin DA, Churchill M, Flores-Suarez LF, Cardiel MH, Wallace D, Martin R, et al. A phase Ib multiple ascending dose study evaluating safety, pharmacokinetics, and early clinical response of brodalumab, a human anti-IL-17R antibody, in methotrexate-resistant rheumatoid arthritis. Arthritis Res Ther. (2013) 15:R164. doi: 10.1186/ar4347

PubMed Abstract | CrossRef Full Text | Google Scholar

217. Genovese MC, Greenwald M, Cho C-S, Berman A, Jin L, Cameron GS, et al. A Phase II randomized study of subcutaneous ixekizumab, an anti–interleukin-17 monoclonal antibody, in rheumatoid arthritis patients who were naive to biologic agents or had an inadequate response to tumor necrosis factor inhibitors. Arthritis Rheumatol. (2014) 66:1693–704. doi: 10.1002/art.38617

PubMed Abstract | CrossRef Full Text | Google Scholar

218. Smolen JS, Agarwal SK, Ilivanova E, Xu XL, Miao Y, Zhuang Y, et al. A randomised phase II study evaluating the efficacy and safety of subcutaneously administered ustekinumab and guselkumab in patients with active rheumatoid arthritis despite treatment with methotrexate. Ann Rheum Dis. (2017) 76:83139. doi: 10.1136/annrheumdis-2016-209831

PubMed Abstract | CrossRef Full Text | Google Scholar

219. Fleischmann RM, Wagner F, Kivitz AJ, Mansikka HT, Khan N, Othman AA, et al. Safety, tolerability, and pharmacodynamics of ABT-122, a tumor necrosis factor– and interleukin-17–targeted dual variable domain immunoglobulin, in patients with rheumatoid arthritis. Arthritis Rheumatol. (2017) 69:2283–91. doi: 10.1002/art.40319

PubMed Abstract | CrossRef Full Text | Google Scholar

220. Genovese MC, Weinblatt ME, Aelion JA, Mansikka HT, Peloso PM, Chen K, et al. ABT-122, a bispecific dual variable domain immunoglobulin targeting tumor necrosis factor and interleukin-17a, in patients with rheumatoid arthritis with an inadequate response to methotrexate. Arthritis Rheumatol. (2018) 70:1710–20. doi: 10.1002/art.40580

PubMed Abstract | CrossRef Full Text | Google Scholar

221. Glatt S, Taylor PC, McInnes IB, Schett G, Landewé R, Baeten D, et al. Efficacy and safety of bimekizumab as add-on therapy for rheumatoid arthritis in patients with inadequate response to certolizumab pegol: a proof-of-concept study. Ann Rheum Dis. (2019) 78:1033–40. doi: 10.1136/annrheumdis-2018-214943

PubMed Abstract | CrossRef Full Text | Google Scholar

222. Voulgarelis M, Tzioufas A. Pathogenetic mechanisms in the initiation and perpetuation of Sjögren's syndrome. Nat Rev Rheumatol. (2010) 6:529–37. doi: 10.1038/nrrheum.2010.118

PubMed Abstract | CrossRef Full Text | Google Scholar

223. Salomonsson S, Jonsson MV, Skarstein K, Brokstad KA, Hjelmström P, Wahren-Herlenius M, et al. Cellular basis of ectopic germinal center formation and autoantibody production in the target organ of patients with Sjögren's syndrome. Arthritis Rheum. (2003) 48:3187–201. doi: 10.1002/art.11311

PubMed Abstract | CrossRef Full Text | Google Scholar

224. Fox R. Sjögren's syndrome. The Lancet. (2005) 366:321–31. doi: 10.1016/S0140-6736(05)66990-5

CrossRef Full Text | Google Scholar

225. Yang XO, Pappu BP, Nurieva R, Akimzhanov A, Kang HS, Chung Y, et al. T helper 17 lineage differentiation is programmed by orphan nuclear receptors RORa and RORg. Immunity. (2007) 28:29–39. doi: 10.1016/j.immuni.2007.11.016

CrossRef Full Text | Google Scholar

226. Nguyen CQ, Hu MH, Li Y, Stewart C, Peck AB. Salivary gland tissue expression of interleukin-23 and interleukin-17 in Sjögren's syndrome: findings in humans and mice. Arthritis Rheum. (2008) 58:734–43. doi: 10.1002/art.23214

PubMed Abstract | CrossRef Full Text | Google Scholar

227. Katsifis GE, Rekka S, Moutsopoulos NM, Pillemer S, Whal SM. Systemic and local interleukin-17 and linked cytokines associated with sjögren's syndrome immunopathogenesis. Am J Pathol. (2009) 175:1167–77. doi: 10.2353/ajpath.2009.090319

PubMed Abstract | CrossRef Full Text | Google Scholar

228. Ciccia F, Guggino G, Rizzo A, Alessandro R, Carubbi F, Giardina AR, et al. Rituximab modulates IL-17 expression in the salivary glands of patients with primary Sjögren's syndrome. Rheumatology. (2014) 53:1313–20. doi: 10.1093/rheumatology/keu004

PubMed Abstract | CrossRef Full Text | Google Scholar

229. Pérez P, Kwon YJ, Alliende C, Leyton L, Aguilera S, Molina C, et al. Increased acinar damage of salivary glands of patients with Sjögren's syndrome is paralleled by simultaneous imbalance of matrix metalloproteinase 3/tissue inhibitor of metalloproteinases 1 and matrix metalloproteinase 9/tissue inhibitor of metalloproteinases 1 ratios. Arthritis Rheum. (2005) 52:2751–60. doi: 10.1002/art.21265

PubMed Abstract | CrossRef Full Text | Google Scholar

230. Mitsdoerffer M, Lee Y, Jäger A, Kim H-J, Korn T, Kolls JK, et al. Proinflammatory T helper type 17 cells are effective B-cell helpers. Proc Natl Acad Sci USA. (2010) 107:14292–7. doi: 10.1073/pnas.1009234107

PubMed Abstract | CrossRef Full Text | Google Scholar

231. Liu R, Gao C, Chen H, Li Y, Jin Y, Qi H. Analysis of Th17-associated cytokines and clinical correlations in patients with dry eye disease. PLoS ONE. (2017) 12:e0173301. doi: 10.1371/journal.pone.0173301

PubMed Abstract | CrossRef Full Text | Google Scholar

232. Gan Y, Zhao X, He J, Liu X, Li Y, Sun X, et al. Increased interleukin-17F is associated with elevated autoantibody levels and more clinically relevant than Interleukin-17A in primary sjögren's syndrome. J Immunol Res. (2017) 2017:4768408. doi: 10.1155/2017/4768408

PubMed Abstract | CrossRef Full Text | Google Scholar

233. Voigt A, Esfandiary L, Wanchoo A, Glenton P, Donate A, Craft WF, et al. Sexual dimorphic function of IL-17 in salivary gland dysfunction of the C57BL/6.NOD-Aec1Aec2 model of Sjögren's syndrome. Sci Rep. (2016) 6:38717. doi: 10.1038/srep38717

PubMed Abstract | CrossRef Full Text | Google Scholar

234. Lin X, Rui K, Deng J, Wang X, Wang S, Ko KH, et al. Th17 cells play a critical role in the development of experimental Sjögren's syndrome. Ann Rheum Dis. (2015) 74:1302–10. doi: 10.1136/annrheumdis-2013-204584

PubMed Abstract | CrossRef Full Text | Google Scholar

235. Alunno A, Bistoni O, Bartoloni E, Caterbi S, Bigerna B, Tabarrini A, et al. IL-17-producing CD4–CD8– T cells are expanded in the peripheral blood, infiltrate salivary glands and are resistant to corticosteroids in patients with primary Sjögren's syndrome. Ann Rheum Dis. (2013) 72:286–92. doi: 10.1136/annrheumdis-2012-201511

PubMed Abstract | CrossRef Full Text | Google Scholar

236. Laurence A, Tato CM, Davidson TS, Kanno Y, Chen Z, Yao Z, et al. Interleukin-2 signaling via STAT5 constrains T helper 17 cell generation. Immunity. (2007) 26:371–81. doi: 10.1016/j.immuni.2007.02.009

PubMed Abstract | CrossRef Full Text | Google Scholar

237. Luo J, Ming B, Zhang C, Deng X, Li P, Wei Z, et al. IL-2 inhibition of Th17 generation rather than induction of treg cells is impaired in primary sjögren's syndrome patients. Front Immunol. (2018) 9:1755. doi: 10.3389/fimmu.2018.01755

PubMed Abstract | CrossRef Full Text | Google Scholar

238. Miao M, Hao Z, Guo Y, Zhang X, Zhang S, Luo J, et al. Short-term and low-dose IL-2 therapy restores the Th17/Treg balance in the peripheral blood of patients with primary Sjögren's syndrome. Ann Rheum Dis. (2018) 77:1838–1840. doi: 10.1136/annrheumdis-2018-213036

PubMed Abstract | CrossRef Full Text | Google Scholar

239. Felten R, Meyer N, Duffaut P, Saadoun D, Hachulla E, Hatron P, et al. Interleukin 6 receptor inhibition in primary Sjögren syndrome: a multicentre double-blind randomised placebo-controlled trial. Ann Rheum Dis. (2019) 2020:218467. doi: 10.1136/annrheumdis-2021-219882

PubMed Abstract | CrossRef Full Text | Google Scholar

240. 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. (2010) 6:123–41. doi: 10.2147/tcrm.s5599

PubMed Abstract | CrossRef Full Text | Google Scholar

241. Grosskreutz CL, Hockey H-U, Serra D, Dryja TP. dry eye signs and symptoms persist during systemic neutralization of IL-1β by canakinumab or IL-17A by secukinumab, Cornea. (2015) 34:1551–6. doi: 10.1097/ICO.0000000000000627

PubMed Abstract | CrossRef Full Text | Google Scholar

242. Heinlen LD, McClain M.T, Merrill J, Akbarali YW, Edgerton CC, Harley JB, et al. Clinical criteria for systemic lupus erythematosus precede diagnosis, and associated autoantibodies are present before clinical symptoms. Arthritis Rheum. (2007) 56:2344–51. doi: 10.1002/art.22665

PubMed Abstract | CrossRef Full Text | Google Scholar

243. Vincent FB, Northcott M, Hoi A, Mackay F, Eric F, Morand EF. Clinical associations of serum interleukin-17 in systemic lupus erythematosus. Arthritis Res Ther. (2013) 15:R97. doi: 10.1186/ar4277

PubMed Abstract | CrossRef Full Text | Google Scholar

244. Wong CK, Lit LCW, Tam LS, Li EKM, Wong PTYW, Lam CWK. Hyperproduction of IL-23 and IL-17 in patients with systemic lupus erythematosus: implications for Th17-mediated inflammation in auto-immunity. Clin Immunol. (2008) 127:385–93. doi: 10.1016/j.clim.2008.01.019

PubMed Abstract | CrossRef Full Text | Google Scholar

245. Crispín JC, Oukka M, Bayliss G, Cohen RA, A Van Beek C, Stillman IE, Kyttaris VC, Juang YT, et al. Expanded double negative T cells in patients with systemic lupus erythematosus produce IL-17 and infiltrate the kidneys. J Immunol. (2008) 181:8761–6. doi: 10.4049/jimmunol.181.12.8761

PubMed Abstract | CrossRef Full Text | Google Scholar

246. Shivakumar JS, Tsokos GC, Datta SK. T cell receptor alpha/beta expressing double-negative (CD4-/CD8-) and CD4+ T helper cells in humans augment the production of pathogenic anti-DNA autoantibodies associated with lupus nephritis. Immunol. (1989) 143:103–12.

PubMed Abstract | Google Scholar

247. Chen DY, Chen YM, Wen MC, Hsieh TY, Hung WT, Lan JL. The potential role of Th17 cells and Th17-related cytokines in the pathogenesis of lupus nephritis Lupus. Lupus. (2012) 21:1385–96. doi: 10.1177/0961203312457718

PubMed Abstract | CrossRef Full Text | Google Scholar

248. Oke V, Brauner S, Larsson A, Gustafsson J, Zickert A, Gunnarsson I, Svenungsson E. IFN-λ1 with Th17 axis cytokines and IFN-α define different subsets in systemic lupus erythematosus (SLE). Arthritis Res Ther. (2017) 19:139. doi: 10.1186/s13075-017-1344-7

PubMed Abstract | CrossRef Full Text | Google Scholar

249. Fischer K, Przepiera-Bedzak H, Sawicki M, Walecka A, Brzosko I, Brzosko M. Serum interleukin-23 in polish patients with systemic lupus erythematosus: association with lupus nephritis, obesity, and peripheral vascular disease. Mediators Inflamm. (2017) 2017:9401432. doi: 10.1155/2017/9401432

PubMed Abstract | CrossRef Full Text | Google Scholar

250. Yang J, Chu Y, Yang X, Gao D, Zhu L, Yang X, et al. Th17 and natural Treg cell population dynamics in systemic lupus erythematosus. Arthritis Rheum. (2009) 60:1472–83. doi: 10.1002/art.24499

PubMed Abstract | CrossRef Full Text | Google Scholar

251. Yang J, Yang X, Yang J, Li M. Hydroxychloroquine inhibits the differentiation of th17 cells in systemic lupus erythematosus. J Rheumatol. (2018) 45:818–26. doi: 10.3899/jrheum.170737

PubMed Abstract | CrossRef Full Text | Google Scholar

252. van Vollenhoven RF, Hahn BH, Tsokos GC, Wagner CL, Lipsky P, Touma Z, et al. Efficacy and safety of ustekinumab, an IL-12 and IL-23 inhibitor, in patients with active systemic lupus erythematosus: results of a multicentre, double-blind, phase 2, randomised, controlled study. Lancet. (2018) 392:1330–9. doi: 10.1016/S0140-6736(18)32167-6

PubMed Abstract | CrossRef Full Text | Google Scholar

Keywords: IL-23, IL-17, IL-23/IL-17 axis, inflammatory diseases, autoimmune diseases

Citation: Schinocca C, Rizzo C, Fasano S, Grasso G, La Barbera L, Ciccia F and Guggino G (2021) Role of the IL-23/IL-17 Pathway in Rheumatic Diseases: An Overview. Front. Immunol. 12:637829. doi: 10.3389/fimmu.2021.637829

Received: 04 December 2020; Accepted: 25 January 2021;
Published: 22 February 2021.

Edited by:

Matteo Vecellio, University of Oxford, United Kingdom

Reviewed by:

Saikat Majumder, University of Pittsburgh, United States
Jonilson Berlink Lima, Federal University of Western Bahia, Brazil

Copyright © 2021 Schinocca, Rizzo, Fasano, Grasso, La Barbera, Ciccia and Guggino. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Giuliana Guggino, giuliana.guggino@unipa.it

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.