ArticlePDF AvailableLiterature Review

The role of interleukin-17 in the pathogenesis of rheumatoid arthritis

Authors:

Abstract and Figures

Interleukin (IL)-17 (also known as IL-17A), the signature cytokine of the newly described T helper 17 (Th17) cell population, has been implicated in the pathogenesis of numerous autoimmune diseases including rheumatoid arthritis. IL-17 is the founding member of a new subclass of cytokines that have highly proinflammatory properties. Studies in rodents and mammalian cell culture systems, as well as clinical settings, support a role for IL-17 in promoting rheumatoid arthritis. This article discusses the history of the discovery of Th17 cells, the potential mechanisms of action of IL-17 in autoimmunity, and perspectives for IL-17-targeted cytokine therapy.
Content may be subject to copyright.
Role of IL-17 in the Pathogenesis of Rheumatoid Arthritis
Sarah L. Gaffen, Ph.D.1
1 Department of Medicine, Division of Rheumatology and Clinical Immunology, University of
Pittsburgh, Pittsburgh PA 15261, USA, sig65@pitt.edu. Ph. 412-383-8903. Fax. 412-383-8864
Abstract
IL-17 (also known as IL-17A) is the signature cytokine of the newly-described “Th17” T helper cell
population, and has been implicated in the pathogenesis of numerous autoimmune diseases including
rheumatoid arthritis. IL-17 is the founding member of a new subclass of cytokines that have highly
pro-inflammatory properties. Studies in rodents, mammalian cell culture systems as well as clinical
settings support a role for IL-17 in promoting rheumatoid arthritis. The history of the discovery of
Th17 cells, the potential mechanisms of action of IL-17 in autoimmunity and perspectives for IL-17-
targeted cytokine therapy are discussed.
Introduction
T helper cells and the new Th17 paradigm
In the last several years, there has been a paradigm-shift in our understanding of the role of T
cells in autoimmunity, particularly with regards to the CD4+ T helper cell subset (Th cells). T
helper cells have long been known as the “masterminds” of the immune system, controlling
the activities of other lymphocytes as well as many aspects of the innate immune response.
Their depletion in the setting of HIV infection highlights the vital role of CD4+ T cells in
coordinating immunity to infectious diseases. T helper cells accomplish their vital tasks
primarily by means of secreting cytokines, protein-based hormones that bind to specific
receptors on target cells. Cytokines have pleiotropic activities, which include triggering
proliferation, cell death, specific gene expression, cellular migration, etc.
In 1986, a seminal model put forth by Coffman and Mossman postulated that different
subpopulations of Th cells could shape immune responses by virtue of differential cytokine
production, and two subsets were initially identified that were termed “Th1 and Th2” (Table
1) (1). This model posits that Th1 cells mediate “cellular immunity,” characterized by
macrophage activation and opsonising antibodies through the actions of the cytokine
interferon-γ (IFNγ). Conversely, Th2 cells mediate “humoral immunity” characterized by
activation of B cells and effector antibodies, which is mediated by interleukin (IL)-4, IL-5 and
IL-13 (Figure 1). Central to this model was the concept that naïve Th cells are not pre-
determined to be either Th1 or Th2, but rather that the environment in which they encounter
antigen dictates their subsequent fate. In accordance with this idea, it was shown that IL-12,
usually derived from antigen-presenting cells (APCs) such as dendritic cells, directs
differentiation of naïve CD4+ T cells into the Th1 lineage, whereas IL-4 promotes development
to the Th2 lineage. Moreover, differentiation of each Th subset is mutually antagonistic and
self-reinforcing by cross-antagonistic signaling pathways mediated by IL-12 and IL-4. For
nearly twenty years the Th1/Th2 model provided a valuable framework in which diseases, both
infectious and autoimmune, were evaluated. Much research effort was devoted to defining the
mechanisms that control Th1/Th2 development, as well as the key cytokines, transcription
factors and microbial/autoimmune stimuli involved in this process (reviewed in (2)).
NIH Public Access
Author Manuscript
Curr Rheumatol Rep. Author manuscript; available in PMC 2010 April 1.
Published in final edited form as:
Curr Rheumatol Rep. 2009 October ; 11(5): 365–370.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
However, over time it became apparent that there were glaring discrepancies with this model,
particularly related to Th1 generation and function (2). The generation of various gene deficient
(“knockout”) mouse strains revealed puzzling differences between the Th1 effector cytokine
IFNγ and the Th1 differentiation cytokine IL-12 (Figure 1). IL-12 is a heterodimeric cytokine
composed of two subunits, IL-12p40 and IL-12p35. IL-12p40-deficient mice were developed
to probe the role of this cytokine in vivo, and IL-12p40-/-mice were found to be resistant to
most autoimmune disease models including the rodent model of RA, collagen induced arthritis
(CIA) (3). However, IFNγ-deficient mice and also IFNγ-receptor deficient mice appeared to
be resistant to CIA as well as many other autoimmune disease models; in fact, in some instances
these mice showed enhanced sensitivity to disease (3). The basis for this paradox began to
become clear when IL-12p35-deficient mice were generated, which appeared to have a highly
similar phenotype to the IFNγ-deficient mice and a less severe phenotype than IL-12p40-/-
animals. Thus, it was apparent that IL-12p40 plays an IL-12-independent role in immunity.
Indeed, IL-12p40 is also component of a new cytokine, IL-23, where it partners with a unique
IL-23p19 subunit (Figure 1). Accordingly, IL-12p40-deficient mice are deficient not only in
IL-12, but also in IL-23. Direct comparisons of IL-23p19-deficient mice and IL-12p35-
deficient began to tease apart the biological functions of these two cytokines, and it became
clear that many of the autoimmune functions that had previously been ascribed to IL-12 (e.g.
in CIA, EAE, IBD), were in fact due to the activities of IL-23 (reviewed in (4)).
Based on these findings, the function of IL-23 became a critical question. In other words, if
Th1 cells were not the main effectors of autoimmunity, was there a new, IL-23-dependent T
cell subset that had previously been overlooked? Indeed, IL-23 was shown to stimulate
production of IL-17 (often referred to as IL-17A) in murine CD4+ T cells (5). IL-17A, although
primarily derived from T cells, was not obviously a Th1 or Th2 cytokine, hinting at the
possibility of anew CD4+ T cell lineage involving this cytokine (6). Interestingly, studies in
Lyme disease arthritis models demonstrated that a distinct subset of CD4+ T cells produced
IL-17A (7), although the connection to IL-23 was not made at that time. Putting this together,
two landmark reports showed that an IL-17A-producing CD4+ T cells subset exists that can
be induced to differentiate in vitro (8,9). Moreover, induction of this lineage was found to be
independent on the classic Th1/Th2 STAT factors STAT4 and STAT6, and development of
this so-called “Th17” population could be inhibited by Th1- and Th2-specific cytokines,
IFNγ and IL-4 (Table 1). A veritable avalanche of reports followed over the next few years,
demonstrating that Th17 cells are indeed a separate lineage (reviewed in (4)). Unexpectedly
and in contrast to IL-12, IL-23 was not essential for differentiation of Th17 cells per se, but
rather for their pathogenicity in vivo (10). However, in mice IL-23 is nonetheless essential for
stable development of Th17 cells, and polymorphisms in the IL-23R gene in humans are linked
to susceptibility to Crohn's Disease (11).
Whereas IL-23 is not required for Th17 differentiation, a combination of TGFβ, IL-1, and IL-6
serve in this capacity, and the transcription factor RORγt was found to be key for differentiation
of these cells (reviewed in (4)). Strikingly, Th17 cells arise in opposition to inducible T
regulatory cells (iTregs), in part by virtue of the fact that both lineages depend on TGFβ for
their differentiation (12,13). Thus, in settings of autoimmunity, an altered balance between
immunosuppressive Tregs and inflammatory Th17 cells appears to be a major component in
disease pathogenesis. Related to this issue, an important and often unappreciated feature of Th
cells in vivo are their lineage plasticity. Specifically, differentiation to specific Th1/2/17/Treg
phenotypes is not fixed, but rather there is considerable interconversion among these cell types
observed in vivo and in vitro (14,15). In particular, Treg and Th17 cells have been shown to
interconvert (16), and understanding precisely how and under what circumstances this occurs
has important implications in terms of treating autoimmune disease.
Gaffen Page 2
Curr Rheumatol Rep. Author manuscript; available in PMC 2010 April 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Although IL-17A is predominantly produced by T cells, it is not only the province of the CD4
+ subset. CD8+ T cells also produce considerable amounts of IL-17A, as do γδ-T cells, NKT
cells and lymphoid tissue inducer (LTi) cells (reviewed in (17)).
IL-17 receptor signaling bridges innate and adaptive immunity
Although IL-17A is eponymous with the Th17 lineage, these cells also produce IL-17F, IL-21,
IL-22, IL-26 (the gene for which is found only in humans), and in some reports TNFα, CCL20,
and G-CSF (Table 1) (17). Although these are very different cytokines, they all function in a
similar manner to promote inflammation, and in some cases can act cooperatively or
synergistically (discussed in more detail below). IL-17A and IL-17F are quite homologous
(55%) and can form heterodimers (termed IL-17A/F) that appear to be the major isoform of
the cytokine in human PBMCs (18). All three cytokine forms bind to a common receptor
complex (see below), but exhibit distinct signaling potencies, with IL-17A>IL-17A/F>IL-17F.
IL-17A and IL-17F bind to a multimeric receptor complex composed of at least two subunits,
IL-17RA and IL-17RC. These receptors belong to a specific subclass of cytokine receptors
with distinct molecular features that distinguish this family from other types of receptors
(19). IL-17A and IL-17F signaling through their receptors is unusual compared to typical
adaptive T helper cell cytokines. Rather than activating JAK-STAT pathways, the IL-17-family
cytokines activate pro-inflammatory pathways more typical of innate, pro-inflammatory
cytokines such as IL-1 or TLR agonists. All these cytokines activate NF-κB, and many also
have been shown to induce MAPK signaling and the C/EBP transcription factors. Thus, the
net effect of IL-17A signaling is to induce an innate-type inflammatory effector gene
expression program that mediates potent inflammation and plays a key role in host defense
(reviewed in (20)). Conversely, in conditions of dysregulation, this inflammatory profile can
also promote inflammatory pathology in autoimmunity.
Much of our understanding of IL-17A biology in both infection and autoimmunity comes from
microarray studies of downstream target genes. Receptors for IL-17 are expressed ubiquitously,
but the key responsive cells tend to be non-immune cells such as epithelial cells, mesenchymal
cells (myoblasts, fibroblasts and adipocytes, osteoblasts) and keratinocytes. IL-17 stimulates
expression of inflammatory genes, including cytokines (IL-6, G-CSF, OSM, IL-32),
chemokines (CXCL1, CXCL2, CXCL5, CCL20) and other inflammatory effectors (iNOS,
anti-microbial genes, acute phase response genes) (reviewed in (20)). Accordingly, although
IL-17A is made by T cells, its downstream signals are similar to those induced by typical
“innate” immune receptors such as Toll-like recepor ligands (e.g. LPS) or IL-1β.
As indicated above, IL-17A and IL-17F synergize potently with TNFα, although the
mechanisms underlying this synergy are only partially understood. Thus, IL-17A may act as
a “rheostat” for TNFα signaling. A recent study in human synovial fibroblasts (21) compared
genes induced by IL-17A versus IL-17F, and found that they promoted similar profiles of gene
expression, although IL-17A was quantitatively more active. In the presence of TNFα, the
patterns of induced genes were highly similar. Interestingly, IL-17A and IL-17F upregulate
expression of TNFRII in synoviocytes (21), which may help explain the basis for synergy
between these cytokines. IL-17A also cooperates with IL-1β and IL-22, although the synergy
is generally not as dramatic as that seen with TNFα (22). Therefore, Th17-related cytokines
act cooperatively to amplify inflammatory cascades, which is beneficial in host defense but
deleterious in many autoimmune settings.
IL-17 promotes disease in mouse models of RA
The pathogenic role of IL-17A in murine models of inflammatory arthritis is unequivocal
(23). Elegant work from Erik Lubberts and colleagues has demonstrated a key role for IL-17
Gaffen Page 3
Curr Rheumatol Rep. Author manuscript; available in PMC 2010 April 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
in collage-induced arthritis. This occurs in part due to an altered RANKL:OPG ratio, and is
independent of IL-1 and TNFα (reviewed in (24)). IL-17-deficient mice are resistant to CIA
(25) as well as spontaneous arthritis in IL-1Ra-deficient mice (26). Blocking IL-17 or its
receptor with antibodies reduces disease in mice (reviewed in (24)). Although the mechanisms
are not fully elucidated, IL-17 has both direct and indirect bone destructive properties. Th17
cells express much higher levels of RANKL than Th1 or Th2 cells and thus may be very
efficient at promoting bone turnover (27). IL-17-induced proteins include matrix
metalloproteinases, RANKL and pro-inflammatory effectors such as iNOS that can
cumulatively promote bone loss (23). IL-17 signals directly in osteoblasts and synovial
fibroblasts to promote inflammation (21,28). Accordingly, IL-17 and other Th17 related
cytokines are considered prime targets of anti-cytokine therapy in autoimmune arthritis (24).
The roles of IL-17 and IL-23 on other bone-destructive conditions, however, are complex, and
not necessarily pathogenic. For example, the most common form of bone loss in humans is
actually due to infectious diseases in the oral cavity that lead to periodontal disease and
destruction of the alveolar bone crest of the jaw. Although elevated levels of IL-17 are found
in some instances of human periodontal disease (29), in a mouse model IL-17 receptor signaling
is highly bone protective through its ability to promote neutrophil expansion and recruitment
and hence limit infection (30). IL-23-deficient mice also show abnormalities in maintaining
normal bone mass, suggestive of bone-protective properties unrelated to inflammation (31).
The extent to which these findings in mice will apply to humans is still unknown, but these
observations may raise flags concerning the efficacy of blocking IL-17A or its receptor to treat
bone-destructive diseases.
IL-17 in human RA
There is strong evidence for a role for IL-17 in promoting human RA, although not all studies
agree fully. As much as a decade ago, reports began to indicate that high levels of IL-17 and
its receptor are found in RA synovial fluid and tissue explants, and IL-17 can promote joint
degradation in ex vivo models (32-37). In contrast, there is no convincing data supporting a
role for IL-17 in OA. Moreover, IL-17 together with TNFα was found to be predictive for poor
outcome in RA (38). IL-17 promotes recruitment of both neutrophils and monocytes by means
of inducing various chemokines (37), which can in turn mediate inflammation in RA. However,
studies have been reported suggesting that Th1 cells may be more important than Th17 cells
(39). The impending use of therapeutics that target IL-17A directly will no doubt shed
important light on this issue.
Implications for anti-cytokine therapy
Blocking TNFα has become a mainstay for treating RA in humans, but is not successful in all
situations (reviewed in (40)). TNFα is produced by Th cells but also by macrophages, DCs and
other innate immune cells. However, it is also plausible that anti-TNF therapy directly impacts
generation of Th17 cells and hence production of IL-17. Consistent with this hypothesis, a
recent report examined the consequences of TNF blockade in psoriasis (41). This study of 20
patients reported that 50 mg of etanercept treatment led to improved PASI scores and also to
reduced levels of IL-17A and IL-22 in psoriatic lesions. Antibodies that block IL-12p40
(ustekinumab), which hence block both IL-12 and IL-23, were developed based on the premise
that Th1 cells promote autoimmune disease. Fortuitously, this subunit is shared with IL-23,
and thus this drug presumably limits IL-17A production by blocking Th17 cells as well as Th1
cells. Ustekinumab is quite effective in Crohn's disease and psoriasitic arthritis, although not
for multiple sclerosis (42). Similarly, antibodies to the IL-6 receptor (tocilizumab) have shown
success for treating RA (43), and IL-6 is both upstream of Th17 cells and a major gene target
of IL-17. In contrast, IL-1 appears to be a key driver of Th17 development in human systems,
yet Anakinra (a soluble IL-1R antagonist) failed to be an effective therapy for RA even though
Gaffen Page 4
Curr Rheumatol Rep. Author manuscript; available in PMC 2010 April 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
it is highly successful in diseases involving inflammasome defects (44). Although this could
be due to its short half-life in vivo, a connection to Th17 cells cannot be ruled out. Antibodies
to IL-17A or IL-17RA are presently in Phase I or Phase II clinical trials for a variety of
autoimmune diseases, including psoriasis, rheumatoid arthritis, psoriatic arthritis, ankylosing
spondylitis and Crohn's Disease (www.clinicaltrials.gov). Early reports of a small psoriasis
study indicate that blocking IL-17A is very efficacious, but clearly larger studies will be needed
to determine its utility in the clinic (D. Patel, unpublished data). It is likely that, even if blocking
IL-17A or its receptor proves efficacious clinically in many patients, it will not be effective in
all who take it.
What are the likely safety issues regarding therapeutic blockade of IL-17A? Blocking TNFα
therapeutically has proved surprisingly safe, although it can promote development of
tuberculosis in patients with latent infections (44,45). Similarly, few major adverse events were
reported with the anti-IL-6R Abs (43). Studies using IL-17A-/- or IL-17RA-/- mice have shown
an important role for this cytokine system in preventing infections to a long list of microbial
pathogens, particularly those that act extracellularly such as gram negative bacteria and the
commensal yeast Candida albicans (46,47). An “experiment of nature” has provided valuable
insight into the possible side effects of blocking Th17 cells in humans. Specifically, humans
with hyper-IgE syndrome (HIES, Job's Syndrome) have recently been shown to have an
inherited, dominant-negative defect in the transcription factor STAT3. STAT3 is required for
development of Th17 cells due to its role in transducing essential signals from IL-23 and IL-6.
Accordingly, HIES patients have a paucity of Th17 cells and IL-17A, but they have normal
levels of Th1 cells and IFNγ (reviewed in (48)). These patients experience a number of recurrent
infectious diseases, including pulmonary infections, staphylococcal abscesses, eczema, and
mucocutaneous Candida albicans infections. There are also bone and connective tissue
disorders related to other functions of STAT3 (reviewed in (49)). Thus, blocking IL-17A may
prove problematic in terms of enhancing susceptibility to common infections.
Conclusions and perspectives
The past few years have been a watershed in our understanding of the T cell-mediated
pathogenesis of autoimmune disease. The impact of the revision of the Th1/Th2 paradigm to
include Th17 cells cannot be overstated. Nonetheless, it is striking that many of the anti-
cytokine therapies that have been developed to target Th1 cells in fact seem to act through the
Th17 lineage instead. It should be noted that, although the Th1 and Th17 cell types are often
considered separately, in fact there is much crosstalk between them (reviewed in {Korn, 2009
#2199; O'Quinn, 2008 #2216}). Understanding how Th17 cells and their downstream cytokines
act at a fundamental level is likely to reveal new strategies for treating RA and other
autoimmune conditions. Conversely, defining the potential side effects of blocking Th17 cells
in animal models as well as rare human populations with genetic defects in these cells is critical
for predicting and being prepared for potential complications associated with this approach.
Acknowledgments
SLG is supported by research grants from the Alliance for Lupus Research, Amgen and the NIH (AR054389,
DE019424). SLG has received travel reimbursement, honoraria and a research grant from Amgen. I thank D. Patel
(Novartis) for sharing unpublished information.
References
1. Mosmann TR, Cherwinski H, Bond MW, et al. Two types of murine helper T cell clone. I. Definition
according to profiles of lymphokine activities and secreted proteins. J Immunol 1986;136:2348–2357.
[PubMed: 2419430]
Gaffen Page 5
Curr Rheumatol Rep. Author manuscript; available in PMC 2010 April 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
2. Steinman L. A brief history of T(H)17, the first major revision in the T(H)1/T(H)2 hypothesis of T
cell-mediated tissue damage. Nature Med 2007;13:139–145. [PubMed: 17290272] * This is an
excellent review article detailing the historical perspective by which discrepancies in the Th1/Th2
paradigm were viewed and the significance of the Th17 cell discovery.
3. Murphy CA, Langrish CL, Chen Y, et al. Divergent pro- and antiinflammatory roles for IL-23 and
IL-12 in joint autoimmune inflammation. The Journal of experimental medicine 2003;198:1951–1957.
[PubMed: 14662908]
4. McGeachy MJ, Cua DJ. The link between IL-23 and Th17 cell-mediated immune pathologies. Semin
Immunol 2007;19:372–376. [PubMed: 18319054]
5. Aggarwal S, Ghilardi N, Xie MH, et al. Interleukin 23 promotes a distinct CD4 T cell activation state
characterized by the production of interleukin 17. The Journal of biological chemistry 2002;3:1910–
1914. [PubMed: 12417590]
6. Aarvak T, Chabaud M, Miossec P, Natvig JB. IL-17 is produced by some proinflammatory Th1/Th0
cells but not by Th2 cells. J Immunol 1999;162:1246–1251. [PubMed: 9973376]
7. Infante-Duarte C, Horton HF, Byrne MC, Kamradt T. Microbial lipopeptides induce the production
of IL-17 in Th cells. J Immunol 2000;165:6107–6115. [PubMed: 11086043]
8. Park H, Li Z, Yang XO, et al. A distinct lineage of CD4 T cells regulates tissue inflammation by
producing interleukin 17. Nature immunology 2005;6:1133–1141. [PubMed: 16200068]
9. Harrington LE, Hatton RD, Mangan PR, et al. Interleukin 17-producing CD4+ effector T cells develop
via a lineage distinct from the T helper type 1 and 2 lineages. Nature immunology 2005;6:1123–1132.
[PubMed: 16200070]
10. McGeachy MJ, Bak-Jensen KS, Chen Y, 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. Nature immunology 2007;8:1390–
1397. [PubMed: 17994024]
11. Duerr RH, Taylor KD, Brant SR, et al. A Genome-Wide Association Study Identifies IL23R as an
Inflammatory Bowel Disease Gene. Science 2006;314:1461–1463. [PubMed: 17068223]
12. Bettelli E, Carrier Y, Gao W, et al. Reciprocal developmental pathways for the generation of
pathogenic effector T(H)17 and regulatory T cells. Nature 2006;441:235–238. [PubMed: 16648838]
13. Mangan PR, Harrington LE, O'Quinn DB, et al. Transforming growth factor-beta induces
development of the T(H)17 lineage. Nature 2006;441:231–234. [PubMed: 16648837]
14. Wei G, Wei L, Zhu J, et al. Global mapping of H3K4me3 and H3K27me3 reveals specificity and
plasticity in lineage fate determination of differentiating CD4+ T cells. Immunity 2009;30:155–167.
[PubMed: 19144320]
15. Lee YK, Turner H, Maynard CL, et al. Late developmental plasticity in the T helper 17 lineage.
Immunity 2009;30:92–107. [PubMed: 19119024]
16. Lohr J, Knoechel B, Caretto D, Abbas AK. Balance of Th1 and Th17 effector and peripheral regulatory
T cells. Microbes Infect. 2009
17. Korn T, Bettelli E, Oukka M, Kuchroo VK. IL-17 and Th17 Cells. Annual review of immunology
2009;27:485–518.
18. Wright JF, Guo Y, Quazi A, et al. Identification of an Interleukin 17F/17A Heterodimer in Activated
Human CD4+ T Cells. The Journal of biological chemistry 2007;282:13447–13455. [PubMed:
17355969]
19. Gaffen SL. Structure and signalling in the IL-17 receptor superfamily. Nat Rev Immunol. 2009 in
press.
20. Shen F, Gaffen SL. Structure-function relationships in the IL-17 receptor: Implications for signal
transduction and therapy. Cytokine 2008;41:92–104. [PubMed: 18178098]
21. Zrioual S, Ecochard R, Tournadre A, et al. Genome-wide comparison between IL-17A- and IL-17F-
induced effects in human rheumatoid arthritis synoviocytes. J Immunol 2009;182:3112–3120.
[PubMed: 19234208] * This paper elegantly compares genes induced by IL-17A versus IL-17F in
human RA synovial fibroblasts, and reveals important insights into mechanisms of synergy with
TNFα
22. Liang SC, Tan XY, Luxenberg DP, et al. Interleukin (IL)-22 and IL-17 are coexpressed by Th17 cells
and cooperatively enhance expression of antimicrobial peptides. The Journal of experimental
medicine 2006;203:2271–2279. [PubMed: 16982811]
Gaffen Page 6
Curr Rheumatol Rep. Author manuscript; available in PMC 2010 April 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
23. Toh ML, Miossec P. The role of T cells in rheumatoid arthritis: new subsets and new targets. Curr
Opin Rheumatol 2007;19:284–288. [PubMed: 17414957]
24. Lubberts E. IL-17/Th17 targeting: on the road to prevent chronic destructive arthritis? Cytokine
2008;41:84–91. [PubMed: 18039580] ** This review summarizes the role of IL-17 and its receptor
in RA, and provides important clinical perspectives into its use in therapy
25. Nakae S, Nambu A, Sudo K, Iwakura Y. Suppression of immune induction of collagen-induced
arthritis in IL-17-deficient mice. J Immunol 2003;171:6173–6177. [PubMed: 14634133]
26. Nakae S, Saijo S, Horai R, et al. IL-17 production from activated T cells is required for the spontaneous
development of destructive arthritis in mice deficient in IL-1 receptor antagonist. Proc Natl Acad Sci
U S A 2003;100:5986–5990. [PubMed: 12721360]
27. Sato K, Suematsu A, Okamoto K, et al. Th17 functions as an osteoclastogenic helper T cell subset
that links T cell activation and bone destruction. The Journal of experimental medicine
2006;203:2673–2682. [PubMed: 17088434]
28. Shen F, Ruddy MJ, Plamondon P, Gaffen SL. Cytokines link osteoblasts and inflammation:
microarray analysis of interleukin-17- and TNF-alpha-induced genes in bone cells. J Leukoc Biol
2005;77:388–399. [PubMed: 15591425]
29. Gaffen SL, Hajishengallis G. A New Inflammatory Cytokine on the Block: Rethinking periodontal
disease and the Th1/Th2 paradigm in the context of Th17 cells and IL-17. J Dent Res 2008;87:817–
828. [PubMed: 18719207]
30. Yu J, Ruddy M, Wong G, et al. An essential role for IL-17 in preventing pathogen-initiated bone
destruction: Recruitment of neutrophils to inflamed bone requires IL-17 receptor-dependent signals.
Blood 2007;109:3794–3802. [PubMed: 17202320]
31. Quinn JM, Sims NA, Saleh H, et al. IL-23 inhibits osteoclastogenesis indirectly through lymphocytes
and is required for the maintenance of bone mass in mice. J Immunol 2008;181:5720–5729. [PubMed:
18832731]
32. Kotake S, Udagawa N, Takahashi N, et al. IL-17 in synovial fluids from patients with rheumatoid
arthritis is a potent stimulator of osteoclastogenesis. J Clin Invest 1999;103:1345–1352. [PubMed:
10225978]
33. Ziolkowska M, Koc A, Luszczukiewicz G, et al. High levels of IL-17 in rheumatoid arthritis patients:
IL-15 triggers in vitro IL-17 production via cyclosporin A-sensitive mechanism. J Immunol
2000;164:2832–2838. [PubMed: 10679127]
34. Honorati MC, Meliconi R, Pulsatelli L, et al. High in vivo expression of interleukin-17 receptor in
synovial endothelial cells and chondrocytes from arthritis patients. Rheumatology (Oxford)
2001;40:522–527. [PubMed: 11371660]
35. Chabaud M, Fossiez F, Taupin JL, Miossec P. Enhancing effect of IL-17 on IL-1-induced IL-6 and
leukemia inhibitory factor production by rheumatoid arthritis synoviocytes and its regulation by Th2
cytokines. J Immunol 1998;161:409–414. [PubMed: 9647250]
36. Cai L, Yin JP, Starovasnik MA, et al. Pathways by which interleukin 17 induces articular cartilage
breakdown in vitro and in vivo. Cytokine 2001;16:10–21. [PubMed: 11669582]
37. Shahrara S, Pickens SR, Dorfleutner A, Pope RM. IL-17 induces monocyte migration in rheumatoid
arthritis. J Immunol 2009;182:3884–3891. [PubMed: 19265168]
38. Kirkham BW, Lassere MN, Edmonds JP, et al. Synovial membrane cytokine expression is predictive
of joint damage progression in rheumatoid arthritis: a two-year prospective study (the DAMAGE
study cohort). Arthritis and rheumatism 2006;54:1122–1131. [PubMed: 16572447]
39. Yamada H, Nakashima Y, Okazaki K, et al. Th1 but not Th17 cells predominate in the joints of
patients with rheumatoid arthritis. Annals of the rheumatic diseases 2008;67:1299–1304. [PubMed:
18063670]
40. Scheinecker C, Redlich L, Smolen J. Cytokines as thereapeutic targets: Advances and limitations.
Immunity 2008;28:440–444. [PubMed: 18400186]
41. Zaba LC, Cardinale I, Gilleaudeau P, et al. Amelioration of epidermal hyperplasia by TNF inhibition
is associated with reduced Th17 responses. The Journal of experimental medicine 2007;204:3183–
3194. [PubMed: 18039949] ** This paper shows that the success of TNFα blockade may in part be
due to suppressive effects on Th17 activity
Gaffen Page 7
Curr Rheumatol Rep. Author manuscript; available in PMC 2010 April 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
42. Reich K, Yasothan U, Kirkpatrick P. Ustekinumab. Nat Rev Drug Discov 2009;8:355–356. [PubMed:
19404310]
43. Smolen JS, Beaulieu A, Rubbert-Roth A, et al. Effect of interleukin-6 receptor inhibition with
tocilizumab in patients with rheumatoid arthritis (OPTION study): a double-blind, placebo-
controlled, randomised trial. Lancet 2008;371:987–997. [PubMed: 18358926]
44. Mclnnes IB, Schett G. Cytokines in the pathogenesis of rheumatoid arthritis. Nat Rev Immunol
2007;7:429–442. [PubMed: 17525752]
45. Strangfeld A, Listing J. Infection and musculoskeletal conditions: Bacterial and opportunistic
infections during anti-TNF therapy. Best Pract Res Clin Rheumatol 2006;20:1181–1195. [PubMed:
17127203]
46. O'Quinn D, Palmer M, Lee Y, Weaver C. Emergence of the Th17 pathway and its role in host defense.
Adv Immunol 2008;99:115–163. [PubMed: 19117534]
47. Conti H, Shen F, Nayyar N, et al. Th17 cells and IL-17 receptor signaling are essential for mucosal
host defense against oral candidiasis. The Journal of experimental medicine 2009;206:299–311.
[PubMed: 19204111]
48. Fischer A. Connecting STAT3, Th17 and human mucosal immunity. Immunol Cell Biol
2008;86:549–551. [PubMed: 18645579] * This review summarizes the discovery of Th17-deficient
humans with Hyper-IgE syndrome, which may provide important insight into the consequences of
IL-17 blockade for therapy
49. Freeman AF, Holland SM. The hyper-IgE syndromes. Immunology and allergy clinics of North
America 2008;28:277–291. viii. [PubMed: 18424333]
Gaffen Page 8
Curr Rheumatol Rep. Author manuscript; available in PMC 2010 April 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Figure 1. Old versus new models of Th cell development
In the original paradigm of CD4+ T cell differentiation, most CD4+ T helper cells could be
classified into Th1 cells (typified by production of IFNγ) or Th2 cells (typified by production
of IL-4, IL-5 and IL-13). Th1 cells were considered to promote host defense against most
infectious microbes (both intracellular and extracellular), whereas Th2 cells were thought to
act primarily against large Helminth worms. Th1 cells were also considered to be the major
driver of autoimmune pathology in RA as well as other autoimmune diseases such as psoriasis,
multiple sclerosis and Crohn's Disease. The heterodimeric cytokine IL-12, composed of the
IL-12p40 and IL-12p35 subunits, was shown to be critical for development of Th1 cells, and
hence efforts to block IL-12 with antibodies against the IL-12p40 subunit were pursued. In
2005, the discovery of the Th17 subset revised this model, which was partly revealed based
on a new understanding of the shared role of the IL-12p40 subunit in the cytokine IL-23. Th17
cells are driven to differentiation by a combination of IL-1, IL-6 and TGFβ, and IL-23
(composed of the IL-12p40 and the IL-23p19 subunits) serves to expand and stabilize this
lineage.
Gaffen Page 9
Curr Rheumatol Rep. Author manuscript; available in PMC 2010 April 1.
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
NIH-PA Author Manuscript NIH-PA Author Manuscript NIH-PA Author Manuscript
Gaffen Page 10
Table 1
T helper subsets and their major functions
Th1 Th2 Th17 iTreg
Major cytokines IFNγIL-4, IL-5, IL-13 IL-17A, IL-17F,
IL-21, IL-22, IL-26
(humans), TNFα,
CCL20
IL-10, TGFβ
Main Antimicrobial activity Intracellular microbes Helminths Extracellular microbes Limit immune-
mediated
damage,
prevent
autoimmunity
Inductive cytokines IL-12 IL-4 IL-1, IL-6, TGFβTGFβ, IL-2
Inductive transcription factors STAT1, Tbet STAT6, GATA3 STAT3, RORγt STAT5, Foxp3
Curr Rheumatol Rep. Author manuscript; available in PMC 2010 April 1.
... CRP is also deposited in the synovium (33), raising the possibility that CRP is present in the synovium in both, native and non-native, pentameric conformations. It has also been shown that the serum levels of IL-17, IL-6, TNF-a, IL-10, IL-2 and IL-1b are altered in RA patients (28,34,35). CRP is used as a nonspecific biomarker of inflammation during the development of RA (3,36). ...
... IL-17 and TNF-a play crucial roles in the development of CIA (34,57,69). That serum levels of IL-17 and TNF-a increase in CIA mice has also been reported previously (57,(70)(71)(72)(73)(74)(75)(76). ...
Article
Full-text available
The biosynthesis of C-reactive protein (CRP) in the liver is increased in inflammatory diseases including rheumatoid arthritis. Previously published data suggest a protective function of CRP in arthritis; however, the mechanism of action of CRP remains undefined. The aim of this study was to evaluate the effects of human CRP on the development of collagen-induced arthritis (CIA) in mice which is an animal model of autoimmune inflammatory arthritis. Two CRP species were employed: wild-type CRP which binds to aggregated IgG at acidic pH and a CRP mutant which binds to aggregated IgG at physiological pH. Ten CRP injections were given on alternate days during the development of CIA. Both wild-type and mutant CRP reduced the incidence of CIA, that is, reduced the number of mice developing CIA; however, CRP did not affect the severity of the disease in arthritic mice. The serum levels of IL-17, IL-6, TNF-α, IL-10, IL-2 and IL-1β were measured: both wild-type and mutant CRP decreased the level of IL-17 and IL-6 but not of TNF-α, IL-10, IL-2 and IL-1β. These data suggest that CRP recognizes and binds to immune complexes, although it was not clear whether CRP functioned in its native pentameric or in its structurally altered pentameric form in the CIA model. Consequently, ligand-complexed CRP, through an as-yet undefined mechanism, directly or indirectly, inhibits the production of IL-17 and eventually protects against the initiation of the development of arthritis. The data also suggest that IL-17, not TNF-α, is critical for the development of autoimmune inflammatory arthritis.
... The image was created using Adobe Illustrator (ver. 27 strongly related to IL-17 signaling and JAK-STAT signaling pathways, which are important for autoimmune inflammation, as well as an innate immune response such as trypanosomiasis or malaria [29,30]. ...
Article
Full-text available
Objectives Compared to conventional disease-modifying antirheumatic drugs (DMARDs), biological DMARDs demonstrate superior efficacy but come with higher costs and increased infection risks. The ability to stop and resume biological DMARD treatment while maintaining remission would significantly alleviate these barriers and anxieties. The objective of this study was to identify biomarkers that can predict an imminent relapse, hopefully enabling the timely resumption of biological DMARDs before relapse occurs. Methods Forty patients with rheumatoid arthritis who had been in remission for more than 12 months were included in the study. The patients discontinued their biological DMARD treatment and were monitored monthly for the next 24 months. Out of the 40 patients, 14 (35%) remained in remission at the end of the 24-month period, while 26 (65%) experienced relapses at different time points. Among the relapse cases, 13 patients experienced early relapse within 6 months, and another 13 patients had late relapse between 6 months and 24 months. Seventy-three cytokines in the sera collected longitudinally from the 13 patients with late relapse were measured by multiplex immunoassay. Using cytokines at two time points, immediately after withdrawal and just before relapse, volcano plot and area under the receiver operating characteristic curves (AUC) were drawn to select cytokines that distinguished imminent relapse. Univariate and multivariate logistic regression analyses were used for the imminent relapse prediction model. Results IL-6, IL-29, MMP-3, and thymic stromal lymphopoietin (TSLP) were selected as potential biomarkers for imminent relapse prediction. All four cytokines were upregulated at imminent relapse time point. Univariate and multivariate logistic regression showed that a combination model with IL-6, MMP-3, and TSLP yielded an AUC of 0.828 as top predictors of imminent relapse. Conclusions This methodology allows for the prediction of imminent relapse while patients are in remission, potentially enabling the implementation of on- and off-treatments while maintaining remission. It also helps alleviate patient anxiety regarding the high cost and infection risks associated with biological DMARDs, which are the main obstacles to benefiting from their superb efficacy.
... IL-1β and TNF-α, two major proinflammatory cytokines, are both known to be present at high concentrations in serum and synovial fluid in patients with RA [5]. IL-1β and TNF-α stimulate their own production and the production of other cytokines, amplifying the inflammation process [8] and contribute synergistically to produce the inflammasome [17]. In our study, TNF-α and IL-1β expression were significantly reduced only by Glu+Hyal A+Sal. ...
... In this study, we planned to gauge the serum IL-17 levels of menopausal women, while a simple expansion in circulating IL-17 may not be the reaction to estradiol depletion. Likewise, some studies noticed a rise in this cytokine in the synovial fluid of patients with rheumatoid arthritis [28]. Given the design of the study, we did not perform invasive procedures. ...
Article
Full-text available
Introduction The reciprocation between systemic inflammatory markers (SIMs), dyslipidemia, and hot flashes (HFs) can play a part in the pathogenesis of endothelial dysfunction through menopause. This study intended to determine the association between some SIMs, lipids, and HFs in healthy menopausal women. Materials and methods We designed a cross-sectional study in which 160 healthy menopausal women aged 40–60 were enrolled. Concerning their HFs status, they were stratified into two groups by consecutive sampling: without HFs (n = 40) and with HFs (n = 120). In addition to clinical variables and HFs experience, we measured the fasting serum levels of SIMs and lipid profiles (LPs), including Interleukin-17 (IL-17), high- sensitivity C-Reactive Protein (hs-CRP), Total Cholesterol (TC), Triglycerides (TG), Low-Density Lipoprotein Cholesterol (LDL-C), and High-Density Lipoprotein Cholesterol (HDL-C) in each group. Then, we calculated TC/HDL-C concerning the related variables and determined Neutrophil-to-Lymphocyte Ratio (NLR), and Lymphocyte-to-Monocyte Ratio (LMR), according to Complete Blood Count (CBC) quantitative parameters in each group. Furthermore, we used logistic regression analysis to assess the association between SIMs, LPs, and HFs. Settings We performed this study in a governmental teaching hospital, Guilan/Rasht, Iran, from April to September 2021. Results The two groups of menopausal women without and with HFs were not significantly different regarding the median of IL-17, hs-CRP, NLR, LMR, TG, HDL-C, and TC/HDL-C, and the mean of TC and LDL-C. Based on multiple logistic regression, TG levels appeared to be associated with the incidence of HFs (B = 0.004, P = 0.040, Odds Ratio:1.004, 95%CI:1.000–1.009). NLR seemed to have an increasing impact on the HFs severity, according to ordinal logistic regression (B = 0.779, P = 0.005, Odds Ratio = 2.180, 95%CI:1.270–3.744). Furthermore, hs-CRP negatively correlated with TG (r = -0.189, P = 0.039) and TC/HDL-C (r = -0.268, P = 0.003) in menopausal women with HFs. Conclusion This study indicated an association between SIMs, lipids, and HFs. These connections may suggest HFs as links between SIMs/LPs alterations and their outcomes.
... IL-17A primarily acts on synoviocytes and osteoblasts to mediate cartilage and bone degradation in RA. In vitro experiments reveal that IL-17A causes synoviocyte activation (e.g., the production of IL-6 and IL-8), migration, and invasion, all of which increase cartilage degradation [7]. ...
Article
Full-text available
Rheumatoid arthritis (RA) is an autoimmune inflammatory disease that causes multi-articular synovitis. The illness is characterized by worsening inflammatory synovitis, which causes joint swelling and pain. Synovitis erodes articular cartilage and marginal bone, resulting in joint deterioration. This bone injury is expected to be permanent. Cytokines play a prominent role in the etiology of RA and could be useful as early diagnostic biomarkers. This research was carried out at Riyadh’s King Khalid University Hospital (KKUH). Patients were enrolled from the Rheumatology unit. Seventy-eight RA patients were recruited (67 (85.9%) females and 11 (14.1%) males). Patients were selected for participation by convenience sampling. Demographic data were collected, and disease activity measurements at 28 joints were recorded using the disease activity score (DAS-28). Age- and sex-matched controls from the general population were included in the study. A panel of 27 cytokines, chemokines, and growth factors was determined in patient and control sera. Binary logistic regression (BLR) and discriminant analysis (DA) were used to analyze the data. We show that multiple cytokine biomarker profiles successfully distinguished RA patients from healthy controls. IL-17, IL-4, and RANTES were among the most predictive variables and were the only biomarkers incorporated into both BLR and DA predictive models for pooled participants (men and women). In the women-only models, the significant cytokines incorporated in the model were IL-4, IL-17, MIP-1b, and RANTES for the BLR model and IL-4, IL-1Ra, GM-CSF, IL-17, and eotaxin for the DA model. The BLR and DA men-only models contained one cytokine each, eotaxin for BLR and platelet-derived growth factor-bb (PDGF-BB) for DA. We show that BLR has a higher fidelity in identifying RA patients than DA. We also found that the use of gender-specific models marginally improves detection fidelity, indicating a possible benefit in clinical diagnosis. More research is needed to determine whether this conclusion will hold true in various and larger patient populations.
... The study compared the level promoting rheumatoid arthritis, owing to the downregulation of IL-17A-induced proinflammatory mediators (IL-1 and TNF-α) and a modulation in the RANKL/OPG ratio. The important involvement of IL-17A in RA was evidenced by high levels of Th17 cells, IL-17A, and its receptor in synovial fluid of the RA patients (Kohno et al., 2008;Gaffen, 2009;Elhewala et al., 2015;Dhaouadi et al., 2018). ...
Article
Full-text available
Bone metabolism requires a balance mechanism between intricate processes of bone formation and resorption, which is affected by the essential interaction between osteoblasts and osteoclasts. Interleukin-17 (IL-17) is a family of cytokines consisting of six isoforms: IL-17A, IL-17B, IL-17C, IL-17D, IL-17E, and IL-17F. Among these isoforms, IL-17A has shown promising and novel roles in the regulation of bone metabolism. IL-17A has also captivated the interest of many researchers since its discovery because of its role as a pro-inflammatory cytokine which mediates a few inflammatory processes. This review describes the biology of IL-17A and its receptor as well as summarises the regulatory role of IL-17A in bone metabolism and diseases through some known pathways. Understanding the function of IL-17A in bone metabolism may lead to the development of therapeutics or diagnostic strategies for some bone diseases; as well as future therapy using tissue engineering and regenerative medicine approaches.
... The systemic inflammation observed in RA shows extensive damage beyond the joints to include for example the cardiovascular, skin, lungs, and eyes (Scott et al., 2010;Bordy et al., 2018). The proinflammatory cytokine IL-17 that is produced by specialized CD+ T helper (Th17) cells and oxidative stress such as ROS, are associated with the pathogenesis of RA in both, human and animal models (Gaffen, 2009;García-González et al., 2015). IL-17A increased autoimmunity which is a characteristic feature of RA (Binger et al., 2017), ROS production (Dhillion et al., 2012), and fibrosis . ...
Article
Full-text available
Rheumatoid arthritis (RA) that affects the synovial knee joint causes swelling of the synovial membrane and tissue damage. Interleukin-17A (IL-17A) and the enzyme glycogen synthase kinase-3β (GSK3β) are involved in the pathogenesis of RA. The link between IL-17A, GSK3β, the oxidative stress, and the profibrogenic marker alpha-smooth muscle actin (α-SMA) with and without TDZD-8, GSK3β inhibitor has not been studied before. Consequently, active immunization of rats was performed to induce RA after three weeks using collagen type II (COII) injections. The treated group received daily injection of 1 mg/kg TDZD-8 for 21 days following the immunization protocol (COII+TDZD-8). Blood and synovium tissue samples were harvested at the end of the experiment. RA development was confirmed as corroborated by a substantial increase in blood levels of the highly specific autoantibody for RA, anti-citrullinated protein antibody as well as augmentation of reactive oxidative species (ROS) levels measured as lipid peroxidation. RA induction also increased synovium tissue levels of IL-17A and the profibrogenic marker, α-SMA. All these parameters seemed to be significantly (p<0.0001) ameliorated by TDZD-8. Additionally, a significant correlation between IL-17A, ROS, and α-SMA and biomarkers of RA was observed. Thus, knee joint synovium RA induction augmented IL-17A/GSK3β/ROS/α-SMA axis mediated arthritis in a rat model of RA, which was inhibited by TDZD-8.
Article
A bstract Objective To present a comprehensive flow cytometry panel for idiopathic subglottic stenosis (iSGS). Study Design Controlled ex vivo cohort study. Setting Tertiary care academic hospital in a metropolitan area. Methods Flow cytometry and single‐cell RNA sequencing were performed on 9 paired normal and scar tissue samples from iSGS patients. Flow cytometry was used to assess the presence of myeloid (CD11b, CD14, CD15, Siglec8), lymphoid (CD3, CD4, CD8, gamma delta [γδ], FOXP3), endothelial (CD31), fibroblast (CD90, SMA), and epithelial (CD326, CK5) markers. Results On flow cytometry, iSGS scar is characterized by an increased presence of myeloid, lymphoid, endothelial, and fibroblast cell types, but a decreased presence of epithelial cells. In the myeloid lineage, iSGS scar samples demonstrated increased CD11b ⁺ monocytes ( P < .001), Siglec8 ⁺ eosinophils ( P = .03), and CD14 ⁺ monocytes ( P = .02). In the lymphoid lineage, iSGS scar demonstrated increased CD3 ⁺ T‐cells ( P < .001), CD4 ⁺ helper T‐cells ( P < .001), γδ ⁺ T‐cells ( P < .001), and FOXP3 ⁺ regulatory T‐cells ( P = .002). iSGS scar exhibited specific increases in CD90 ⁺ ( P = .04) and SMA ⁺ ( P < .001) fibroblasts but decreased CD326 ⁺ (E‐cadherin) epithelial cells ( P = .01) relative to normal samples. Conclusion We present a comprehensive flow cytometry panel for iSGS. This flow panel may serve as a common platform among airway scientists to elucidate the cellular mechanisms underpinning iSGS and other upper airway pathologies. Scar iSGS samples demonstrate a distinct cellular profile relative to normal iSGS specimens, exhibiting increased fibroblast, endothelial, and inflammatory cell types but decreased epithelium.
Article
Introduction: Gout arthritis (GA) is an intermittent inflammatory disease affecting approximately 10% of the worldwide population. Symptomatic phases (acute flares) are timely spaced by asymptomatic periods. During an acute attack, redness, joint swelling, limited movement, and excruciating pain are common symptoms. However, the current available therapies are not fully effective in reducing symptoms and offered numerous side effects. Therefore, unveiling new drug targets and effector molecules are required in developing novel GA therapeutics. Areas covered: This review discusses the pathophysiological mechanisms of GA and explore potential pharmacological targets to ameliorate disease outcome. In addition, we listed promising pre-clinical studies demonstrating effector molecules with therapeutical potential. Among those, we emphasized the importance of natural products, including traditional Chinese medicine formulas and their multitarget mechanisms of action. Expert opinion: In our search, we observed that there is a massive gap between pre-clinical and clinical knowledge. Only a minority (4.4%) of clinical trials aimed to intervene applying natural products, or current hot targets described herein. In this sense, we envisage four possibilities for GA therapeutics, which includes the repurposing of existing therapies, ALX/FPR2 agonism for improvement in disease outcome, the use of multitarget drugs (e.g. natural products), and targeting the neuroinflammatory component of GA.
Article
Full-text available
Studies have shown that transforming growth factor- (TGF-) and interleukin 6 (IL-6) are required for the lineage commitment of pathogenic IL-17-producing T helper cells (TH-17 cells). Unexpectedly, here we found that stimulation of myelin-reactive T cells with TGF- plus IL-6 completely abrogated their pathogenic function despite upregulation of IL-17 production. Cells stimulated with TGF- plus IL-6 were present in the spleen as well as the central nervous system, but they failed to upregulate the proinflammatory chemokines crucial for central nervous system inflammation. In addition, these cells produced IL-10, which has potent anti-inflammatory activities. In contrast, stimulation with IL-23 promoted expression of IL-17 and proinflammatory chemokines but not IL-10. Hence, TGF- and IL-6 'drive' initial lineage commitment but also 'restrain' the pathogenic potential of TH-17 cells. Our findings suggest that full acquisition of pathogenic function by effector TH-17 cells is mediated by IL-23 rather than by TGF- and IL-6.
Article
Full-text available
Interleukin-17A (IL-17A), the hallmark cytokine of the newly defined T helper 17 (T(H)17) cell subset, has important roles in protecting the host against extracellular pathogens, but also promotes inflammatory pathology in autoimmune disease. IL-17A and its receptor (IL-17RA) are the founding members of a newly described family of cytokines and receptors that have unique structural features which distinguish them from other cytokine families. Research defining the signal transduction pathways induced by IL-17R family cytokines has lagged behind that of other cytokine families, but studies in the past 2 years have begun to delineate unusual functional motifs and new proximal signalling mediators used by the IL-17R family to mediate downstream events.
Article
Nature Reviews Immunology 9, 556–567 (2009); published online 3 July 2009; corrected after print 7 August 2009 In the version of this article initially published, reference 22 was incorrectly stated to be the first report to directly compare Il17a−/− and Il17f−/− mice and to show that these cytokines have markedly different functions in vivo.
Article
For over 35 years, immunologists have divided T-helper (TH) cells into functional subsets. T-helper type 1 (TH1) cells - long thought to mediate tissue damage - might be involved in the initiation of damage, but they do not sustain or play a decisive role in many commonly studied models of autoimmunity, allergy and microbial immunity. A major role for the cytokine interleukin-17 (IL-17) has now been described in various models of immune-mediated tissue injury, including organ-specific autoimmunity in the brain, heart, synovium and intestines, allergic disorders of the lung and skin, and microbial infections of the intestines and the nervous system. A pathway named TH17 is now credited for causing and sustaining tissue damage in these diverse situations. The TH1 pathway antagonizes the T H17 pathway in an intricate fashion. The evolution of our understanding of the TH17 pathway illuminates a shift in immunologists' perspectives regarding the basis of tissue damage, where for over 20 years the role of TH1 cells was considered paramount.
Article
In January 2009, ustekinumab (Stelara; Janssen Cilag) was granted marketing authorization by the European Commission for the treatment of moderate to severe plaque psoriasis in adults who failed to respond to, or who have a contraindication to, or are intolerant to other therapies including cyclosporine, methotrexate and phototherapy.
Article
Immunology and Cell Biology focuses on the general functioning of the immune system in its broadest sense, with a particular emphasis on its cell biology. Areas that are covered include but are not limited to: Cellular immunology, Innate and adaptive immunity, Immune responses to pathogens,Tumour immunology,Immunopathology, Immunotherapy, Immunogenetics, Immunological studies in humans and model organisms (including mouse, rat, Drosophila etc)
Article
Transfer of antigen-specific T cells into antigen-expressing lymphopenic recipients leads to the sequential generation of Th1 and Th17 effector and protective CD25(+)FoxP3(+) regulatory cells in the periphery with surprisingly different kinetics. Such an experimental model is potentially valuable for defining the stimuli that regulate lineage decision and plasticity of various T cell effectors and peripheral regulatory T cells. Our studies have shown that IL-17 production occurs rapidly and declines within the first week with the appearance of IFN-gamma producing T cells. Regulatory T cells appear during the recovery phase of the disease. The factors that mediate this complex differentiation originating from a starting naïve T cell population remain to be defined.