Gaffen2009 1 3

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 3

The Role of Interleukin-17 in the

Pathogenesis of Rheumatoid Arthritis


Sarah L. Gaffen, PhD

Corresponding author by macrophage activation and opsonizing antibodies


Sarah L. Gaffen, PhD through the actions of the cytokine interferon (IFN)-γ.
Department of Medicine, Division of Rheumatology and Clinical Conversely, Th2 cells mediate humoral immunity char-
Immunology, University of Pittsburgh, Pittsburgh, PA 15261, USA.
E-mail: sig65@pitt.edu
acterized by activation of B cells and effector antibodies,
which is mediated by interleukin (IL)-4, IL-5, and IL-13
Current Rheumatology Reports 2009, 11:365–370
Current Medicine Group LLC ISSN 1523-3774 (Fig. 1). Central to this model was the concept that naïve
Copyright © 2009 by Current Medicine Group LLC Th cells are not predetermined to be either Th1 or Th2,
but rather that the environment in which they encounter
an antigen dictates their subsequent fate. In accordance
Interleukin (IL)-17 (also known as IL-17A), the sig- with this idea, it was shown that IL-12, usually derived
nature cytokine of the newly described T helper 17 from antigen-presenting cells, such as dendritic cells,
(Th17) cell population, has been implicated in the directs differentiation of naïve CD4+ T cells into the Th1
pathogenesis of numerous autoimmune diseases lineage, whereas IL-4 promotes development to the Th2
including rheumatoid arthritis. IL-17 is the found- lineage. Moreover, differentiation of each Th subset is
ing member of a new subclass of cytokines that have mutually antagonistic and self-reinforcing by cross-antag-
highly proinflammatory properties. Studies in rodents onistic signaling pathways mediated by IL-12 and IL-4.
and mammalian cell culture systems, as well as clinical For nearly 20 years, the Th1/Th2 model provided a valu-
settings, support a role for IL-17 in promoting rheu- able framework in which diseases, both infectious and
matoid arthritis. This article discusses the history of autoimmune, were evaluated. Much research effort was
the discovery of Th17 cells, the potential mechanisms devoted to defi ning mechanisms that control Th1/Th2
of action of IL-17 in autoimmunity, and perspectives development, as well as the key cytokines, transcription
for IL-17–targeted cytokine therapy. factors, and microbial/autoimmune stimuli involved in
this process [2•].
However, over time, glaring discrepancies became
Introduction apparent with this model, particularly related to Th1
During the past several years, there has been a paradigm generation and function [2•]. The generation of various
shift in our understanding of the role of T cells in autoim- gene deficient (knockout) mouse strains revealed puzzling
munity, particularly with regard to the CD4+ T helper (Th) differences between the Th1 effector cytokine IFN-γ and
cell subset. Th cells have long been known as the “master- the Th1 differentiation cytokine IL-12 (Fig. 1). IL-12
minds” of the immune system, controlling the activities of is a heterodimeric cytokine composed of two subunits,
other lymphocytes and many aspects of the innate immune IL-12p40 and IL-12p35. IL-12p40–deficient mice were
response. Their depletion in the setting of HIV infection developed to probe the role of this cytokine in vivo, and
highlights the vital role of CD4+ T cells in coordinating IL-12p40 –/– mice were found resistant to most autoimmune
immunity to infectious diseases. Th cells accomplish their disease models, including the rodent model of RA, colla-
vital tasks primarily by means of secreting cytokines, gen-induced arthritis (CIA) [3]. However, IFN-γ deficient
protein-based hormones that bind to specific receptors on mice and also IFN-γ–receptor deficient mice appeared
target cells. Cytokines have pleiotropic activities, which resistant to CIA and many other autoimmune disease mod-
include, for example, triggering proliferation, cell death, els; in fact, in some instances these mice showed enhanced
specific gene expression, and cellular migration. sensitivity to disease [3]. The basis for this paradox began
In 1986, a seminal model by Mossman et al. [1] pos- to become clear when IL-12p35-deficient mice were gener-
tulated that different subpopulations of Th cells could ated, which appeared to have a highly similar phenotype
shape immune responses by virtue of differential cytokine to the IFN-γ–deficient mice and a less severe phenotype
production. Two subsets were initially identified that than IL-12p40 –/– animals. Thus, it was apparent that IL-
were termed Th1 and Th2 (Table 1). This model posits 12p40 plays an IL-12–independent role in immunity.
that Th1 cells mediate cellular immunity, characterized Indeed, IL-12p40 is also a component of a new cytokine,
366
I Rheumatoid Arthritis

Table 1. T-helper subsets: inductive factors and their major functions


Main antimicrobial Inductive transcription
Th subset Major cytokines activity Inductive cytokines factors
Th1 IFN-γ Intracellular microbes IL-12 STAT1, T-bet
Th2 IL-4, -5, -13 Helminths IL-4 STAT6, GATA3
Th17 IL-17A, -17F, -21, Extracelluar microbes IL-1, IL-6, TGF-β STAT3, RORγt
-22, -26 (humans),
TNF-α, CCL20
Treg IL-10, TGF-β Limit immune- TGF-β, IL-2 STAT5, FOXP3
mediated damage,
prevent autoimmunity
CCL20—chemokine (C-C motif) ligand 20; IFN—interferon; IL—interleukin; RORγt—retinoic acid receptor–related orphan
receptor γ–tissue distribution; STAT—signal transducers and activators of transcription; T-bet—transcription factor of the T-box family;
TGF-β–transforming growth factor–β; Th—T helper; TNF-α—tumor necrosis factor–α; Treg—T regulatory cells.

Figure 1. Old versus new models of T helper (Th) cell development. In the original paradigm of CD4+ T cell differentiation, most CD4+ Th
cells (Thp) could be classified into Th1 cells (typified by production of interferon [IFN]-γ) or Th2 cells (typified by production of interleukin
[IL]-4, IL-5, and IL-13). Th1 cells were considered to promote host defense against most infectious microbes (both intracellular and extracel-
lular), 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 rheumatoid arthritis and 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 tumor necrosis factor–α, and IL-23 (composed of the IL-12p40 and the
IL-23p19 subunits) serves to expand and stabilize this lineage. TGF-β–transforming growth factor–β.

IL-23, where it partners with a unique IL-23p19 subunit cytokine [6]. Interestingly, studies in Lyme disease arthritis
(Fig. 1). Accordingly, IL-12p40–deficient mice are deficient models demonstrated that a distinct subset of CD4+ T cells
not only in IL-12, but also in IL-23. Direct comparisons produced IL-17A [7], although the connection to IL-23 was
of IL-23p19–deficient mice and IL-12p35–deficient began not made at that time. Putting this together, two landmark
to tease apart the biological functions of these two cyto- reports showed that an IL-17A–producing CD4+ T cells
kines, and it became clear that many of the autoimmune subset exists that can be induced to differentiate in vitro
functions that had previously been ascribed to IL-12 (eg, in [8,9]. Moreover, induction of this lineage was found to
CIA, encephalomyelitis, inflammatory bowel disease), were be independent on the classic Th1/Th2 signal transducers
in fact due to the activities of IL-23 [4]. and activators of transcription (STAT) factors, STAT4 and
Based on these findings, the function of IL-23 became STAT6, and development of this so-called Th17 population
a critical question. In other words, if Th1 cells were not could be inhibited by Th1- and Th2-specific cytokines,
the main effectors of autoimmunity, was there a new, IFN-γ, and IL-4 (Table 1). A veritable avalanche of reports
IL-23–dependent T cell subset that had previously been followed in the next few years, demonstrating that Th17
overlooked? Indeed, IL-23 was shown to stimulate produc- cells are indeed a separate lineage [4]. Unexpectedly and in
tion of IL-17 (often referred to as IL-17A) in murine CD4+ contrast to IL-12, IL-23 was not essential for differentiation
T cells [5]. IL-17A, although primarily derived from T of Th17 cells, per se, but rather for their pathogenicity in
cells, was not obviously a Th1 or Th2 cytokine, hinting at vivo [10]. However, in mice, IL-23 is nonetheless essential
the possibility of a new CD4+ T cell lineage involving this for stable development of Th17 cells, and polymorphisms
The Role of IL-17 in the Pathogenesis of RA
I Gaffen
I 367

in the IL23R gene in humans are linked to susceptibility to cytokines such as IL-1 or Toll-like receptor agonists. All
Crohn’s disease [11]. these cytokines activate nuclear factor κ-light-chain-
Whereas IL-23 is not required for Th17 differentiation, enhancer of activated B cells (NF-κB), and many also have
a combination of transforming growth factor (TGF)-β, been shown to induce mitogen-activated protein kinases
IL-1, and IL-6 serve in this capacity, and the transcription (MAPK) signaling and the cytidine-cytidine-adenosine-
factor RAR-related orphan receptor γ–tissue distribu- adenosine-thymidine (CCAAT)-enhancer-binding protein
tion (RORγt) was found to be key for differentiation of transcription factors. Thus, the net effect of IL-17A signal-
these cells [4]. Strikingly, Th17 cells arise in opposition ing is to induce an innate-type inflammatory effector gene
to inducible T regulatory cells (Tregs), in part by virtue expression program that mediates potent inflammation and
of the fact that both lineages depend on TGF-β for their plays a key role in host defense [20]. Conversely, in condi-
differentiation [12,13]. Thus, in settings of autoimmunity, tions of dysregulation, this inflammatory profile can also
an altered balance between immunosuppressive Tregs promote inflammatory pathology in autoimmunity.
and inflammatory Th17 cells appears to be a major com- Much of our understanding of IL-17A biology in
ponent in disease pathogenesis. Related to this issue, an both infection and autoimmunity comes from microar-
important and often unappreciated feature of Th cells in ray studies of downstream target genes. Receptors for
vivo is their lineage plasticity. Specifically, differentiation IL-17 are expressed ubiquitously, but the key responsive
to specific Th1/2/17/Treg phenotypes is not fi xed, but cells tend to be nonimmune cells such as epithelial cells,
rather there is considerable interconversion among these mesenchymal cells (myoblasts, fibroblasts and adipocytes,
cell types observed in vivo and in vitro [14,15]. In particu- osteoblasts), and keratinocytes. IL-17 stimulates expres-
lar, Treg and Th17 cells have been shown to interconvert. sion of inflammatory genes, including cytokines (IL-6,
Understanding precisely how and under what circum- G-CSF, oncostatin M, IL-32), chemokines (chemokine
stances this occurs has important implications in terms of [C-X-C motif] ligand 1; chemokine [C-X-C motif] ligand
treating autoimmune disease [16]. 2; chemokine [C-X-C motif] ligand 5; chemokine [C-C
Although IL-17A is predominantly produced by T motif] ligand 20) and other inflammatory effectors (induc-
cells, it is not only the province of the CD4+ subset. CD8+ ible nitric oxide synthase [iNOS], antimicrobial genes,
T cells also produce considerable amounts of IL-17A, as acute phase response genes) [20]. Accordingly, although
do γδ-T cells, natural killer T cells, and lymphoid tissue IL-17A is made by T cells, its downstream signals are sim-
inducer cells [17]. ilar to those induced by typical innate immune receptors
such as TLR ligands (eg, lipopolysaccharide) or IL-1β.
As indicated above, IL-17A and IL-17F synergize
IL-17 Receptor Signaling Bridges Innate potently with TNF-α, although the mechanisms under-
and Adaptive Immunity lying this synergy are only partially understood. Thus,
Although IL-17A is eponymous with the Th17 lineage, IL-17A may act as a rheostat for TNF-α signaling. A
these cells also produce IL-17F, IL-21, IL-22, IL-26 (the recent study in human synovial fibroblasts compared
gene for which is found only in humans), and in some genes induced by IL-17A versus IL-17F, and found that
reports tumor necrosis factor (TNF)-α, chemokine (C- they promoted similar profi les of gene expression,
C motif) ligand 20, and granulocyte-colony stimulating although IL-17A was quantitatively more active [21•]. In
factor (G-CSF) (Table 1) [17]. Although these are very dif- the presence of TNF-α, the patterns of induced genes were
ferent cytokines, they all function in a similar manner to highly similar. Interestingly, IL-17A and IL-17F upregu-
promote inflammation, and in some cases can act coop- late expression of TNF receptor 2 in synoviocytes [21•],
eratively or synergistically. IL-17A and IL-17F are quite which may help explain the basis for synergy between
homologous (about 55%) and can form heterodimers these cytokines. IL-17A also cooperates with IL-1β and
(IL-17A/F) that appear to be the major isoform of the IL-22, although the synergy is generally not as dramatic
cytokine in human peripheral blood mononuclear cells as that seen with TNF-α [22]. Therefore, Th17-related
[18]. All three cytokine forms bind to a common receptor cytokines act cooperatively to amplify inflammatory cas-
complex, but exhibit distinct signaling potencies, with the cades, which is beneficial in host defense but deleterious
following activities: IL-17A > IL-17A/F > IL-17F. in many autoimmune settings.
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 IL-17 Promotes Disease in Murine Models
cytokine receptors with distinct molecular features that of Rheumatoid Arthritis
distinguish this family from other types of receptors [19]. The pathogenic role of IL-17A in murine models of
IL-17A and IL-17F signaling through their receptors is inflammatory arthritis is unequivocal [23]. Elegant work
unusual compared with typical adaptive Th cell cytokines. by Lubberts et al. [24••] has demonstrated a key role for
Rather than activating Janus tyrosine kinase (JAK)–STAT IL-17 in collage-induced arthritis. This occurs in part
pathways, the IL-17 family cytokines activate proinflam- due to an altered receptor activator for nuclear factor κ
matory pathways more typical of innate, proinflammatory B ligand (RANKL)–osteoprotegerin (OPG) ratio, and is

You might also like