Overview of IL-17 Family




(1)
VA Portland Health Care System, Oregon Health & Science University, Portland, Oregon, USA

 




1.1 Discovery and Structure of IL-17


The name of IL-17 was first proposed by Yao et al. [1] in 1995 when they discovered that an open reading frame of the T lymphotropic herpesvirus saimiri gene 13 (HSV13) exhibits 58 % homology with a previously cloned molecule, mouse cytotoxic T lymphocyte-associated protein 8 (CTLA-8) [2]. Recombinant HVS13 and CTLA-8 stimulate transcription factor NFκB activity and IL-6 secretion in fibroblasts and co-stimulate T cell proliferation. A novel cytokine receptor was also isolated and shown to bind both HVS13 and CTLA-8. Therefore, mouse CTLA-8 was named as IL-17, HVS13 as viral IL-17, and the newly cloned cytokine receptor as IL-17 receptor (IL-17R) [1]. Human IL-17 shares 72 % homology with HSV13 and 62 % with mouse IL-17 [3]. Subsequently, by homology-based cloning another 5 cytokines were identified to belong to the IL-17 gene family [4, 5, 6, 7]. The prototypic member IL-17 was renamed as IL-17A and the others as IL-17B through to F (see Table 2.​1) [8].

IL-17E was found to be identical to IL-25, which was described by an independent group [9]. All IL-17 cytokines are homodimeric glycoproteins linked by a disulfide bond except IL-17B, which forms a noncovalent homodimer. All show conservation in their c-terminal region, with five spatially conserved cysteine residues accounting for a characteristic cysteine-knot formation. Of these members, IL-17A and IL-17F are most closely related and share 55 % amino acid sequences. IL-17A and IL-17F also form a heterodimer, IL-17A/F, which exerts similar functions to those of IL-17A and IL-17F (see Table 2.​1) [10, 11, 12]. Homology with IL-17A for IL-17B, IL-17D, and IL-17C is in the order of 29 %, 25 %, and 23 %, respectively, and IL-17E is most distant and with only 19 % homology with IL-17A [8].


1.2 IL-17 Receptors and Signaling


IL-17 receptor (IL-17R) is also a unique cytokine receptor family [1] with five members identified to date, namely, IL-17RA, IL-17RB, IL-17RC, IL-17RD, and IL-17RE (see Table 2.​1) [13]. All these IL-17 receptors contain extracellular domains composed of fibronectin type III (FnIII) domains and cytoplasmic similar expression to fibroblast growth factor (SEF) genes–IL-17R (SEFIR) domains that are loosely homologous to Toll/IL-1R domains [14, 15]. Several studies have shown that IL-17RA serves as a common receptor to mediate signals for several members of the IL-17 family (Fig. 1.1) [16, 17, 18, 19, 20, 21, 22, 23].

A393433_1_En_1_Fig1_HTML.gif


Figure 1.1
IL-17 receptors. The IL-17R family comprises five members: IL-17RA, IL-17RB, IL-17RC, IL-17RD, and IL-17RE. IL-17RA is a common receptor that forms heterodimers with other IL-17Rs for ligand binding and signal transduction: IL-17RA-RC for IL-17A, IL-17F, and IL-17A/F; IL-17RA-RE for IL-17C; IL-17RA-RB for IL-17E (IL-25). It is not clear whether IL-17RB also forms a homodimer for IL-17B; IL-17RD remains an orphan receptor, but it regulates IL-17A signaling and forms TNFR2–IL-17RD receptor complex. The receptor for IL-17D has not been identified. IL-17RA-RC, IL-17RA-RB, and IL-17RA-RE transduce signal via adaptor molecule Act1. Act1 NFκB activator 1 (also known as CIKS connection to IKK and SAPK/JNK), IL-17RA IL-17 receptor adaptor, C/EBPs CCAAT-enhancer-binding proteins, MAPKs mitogen-activated protein kinases, NFκB nuclear factor κB, TRAF tumor necrosis factor-associated factor

Indeed, the major IL-17RA interacting residues are identical among all IL-17 cytokines [24]. But IL-17RA does not bind all ligands with an equal affinity. IL-17RA has high affinity for IL-17A, about hundredfold weaker affinity for IL-17F, an intermediate affinity for IL-17A/F, and much weaker affinities for IL-17B, IL-17C, IL-17D, and IL-17E [4, 5, 16, 17, 19, 22, 25]. Interestingly, IL-17RA pairs with other IL-17Rs to form functional heterodimeric receptor complexes. This is apparently determined by the ligands, IL-17 cytokines. For example, the homodimeric IL-17A or IL-17F disfavors binding of a second molecule of IL-17RA, but selects IL-17RC to form IL-17RA-RC complex (Fig. 1.1) [24]. Similarly, other IL-17 cytokines bind their primary receptors and recruit IL-17RA to form a functional heterodimeric receptor complex. IL-17A and IL-17F and IL-17A/F bind to receptor complex IL-17RA-RC, IL-17C to IL-17RA-RE, and IL-17E (IL-25) to IL-17RA-RB (Fig. 1.1) [13]. The redundant activities of IL-17A and IL-17F are partially attributed to their binding to the same IL-17RA-RC receptor complex, and yet the distinct activities of the two cytokines may be due to the different binding affinities to the receptor complex [26]. Both components of the heterodimeric receptor complex are required for signaling. For instance, deficiency of either IL-17RA or IL-17RC can completely abolish the inflammatory function of IL-17A and IL-17F [12, 21, 27, 28].

The proinflammatory and host defense effects of IL-17A and IL-17F are executed chiefly by activation of NFκB and MAPK pathways. A signal from IL-17R is first relayed by a cytosolic protein, signaling adaptor, called Act1 (NFκB activator 1, also known as CIKS) (Fig. 1.2) [29].

A393433_1_En_1_Fig2_HTML.gif


Figure 1.2
Structure of Act1. Act1 is IL-17R adaptor which relays IL-17 cytokine–IL-17R signaling. It contains a SEFIR domain that binds cytoplasmic SEFIR domain of IL-17Rs. A decoy peptide correspondence of the sequence of CC’ loop of Act1 SEFIR domain can block the IL-17A and IL-17E (IL-25) signaling, indicating interruption of interaction between Act1 and IL-17R is potentially therapeutic. C/EBP CCAAT-enhancer-binding proteins, HLH helix–loop–helix, MAPKs mitogen-activated protein kinases, NFκB nuclear factor κB, SEFIR similar expression to fibroblast growth factor genes and IL-17R, TRAF tumor necrosis factor-associated factor

Act1 is essential for IL-17 signal transduction. Deficiency of Act1 results in a loss of IL-17-dependent NFκB activation and proinflammatory cytokine production [29, 30]. Upon ligand stimulation of IL-17R, Act1 is recruited to the IL-17R complex. Act1 also contains a SEFIR domain, which binds to the SEFIR region of IL-17R through homotypic interactions [13, 31]. The activated Act1 then binds tumor necrosis factor-associated factor (TRAF)-6 via a TRAF binding domains of Act1 (Figs. 1.1 and 1.2). TRAF6 mediates further downstream cascade interactions to lead to activation of NFκB, MAPK, and C/EBP pathways and the activation of target genes [13].

The importance of Act1 in autoimmunity and inflammatory diseases has been demonstrated in animal models of inflammatory diseases. For instance, mice with deficiency of Act1 have reduced demyelination in a cuprizone-induced multiple sclerosis (MS) model, which is known to be mediated by IL-17 signaling [32]. Act1-deficient mice are protected from collagen-induced arthritis (CIA) [33]. These data suggest that Act1 is a candidate target for therapeutic intervention in human inflammatory diseases.

As in other cytokine signaling, IL-17/IL-17R signaling is also finely regulated. Many regulators have been described to involve positive and negative regulation of the IL-17/IL-17R signaling pathway at various stages [13]. Modulation of these regulatory factors is potentially of therapeutic benefit. Soluble IL-17RA, IL-17RB, and IL-17RC naturally exist and may function as decoy receptor to regulate IL-17–IL-17R signaling in vivo. Indeed, fused with immunoglobulin G (IgG) Fc fragment, soluble IL-17RA (IL-17RA-Fc) is able to suppress IL-17A signaling in cultured cells; IL-17RC-Fc suppresses the effect of IL-17A and IL-17F [27], and IL-17RB-Fc is able to block IL-17E signaling [34]. These soluble receptor-Fc fusion proteins are highly likely to be evaluated as therapeutic agents (Fig. 5.1b).

Signaling through the IL-17R complex can influence function of other IL-17 cytokines. Chang et al. [17] observed that IL-17C is highly expressed in the central nervous system of mice with experimental autoimmune encephalomyelitis (EAE). IL-17C knockout mice exhibited a striking reduction of incidence and severity of EAE along with reduction of Th17 cells [17]. IL-17C signaling through IL-17RA-RE [17, 18, 19] further increases IL-17RE expression on Th17 cells and increases IL-17A, IL-17F, and IL-22 production. Skin keratinocytes and intestine epithelial cells constitutively express IL-17RE and can respond to IL-17C [18, 19]. These findings indicate that IL-17C may be another target for therapy in Th17-mediated inflammatory diseases, but caution needs to be taken for its protective effect in intestines.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Jan 18, 2018 | Posted by in RHEUMATOLOGY | Comments Off on Overview of IL-17 Family

Full access? Get Clinical Tree

Get Clinical Tree app for offline access