Suppressors of cytokine signaling (SOCS) proteins are negative-feedback regulators of JAK/STAT

Suppressors of cytokine signaling (SOCS) proteins are negative-feedback regulators of JAK/STAT pathway and SOCS3 contributes to host immunity by regulating the intensity/period of cytokine signals and inflammatory responses. the T cell-mediated autoimmune disease, experimental autoimmune uveitis (EAU). In contrast to the exacerbation of EAE in myeloid-specific SOCS3-deleted mice, Y-27632 2HCl CD4-SOCS3KO mice were guarded from acute and chronic uveitis. Protection from EAU correlated with enhanced manifestation Y-27632 2HCl of CTLA4 and growth of IL-10 generating Tregs with augmented suppressive activities. We further show that SOCS3 interacts with CTLA4 and negatively regulates CTLA4 levels in T cells, providing mechanistic explanation for the growth of Tregs in CD4-SOCS3 during EAU. Contrary to epigenetic studies, Th17/IFN- Y-27632 2HCl and Tc17/IFN- populations were markedly reduced in CD4-SOCS3KO, suggesting that SOCS3 promotes growth of Th17/IFN- subset associated with development of severe uveitis. Thus, SOCS3 is usually a potential therapeutic target in uveitis and other auto-inflammatory diseases. Introduction The JAK/STAT pathway is usually an evolutionarily conserved transmission transduction mechanism that regulates a myriad of physiological processes in mammals (1). The importance of regulating the initiation, duration and intensity of STAT signals is usually underscored by the diverse array of pathologic conditions that arise from disruption or aberrant activation of STATs (2). JAK/STAT pathways are therefore under stringent rules by a number of cytoplasmic proteins including PIAS (protein inhibitors of activated STAT), SHP-1 (SH2-made up of phosphatase 1), Dispatch-2 and suppressors of cytokine signaling (SOCS) family of proteins. In context of immune rules or immune modulation therapy, much interest has focused on SOCS protein, particularly SOCS1 and SOCS3 (3, 4). SOCS protein are rapidly induced in response to cytokines (IFN-, Itga2 IL-2, IL-4, IL-6, IL-10, IL-12, IL-21, IL-23, IL-27) or growth factors (CNTF, LIF, FGF, IGF-1, insulin) and their inhibitory effects derive from direct conversation with cytokine/growth-factor receptors or signaling protein, leading to proteosomal degradation of the receptor complex and termination of the transmission (5). Because of the relatively short half-life of SOCS proteins, their unfavorable regulatory effects are generally transient. However, unabated activation of STAT signaling pathway by chronic inflammation can induce constitutive activation of SOCS manifestation (6). In some tissues this may result in prolonged silencing of crucial cellular pathways and pre-disposition to development of organ-specific diseases (7). SOCS protein have now been implicated in diverse diseases including, autoimmune diseases, diabetes, and malignancy (6C9). SOCS3 regulates the activation and differentiation of na?vat the CD4 T cells, preferentially promoting Th2 and inhibiting Th1 differentiation via the inhibition of IL-12-mediated STAT4 activation (10, 11). It is usually constitutively expressed in na?ve CD4+ T cells and its expression is inversely correlated with the level of IL-2 (11, 12). SOCS3 mediates the IL-27-induced suppression of CD28-mediated IL-2 production (13) and it blocks IL-2 production in response to TCR activation by suppressing calcineurin-dependent dephosphorylation and activation of NFATp (14). Unlike T-helper cells, Tregs are deficient in SOCS3 protein expression and over-expression of SOCS3 in Treg decreased their proliferation and expression of Foxp3, suggesting Y-27632 2HCl the SOCS3/IL-2 axis plays critical role in controlling physiological levels of Tregs. In terms of the potential involvement of SOCS3 in autoimmune diseases, it has recently been shown that mice with deletion in myeloid cells develop severe EAE, suggesting that STAT3/SOCS3 axis regulates neuroinflammation (15, 16). On the other hand, expression of SOCS3 in human arthritic chondrocytes contributes to cartilage damage during arthritis (17, 18). Interestingly, epigenetic suppression of SOCS3 expression in T cells promotes the expansion of a unique Tc17/IFN–double producing CD8+ T cells implicated in several autoimmune diseases (19C22). These observations thus underscore the complexity of SOCS3 functions in the immune system and mechanisms that regulate autoimmune pathology. Intraocular inflammation or uveitis is a major cause of severe visual handicap and includes sight-threatening diseases such as Behcet disease, birdshot retinochoroidopathy, Vogt-Koyanagi-Harada, sympathetic ophthalmia and ocular sarcoidosis (23). Although the cause of most chronic ocular inflammatory diseases of non-infectious etiology is largely unknown, studies in the mouse indicate that it is predominantly a T cell mediated disease. Experimental autoimmune uveoretinitis (EAU) shares essential immunopathologic features with human uveitis and is the animal model of human uveitis (24). The disease is characterized by massive infiltration Y-27632 2HCl of inflammatory cells into the retina, destruction of retinal architecture and Th1 and Th17 cells were thought to be the etiologic agents.