H1 cells have long been considered the principal mediators of disease development. More recently, a role for Th17 cells in psoriasis has been recognized. Th17 cytokines, including IL-17A, IL-17F, and IL-22, are found at higher levels in psoriatic skin lesions than in non-psoriatic and normal skin [3,4]. Additionally, IL-23, a Th17 growth and differentiation factor and its receptor are increased in psoriatic lesions [4,5,6]. Moreover, injection of wild-type (WT) mice 1655472 with IL-23 reproduces several aspects of disease, including epidermal acanthosis, hyperkeratosis and a mixed dermal inflammatory infiltrate that includes mononuclear cells and granulocytes he majority of which are neutrophils [7,8,9]. MedChemExpress Dimethylenastron Finally, recent clinical data demonstrate a critical role for Th17 cytokines. Immunotherapies using antibodies targeting IL-17 [10,11,12] or IL-12/IL-23 [13,14,15,16] are effective psoriasis treatments. Several data suggest that chemokines and their receptors regulate the pathogenesis of inflammatory diseases, including psoriasis by regulating the recruitment of leukocytes into affectedtissues. Th17 cells express the chemokine receptor, CCR6 [8,17,18,19,20], and recent studies demonstrate that the CCR6 ligand, CCL20 is up-regulated in psoriatic plaques [18,21]. The finding that CCR6-deficient mice fail to develop psoriasiform pathology following intradermal injection with IL-23 supports a critical role for CCR6 in this inflammatory skin disorder [9]. The expression of many other chemokines within psoriatic lesions suggests that additional chemokine-driven mechanisms may also regulate disease development. CCR2 has been implicated in the pathogenesis of several inflammatory diseases, and CCR2 antagonists have been developed. CCR2 is expressed on activated T cells ncluding Th17 cells [22,23], as well as monocytes, macrophages, immature dendritic cells, cd T cells and NK cells [24]. CCR2 binds multiple murine chemokine ligands: CCL2 (MCP-1), CCL7 (MCP-3) and CCL12 (MCP-5) [25]. CCL2 is expressed at high levels in psoriatic plaques by keratinocytes [26,27], suggesting a potential role for CCR2 in psoriasis pathogenesis. A requirement for CCR2 in the development of Th17-mediated autoimmune inflammation has been demonstrated [28,29]; EAE disease pathology in CCR2deficient (CCR22/2) mice is ameliorated. Protection from EAE is associated with a decreased IFN-c response [28], although IL-17 and IL-22 cytokine production was not measured in these studies. In contrast, in a mouse model of collagen-induced arthritis, disease severity was exacerbated in CCR22/2 mice, and this correlatedIL-23 Induces Th2 Inflammation in CCR22/2 Micewith an increased Th17 response [30]. Thus, depending on the disease model, CCR2-deficiency may have an inflammatory or anti-inflammatory effect. Recent studies have demonstrated that skewing CD4+ T cell phenotype within psoriatic plaques to a Th2-type immune response can ameliorate disease [31,32,33]. Treatment of psoriasis patients with subcutaneous injections of IL-4 polarizes lesional T cell responses to a Th2-type and 1317923 decreases psoriasis severity [31]. Similarly, transdermal delivery of IL-4 expression plasmid ameliorates disease in a mouse model of psoriasis [32,33]. Thus, induction of a Th2 phenotype of skin infiltrating lymphocytes correlates with disease improvement. In several Bromopyruvic acid chemical information models of inflammation, CCR2 blockade blunts Th1-type immune responses and enhances Th2-type immune responses. Studies using mouse models of infect.H1 cells have long been considered the principal mediators of disease development. More recently, a role for Th17 cells in psoriasis has been recognized. Th17 cytokines, including IL-17A, IL-17F, and IL-22, are found at higher levels in psoriatic skin lesions than in non-psoriatic and normal skin [3,4]. Additionally, IL-23, a Th17 growth and differentiation factor and its receptor are increased in psoriatic lesions [4,5,6]. Moreover, injection of wild-type (WT) mice 1655472 with IL-23 reproduces several aspects of disease, including epidermal acanthosis, hyperkeratosis and a mixed dermal inflammatory infiltrate that includes mononuclear cells and granulocytes he majority of which are neutrophils [7,8,9]. Finally, recent clinical data demonstrate a critical role for Th17 cytokines. Immunotherapies using antibodies targeting IL-17 [10,11,12] or IL-12/IL-23 [13,14,15,16] are effective psoriasis treatments. Several data suggest that chemokines and their receptors regulate the pathogenesis of inflammatory diseases, including psoriasis by regulating the recruitment of leukocytes into affectedtissues. Th17 cells express the chemokine receptor, CCR6 [8,17,18,19,20], and recent studies demonstrate that the CCR6 ligand, CCL20 is up-regulated in psoriatic plaques [18,21]. The finding that CCR6-deficient mice fail to develop psoriasiform pathology following intradermal injection with IL-23 supports a critical role for CCR6 in this inflammatory skin disorder [9]. The expression of many other chemokines within psoriatic lesions suggests that additional chemokine-driven mechanisms may also regulate disease development. CCR2 has been implicated in the pathogenesis of several inflammatory diseases, and CCR2 antagonists have been developed. CCR2 is expressed on activated T cells ncluding Th17 cells [22,23], as well as monocytes, macrophages, immature dendritic cells, cd T cells and NK cells [24]. CCR2 binds multiple murine chemokine ligands: CCL2 (MCP-1), CCL7 (MCP-3) and CCL12 (MCP-5) [25]. CCL2 is expressed at high levels in psoriatic plaques by keratinocytes [26,27], suggesting a potential role for CCR2 in psoriasis pathogenesis. A requirement for CCR2 in the development of Th17-mediated autoimmune inflammation has been demonstrated [28,29]; EAE disease pathology in CCR2deficient (CCR22/2) mice is ameliorated. Protection from EAE is associated with a decreased IFN-c response [28], although IL-17 and IL-22 cytokine production was not measured in these studies. In contrast, in a mouse model of collagen-induced arthritis, disease severity was exacerbated in CCR22/2 mice, and this correlatedIL-23 Induces Th2 Inflammation in CCR22/2 Micewith an increased Th17 response [30]. Thus, depending on the disease model, CCR2-deficiency may have an inflammatory or anti-inflammatory effect. Recent studies have demonstrated that skewing CD4+ T cell phenotype within psoriatic plaques to a Th2-type immune response can ameliorate disease [31,32,33]. Treatment of psoriasis patients with subcutaneous injections of IL-4 polarizes lesional T cell responses to a Th2-type and 1317923 decreases psoriasis severity [31]. Similarly, transdermal delivery of IL-4 expression plasmid ameliorates disease in a mouse model of psoriasis [32,33]. Thus, induction of a Th2 phenotype of skin infiltrating lymphocytes correlates with disease improvement. In several models of inflammation, CCR2 blockade blunts Th1-type immune responses and enhances Th2-type immune responses. Studies using mouse models of infect.