Several inhibitors have been developed to interfere with NF-B activation at different levels

Several inhibitors have been developed to interfere with NF-B activation at different levels. not been fully exploited in the context of IL-23-mediated diseases. In this review, we discuss the current knowledge about proximal signaling events triggered by IL-23 upon binding to its membrane receptor to bring to the spotlight new opportunities for therapeutic intervention in IL-23-mediated pathologies. [32,33], and it induces expression of genes regulating proliferation, wound healing, and apoptosis of intestinal epithelial cells [34]. In addition to its role in host defense, IL-22 provides functional barrier support through induction of cell proliferation, mucins, and antimicrobial peptides [35]. In fact, the interference with the IL-22/IL-22R pathway exacerbated colitis in some mouse models [36,37]. Thus, as for IL-17, both pro-inflammatory and tissue-protective functions have been identified for IL-22. Interestingly, the role in intestinal homeostasis of Th17-derived IL-17 and IL-22 are independent of IL-23 [23,24,38], and thus, the development of selective IL-23 inhibitors hold the promise to interfere especially with pathogenic IL-17-producing cells without affecting maintenance of the gut barrier. GM-CSF has emerged as the key pathogenic effector molecule downstream of IL-23 in the development Molindone hydrochloride of the experimental autoimmune encephalomyelitis (EAE) model of multiple sclerosis [7,8]. GM-CSF is secreted as a monomeric cytokine that binds to the GM-CSF receptor, a heterodimer formed by a specific subunit and a common beta (c) Molindone hydrochloride subunit shared with IL-3 and IL-5 receptors. GM-CSF binding to its cognate receptor promotes the activation of Jak2 and subsequent STAT5 phosphorylation, Src family kinases, and the phosphatidylinositol 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) pathways. The main GM-CSF responder populations are dendritic cells, monocytes, macrophages, granulocytes, neutrophils, and importantly, microglia and astrocytes [39,40]. Despite its initial classification as a hematopoietic growth factor, GM-CSF plays a minor role in myelopoiesis, and it is emerging as a major mediator of tissue inflammation. GM-CSF induces a genetic program involved in inflammasome function, phagocytosis and chemotaxis that participate in tissue destruction and demyelination Rabbit Polyclonal to ARTS-1 [41]. GM-CSF promotes monocyte migration from the bone marrow across the hematoencephalic barrier and into the central nervous system (CNS) [42]. Once at the CNS, GM-CSF promotes the differentiation of infiltrating monocytes into antigen presenting cells that contribute to the maintenance of the pathogenic Th17 cells [43] and also induces production of pro-inflammatory mediators that promote tissue damage, demyelination, and axonal loss [44]. Finally, although less studied than IL-17, IL-22, and GM-CSF, IL-23 also induces the production of TNF, IL-19, and IL-24 in a skin inflammation model [9]. IL-23 is required to provide effective host defense against a wide variety of extracellular pathogens, such as bacteria, parasites, fungi, and viruses Molindone hydrochloride [1]. However, due to their pivotal role in inflammatory diseases, IL-23 and its downstream effector molecules have emerged as attractive therapeutic targets. The emergence of neutralizing antibodies against harmful pro-inflammatory mediators has marked a milestone in the development of new therapeutic strategies. In this context, blocking antibodies against IL-23 and IL-17 have been approved for treatment of plaque psoriasis, and they are currently under Phase II/Phase III clinical trials for inflammatory bowel diseases, multiple sclerosis, and rheumatoid arthritis [1]. Therapeutic interventions using blocking antibodies in the context of IL-23-mediated diseases have been recently and extensively reviewed elsewhere [2,11,45,46,47]. Despite the success of monoclonal antibodies, not all patients respond to these treatments, and others show a partial response. Thus, effective therapies for chronic inflammatory diseases may require the combination of multiple immune-modulatory drugs to prevent disease progression and to improve quality of life. Alternative strategies aimed at inhibiting intracellular signaling cascades using small molecule inhibitors or interfering peptides have not been fully exploited in the context of IL-23-mediated diseases. The interference with intracellular signaling cascades has been successfully applied for the treatment of different types of cancer and inflammatory pathologies [48,49]. In comparison to monoclonal antibodies, small molecule inhibitors have a broader tissue distribution, possibility of development of oral/topical versions, and reduced production costs [50]. These therapies are effective, economic, and thus, suitable for mild clinical symptoms or to be used in combination with monoclonal antibodies therapies. In addition, engineered, non-immunoglobulin protein scaffolds that interfere with IL-23 or the IL-23R represent another therapeutic strategy for treatment of chronic inflammatory diseases. Protein scaffolds are Molindone hydrochloride based in natural proteins and use combinatorial protein engineering to change their affinity and specificity to bind.