6 research outputs found

    Selective targeting of immunoproteasome subunit LMP7 prevents colitis-associated carcinogenesis

    Get PDF
    Chronic inflammation is a well-known risk factor for the development of colonic tumorigenesis. In this study, we show that the immunoproteasome (iP) subunit LMP7 plays a crucial role in the progression of colitis-associated carcinogenesis (CAC). The activity and function of the iP complex has been extensively studied in the context of MHC class I-coupled antigen presentation, inflammation and infectious diseases. Here we show that the absence of LMP7 exerts a protective effect, since the LMP7-deficient mice fail to develop a full scale of carcinogenesis after CAC induction with AOM/DSS treatment. Our findings demonstrate that LMP7 deficiency results in reduced protein expression of pro-tumorigenic cytokines IL-6 and TNF-α in the colon after AOM/DSS treatment. Additionally, LMP7-deficient mice also exhibit significantly decreased mRNA levels of pro-tumorigenic chemokines CXCL1, CXCL2 and CXCL3, as well as cell adhesion molecule VCAM-1, thus highlighting the role of LMP7 in regulation of these pro-tumorigenic factors. The net result of the lack of pro-tumorigenic cytokines and chemokines is an impaired recruitment and subsequent activity of tumour-associated neutrophils (TANs) in the colonic lamina propria. Furthermore, we show that the absence or pharmacological inhibition of LMP7 and the consequent blockade of NF-κB, abrogated the production IL-17A, which possesses a potent carcinogenic activity in the gut. Moreover, in vivo administration of a selective LMP7 inhibitor „ONX-0914‟ during CAC induction reduced the tumour incidence in wild-type (WT) mice. Taken together, we identify the iP complex as a crucial mediator of inflammation-driven colon carcinogenesis and we also propose LMP7 as a potential therapeutic target for CAC to limit the ongoing tumorigenesis in inflammatory bowel disease (IBD) patients

    Prevention of colitis-associated cancer by selective targeting of immunoproteasome subunit LMP7

    Get PDF
    Chronic inflammation is a well-known risk factor in development of intestinal tumorigenesis, although the exact mechanisms underlying development of colitis-associated cancer (CAC) still remain obscure. The activity and function of immunoproteasome has been extensively analyzed in the context of inflammation and infectious diseases. Here, we show that the proteasomal immunosubunit LMP7 plays an essential role in development of CAC. Mice devoid of LMP7 were resistant to chronic inflammation and formation of neoplasia, and developed virtually no tumors after AOM/DSS treatment. Our data reveal that LMP7 deficiency resulted in reduced expression of pro-tumorigenic chemokines CXCL1, CXCL2 and CXCL3 as well as adhesion molecule VCAM-1. As a consequence, an impaired recruitment and activity of tumor-infiltrating leukocytes resulting in decreased secretion of cytokines IL-6 and TNF-α was observed. Further, the deletion or pharmacological inhibition of LMP7 and consequent blockade of NF-κB abrogated the production of IL-17A, which possesses a strong carcinogenic activity in the gut. Moreover, in vivo administration of the selective LMP7 inhibitor ONX-0914 led to a marked reduction of tumor numbers in wild-type (WT) mice. Collectively, we identified the immunoproteasome as a crucial mediator of inflammation-driven neoplasia highlighting a novel potential therapeutic approach to limit colonic tumorigenesis

    Selective targeting of immunoproteasome subunit LMP7 prevents colitis-associated carcinogenesis

    No full text
    Chronic inflammation is a well-known risk factor for the development of colonic tumorigenesis. In this study, we show that the immunoproteasome (iP) subunit LMP7 plays a crucial role in the progression of colitis-associated carcinogenesis (CAC). The activity and function of the iP complex has been extensively studied in the context of MHC class I-coupled antigen presentation, inflammation and infectious diseases. Here we show that the absence of LMP7 exerts a protective effect, since the LMP7-deficient mice fail to develop a full scale of carcinogenesis after CAC induction with AOM/DSS treatment. Our findings demonstrate that LMP7 deficiency results in reduced protein expression of pro-tumorigenic cytokines IL-6 and TNF-α in the colon after AOM/DSS treatment. Additionally, LMP7-deficient mice also exhibit significantly decreased mRNA levels of pro-tumorigenic chemokines CXCL1, CXCL2 and CXCL3, as well as cell adhesion molecule VCAM-1, thus highlighting the role of LMP7 in regulation of these pro-tumorigenic factors. The net result of the lack of pro-tumorigenic cytokines and chemokines is an impaired recruitment and subsequent activity of tumour-associated neutrophils (TANs) in the colonic lamina propria. Furthermore, we show that the absence or pharmacological inhibition of LMP7 and the consequent blockade of NF-κB, abrogated the production IL-17A, which possesses a potent carcinogenic activity in the gut. Moreover, in vivo administration of a selective LMP7 inhibitor „ONX-0914‟ during CAC induction reduced the tumour incidence in wild-type (WT) mice. Taken together, we identify the iP complex as a crucial mediator of inflammation-driven colon carcinogenesis and we also propose LMP7 as a potential therapeutic target for CAC to limit the ongoing tumorigenesis in inflammatory bowel disease (IBD) patients

    The microbial metabolite butyrate induces expression of Th1- associated factors in cD4<sup>+</sup> T cells.

    No full text
    Short-chain fatty acids (SCFAs), which are generated by the bacterial fermentation of dietary fibers, promote expansion of regulatory T cells (Tregs). Potential therapeutic value of SCFAs has been recently highlighted in the experimental models of T cell-mediated autoimmunity and allergic inflammation. These studies suggest that physiological intestinal concentrations of SCFAs within the millimolar range are crucial for dampening inflammation-mediated processes. Here, we describe opposing effects of SCFAs on T cell-mediated immune responses. In accordance with published data, lower butyrate concentrations facilitated differentiation of Tregs in vitro and in vivo under steady-state conditions. In contrast, higher concentrations of butyrate induced expression of the transcription factor T-bet in all investigated T cell subsets resulting in IFN-&gamma;-producing Tregs or conventional T cells. This effect was mediated by the inhibition of histone deacetylase activity and was independent of SCFA-receptors FFA2 and FFA3 as well as of Na + - coupled SCFA transporter Slc5a8. Importantly, while butyrate was not able to induce the generation of Tregs in the absence of TGF-&beta;1, the expression of T-bet and IFN-&gamma; was triggered upon stimulation of CD4 + T cells with this SCFA alone. Moreover, the treatment of germ-free mice with butyrate enhanced the expression of T-bet and IFN-&gamma; during acute colitis. Our data reveal that, depending on its concentration and immunological milieu, butyrate may exert either beneficial or detrimental effects on the mucosal immune system
    corecore