17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      All-Trans Retinoic Acid Modulates TLR4/NF- κB Signaling Pathway Targeting TNF- α and Nitric Oxide Synthase 2 Expression in Colonic Mucosa during Ulcerative Colitis and Colitis Associated Cancer

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Colitis associated cancer (CAC) is the colorectal cancer (CRC) subtype that is associated with bowel disease such as ulcerative colitis (UC). The data on role of NF- κB signaling in development and progression of CAC were derived from preclinical studies, whereas data from human are rare. The aim of this work was to study the contribution of NF- κB pathway during UC and CAC, as well as the immunomodulatory effect of all-trans retinoic acid (AtRA). We analyzed the expression of NOS2, TNF- α, TLR4, and NF- κB, in colonic mucosa. We also studied NO/TNF- α modulation by LPS in colonic mucosa pretreated with AtRA. A marked increase in TLR4, NF- κB, TNF- α, and NOS2 expression was reported in colonic mucosa. The relationship between LPS/TLR4 and TNF- α/NO production, as well as the role of NF- κB signaling, was confirmed by ex vivo experiments and the role of LPS/TLR4 in NOS2/TNF- α induction through NF- κB pathway was suggested. AtRA downregulates NOS2 and TNF- α expression. Collectively, our study indicates that AtRA modulates in situ LPS/TLR4/NF- κB signaling pathway targeting NOS2 and TNF- α expression. Therefore, we suggest that AtRA has a potential value in new strategies to improve the current therapy, as well as in the clinical prevention of CAC development and progression.

          Related collections

          Most cited references76

          • Record: found
          • Abstract: found
          • Article: not found

          LPS-TLR4 Signaling to IRF-3/7 and NF-κB Involves the Toll Adapters TRAM and TRIF

          Toll–IL-1–resistance (TIR) domain–containing adaptor-inducing IFN-β (TRIF)–related adaptor molecule (TRAM) is the fourth TIR domain–containing adaptor protein to be described that participates in Toll receptor signaling. Like TRIF, TRAM activates interferon regulatory factor (IRF)-3, IRF-7, and NF-κB-dependent signaling pathways. Toll-like receptor (TLR)3 and 4 activate these pathways to induce IFN-α/β, regulated on activation, normal T cell expressed and secreted (RANTES), and γ interferon–inducible protein 10 (IP-10) expression independently of the adaptor protein myeloid differentiation factor 88 (MyD88). Dominant negative and siRNA studies performed here demonstrate that TRIF functions downstream of both the TLR3 (dsRNA) and TLR4 (LPS) signaling pathways, whereas the function of TRAM is restricted to the TLR4 pathway. TRAM interacts with TRIF, MyD88 adaptor–like protein (Mal)/TIRAP, and TLR4 but not with TLR3. These studies suggest that TRIF and TRAM both function in LPS-TLR4 signaling to regulate the MyD88-independent pathway during the innate immune response to LPS.
            Bookmark
            • Record: found
            • Abstract: not found
            • Article: not found

            Nitric oxide synthases in mammals.

              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Blocking TNF-alpha in mice reduces colorectal carcinogenesis associated with chronic colitis.

              The inflammatory bowel disease ulcerative colitis (UC) frequently progresses to colon cancer. To understand the mechanisms by which UC patients develop colon carcinomas, we used a mouse model of the disease whereby administration of azoxymethane (AOM) followed by repeated dextran sulfate sodium (DSS) ingestion causes severe colonic inflammation and the subsequent development of multiple tumors. We found that treating WT mice with AOM and DSS increased TNF-alpha expression and the number of infiltrating leukocytes expressing its major receptor, p55 (TNF-Rp55), in the lamina propria and submucosal regions of the colon. This was followed by the development of multiple colonic tumors. Mice lacking TNF-Rp55 and treated with AOM and DSS showed reduced mucosal damage, reduced infiltration of macrophages and neutrophils, and attenuated subsequent tumor formation. WT mice transplanted with TNF-Rp55-deficient bone marrow also developed significantly fewer tumors after AOM and DSS treatment than either WT mice or TNF-Rp55-deficient mice transplanted with WT bone marrow. Furthermore, administration of etanercept, a specific antagonist of TNF-alpha, to WT mice after treatment with AOM and DSS markedly reduced the number and size of tumors and reduced colonic infiltration by neutrophils and macrophages. These observations identify TNF-alpha as a crucial mediator of the initiation and progression of colitis-associated colon carcinogenesis and suggest that targeting TNF-alpha may be useful in treating colon cancer in individuals with UC.
                Bookmark

                Author and article information

                Journal
                Mediators Inflamm
                Mediators Inflamm
                MI
                Mediators of Inflammation
                Hindawi
                0962-9351
                1466-1861
                2017
                20 March 2017
                : 2017
                : 7353252
                Affiliations
                1Team: Cytokines and NO Synthases-Immunity and Pathogenesis, Laboratory of Cellular and Molecular Biology (LBCM), Faculty of Biological Science, University of Sciences and Technology (USTHB), Algiers, Algeria
                2Institut de Biologie de Lille, UMR 8161, CNRS, Institut Pasteur de Lille, Université Lille-Nord de France, Lille, France
                3Anatomic Pathology Service, Mustapha Pacha Hospital, Algiers, Algeria
                4Department of Gastroenterology, Maillot Hospital, Algiers, Algeria
                5Service of Oncology, Rouiba Hospital, Algiers, Algeria
                Author notes
                *Nadira Delhem: nadira.delhem@ 123456ibl.fr and
                *Chafia Touil-Boukoffa: touilboukoffa@ 123456yahoo.fr

                Academic Editor: Kumar S. Bishnupuri

                Author information
                http://orcid.org/0000-0002-2135-3265
                http://orcid.org/0000-0001-5335-4222
                http://orcid.org/0000-0001-7728-2147
                Article
                10.1155/2017/7353252
                5376956
                28408791
                3d343f42-84a7-47db-947f-2b2efc9ad61e
                Copyright © 2017 Hayet Rafa et al.

                This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

                History
                : 5 November 2016
                : 5 January 2017
                : 19 February 2017
                Funding
                Funded by: Agence Thématique de Recherche en Sciences de la Santé
                Funded by: Agence Universitaire de la Francophonie
                Funded by: SIRIC ONCOLille
                Categories
                Research Article

                Immunology
                Immunology

                Comments

                Comment on this article