9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      TGF-βRII Knock-down in Pancreatic Cancer Cells Promotes Tumor Growth and Gemcitabine Resistance. Importance of STAT3 Phosphorylation on S727

      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

          Pancreatic adenocarcinoma (PDAC) is one of the most deadly cancers in the Western world because of a lack of early diagnostic markers and efficient therapeutics. At the time of diagnosis, more than 80% of patients have metastasis or locally advanced cancer and are therefore not eligible for surgical resection. Pancreatic cancer cells also harbour a high resistance to chemotherapeutic drugs such as gemcitabine that is one of the main palliative treatments for PDAC. Proteins involved in TGF-β signaling pathway (SMAD4 or TGF-βRII) are frequently mutated in PDAC (50–80%). TGF-β signalling pathway plays antagonistic roles during carcinogenesis by initially inhibiting epithelial growth and later promoting the progression of advanced tumors and thus emerged as both tumor suppressor and oncogenic pathways. In order to decipher the role of TGF-β in pancreatic carcinogenesis and chemoresistance, we generated CAPAN-1 and CAPAN-2 cell lines knocked down for TGF-βRII (first actor of TGF-β signaling). The impact on biological properties of these TGF-βRII-KD cells was studied both in vitro and in vivo. We show that TGF-βRII silencing alters tumor growth and migration as well as resistance to gemcitabine. TGF-βRII silencing also leads to S727 STAT3 and S63 c-Jun phosphorylation, decrease of MRP3 and increase of MRP4 ABC transporter expression and induction of a partial EMT phenotype. These markers associated with TGF-β signaling pathways may thus appear as potent therapeutic tools to better treat/manage pancreatic cancer.

          Related collections

          Most cited references30

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

          Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal.

          The cBioPortal for Cancer Genomics (http://cbioportal.org) provides a Web resource for exploring, visualizing, and analyzing multidimensional cancer genomics data. The portal reduces molecular profiling data from cancer tissues and cell lines into readily understandable genetic, epigenetic, gene expression, and proteomic events. The query interface combined with customized data storage enables researchers to interactively explore genetic alterations across samples, genes, and pathways and, when available in the underlying data, to link these to clinical outcomes. The portal provides graphical summaries of gene-level data from multiple platforms, network visualization and analysis, survival analysis, patient-centric queries, and software programmatic access. The intuitive Web interface of the portal makes complex cancer genomics profiles accessible to researchers and clinicians without requiring bioinformatics expertise, thus facilitating biological discoveries. Here, we provide a practical guide to the analysis and visualization features of the cBioPortal for Cancer Genomics.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Integrated Genomic Characterization of Pancreatic Ductal Adenocarcinoma

            (2017)
            We performed integrated genomic, transcriptomic, and proteomic profiling of 150 pancreatic ductal adenocarcinoma (PDAC) specimens, including samples with characteristic low neoplastic cellularity. Deep whole-exome sequencing revealed recurrent somatic mutations in KRAS, TP53, CDKN2A, SMAD4, RNF43, ARID1A, TGFβR2, GNAS, RREB1, and PBRM1. KRAS wild-type tumors harbored alterations in other oncogenic drivers, including GNAS, BRAF, CTNNB1, and additional RAS pathway genes. A subset of tumors harbored multiple KRAS mutations, with some showing evidence of biallelic mutations. Protein profiling identified a favorable prognosis subset with low epithelial-mesenchymal transition and high MTOR pathway scores. Associations of non-coding RNAs with tumor-specific mRNA subtypes were also identified. Our integrated multi-platform analysis reveals a complex molecular landscape of PDAC and provides a roadmap for precision medicine.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              TGF-beta signaling in cancer--a double-edged sword.

              Transforming growth factor (TGF) beta1 is a potent growth inhibitor, with tumor-suppressing activity. Cancers are often refractile to this growth inhibition either because of genetic loss of TGF-beta signaling components or, more commonly, because of downstream perturbation of the signaling pathway, such as by Ras activation. Carcinomas often secrete excess TGF-beta1 and respond to it by enhanced invasion and metastasis. Therapeutic approaches should aim to inhibit the TGF-beta-induced invasive phenotype, but also to retain its growth-inhibitory and apoptosis-inducing effects.
                Bookmark

                Author and article information

                Journal
                Cancers (Basel)
                Cancers (Basel)
                cancers
                Cancers
                MDPI
                2072-6694
                31 July 2018
                August 2018
                : 10
                : 8
                : 254
                Affiliations
                [1 ]INSERM, UMR-S1172, Jean Pierre Aubert Research Center, “Mucins, Epithelial Differentiation and Carcinogenesis” Team, rue Polonovski, 59045 Lille CEDEX, France; v.drubay54@ 123456gmail.com (V.D.); nicolas.skrypek@ 123456gmail.com (N.S.); lucie.cordiez@ 123456gmail.com (L.C.); romain.vasseur@ 123456inserm.fr (R.V.); celine.schulz@ 123456univ-lille.fr (C.S.); nihad.boukrout@ 123456inserm.fr (N.B.); belinda.duchene@ 123456inserm.fr (B.D.); lucie.coppin@ 123456inserm.fr (L.C.); isabelle.vanseuningen@ 123456inserm.fr (I.V.S.)
                [2 ]UMR-S 1172—JPArc, Université Lille Nord de France, 1 Place de Verdun, 59045 Lille CEDEX, France
                [3 ]Centre Hospitalier Régional et Universitaire de Lille, Place de Verdun, 59037 Lille CEDEX, France
                [4 ]CNRS, UMR 8576-UGSF-Unité de Glycobiologie Structurale et Fonctionnelle, Université de Lille, F 59000 Lille, France
                Author notes
                [* ]Correspondence: nicolas.jonckheere@ 123456inserm.fr ; Tel.: +33-3-2029-8850; Fax: +33-3-2053-8562
                [†]

                Authors contributed equally to this manuscript.

                Author information
                https://orcid.org/0000-0001-6072-7963
                https://orcid.org/0000-0002-6596-720X
                https://orcid.org/0000-0002-3131-2694
                https://orcid.org/0000-0002-0496-0661
                Article
                cancers-10-00254
                10.3390/cancers10080254
                6116183
                30065235
                35cf7009-8ab4-4063-85cf-6a7ac20390cf
                © 2018 by the authors.

                Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license ( http://creativecommons.org/licenses/by/4.0/).

                History
                : 10 July 2018
                : 27 July 2018
                Categories
                Article

                tgf-βrii receptor,stat3,metastasis,gemcitabine,abc transporters,pancreas

                Comments

                Comment on this article