19
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Inhibition of Nox4‐dependent ROS signaling attenuates prostate fibroblast activation and abrogates stromal‐mediated protumorigenic interactions

      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

          Carcinoma‐associated fibroblasts (CAFs) play a key onco‐supportive role during prostate cancer (PCa) development and progression. We previously reported that the reactive oxygen species (ROS)‐producing enzyme NADPH oxidase 4 (Nox4) is essential for TGFβ1‐mediated activation of primary prostate human fibroblasts to a CAF‐like phenotype. This study aimed to further investigate the functional relevance of prostatic Nox4 and determine whether pharmacological inhibition of stromal Nox4 abrogates paracrine‐mediated PCa‐relevant processes. RNA in situ hybridization revealed significantly elevated Nox4 mRNA levels predominantly in the peri‐tumoral stroma of clinical PCa with intense stromal Nox4 staining adjacent to tumor foci expressing abundant TGFβ protein levels. At pharmacologically relevant concentrations, the Nox1/Nox4 inhibitor GKT137831 attenuated ROS production, CAF‐associated marker expression and migration of TGFβ1‐activated but not nonactivated primary human prostate fibroblasts. Similar effects were obtained upon shRNA‐mediated silencing of Nox4 but not Nox1 indicating that GKT137831 primarily abrogates TGFβ1‐driven fibroblast activation via Nox4 inhibition. Moreover, inhibiting stromal Nox4 abrogated the enhanced proliferation and migration of PCa cell lines induced by TGFβ1‐activated prostate fibroblast conditioned media. These effects were not restricted to recombinant TGFβ1 as conditioned media from PCa cell lines endogenously secreting high TGFβ1 levels induced fibroblast activation in a stromal Nox4‐ and TGFβ receptor‐dependent manner. Importantly, GKT137831 also attenuated PCa cell‐driven fibroblast activation. Collectively, these findings suggest the TGFβ‐Nox4 signaling axis is a key interface to dysregulated reciprocal stromal–epithelial interactions in PCa pathophysiology and provide a strong rationale for further investigating the applicability of Nox4 inhibition as a stromal‐targeted approach to complement current PCa treatment modalities.

          Abstract

          What's new?

          While the tumor microenvironment plays a critical role in prostate adenocarcinoma pathogenesis, underlying mechanisms remain unclear. This study demonstrates the functional relevance of elevated expression of Nox4—which is a major source of cellular reactive oxygen species (ROS)—in the tumor microenvironment and downstream consequences on prostate cancer (PCa)‐relevant processes. Findings identify stromal Nox4 as a central mediator of reciprocal epithelial‐stromal cell crosstalk, fibroblast activation and stromal‐driven tumor (cell)‐promoting hallmarks. The data thus provide a strong mechanistic rationale for therapeutic inhibition of Nox4 as a stromal‐targeted approach to complement current treatment strategies for PCa and restore dysregulated reciprocal stromal–epithelial interactions.

          Related collections

          Most cited references35

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

          Functional analysis of Nox4 reveals unique characteristics compared to other NADPH oxidases.

          Reactive oxygen species (ROS) are important signal transduction molecules in ligand-induced signaling, regulation of cell growth, differentiation, apoptosis and motility. Recently NADPH oxidases (Nox) homologous to Nox2 (gp91phox) of phagocyte cytochrome b558 have been identified, which are an enzymatic source for ROS generation in epithelial cells. This study was undertaken to delineate the requirements for ROS generation by Nox4. Nox4, in contrast to other Nox proteins, produces large amounts of hydrogen peroxide constitutively. Known cytosolic oxidase proteins or the GTPase Rac are not required for this activity. Nox4 associates with the protein p22phox on internal membranes, where ROS generation occurs. Knockdown and gene transfection studies confirmed that Nox4 requires p22phox for ROS generation. Mutational analysis revealed structural requirements affecting expression of the p22phox protein and Nox activity. Mechanistic insight into ROS regulation is significant for understanding fundamental cell biology and pathophysiological conditions.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Reactive stroma in human prostate cancer: induction of myofibroblast phenotype and extracellular matrix remodeling.

            Generation of a reactive stroma environment occurs in many human cancers and is likely to promote tumorigenesis. However, reactive stroma in human prostate cancer has not been defined. We examined stromal cell phenotype and expression of extracellular matrix components in an effort to define the reactive stroma environment and to determine its ontogeny during prostate cancer progression. Normal prostate, prostatic intraepithelial neoplasia (PIN), and prostate cancer were examined by immunohistochemistry. Tissue samples included radical prostatectomy specimens, frozen biopsy specimens, and a prostate cancer tissue microarray. A human prostate stromal cell line was used to determine whether transforming growth factor beta1 (TGF-beta1) regulates reactive stroma. Compared with normal prostate tissue, reactive stroma in Gleason 3 prostate cancer showed increased vimentin staining and decreased calponin staining (P < 0.001). Double-label immunohistochemistry revealed that reactive stromal cells were vimentin and smooth muscle alpha-actin positive, indicating the myofibroblast phenotype. In addition, reactive stroma cells exhibited elevated collagen I synthesis and expression of tenascin and fibroblast activation protein. Increased vimentin expression and collagen I synthesis were first observed in activated periacinar fibroblasts adjacent to PIN. Similar to previous observations in prostate cancer, TGF-beta1-staining intensity was elevated in PIN. In vitro, TGF-beta1 stimulated human prostatic fibroblasts to switch to the myofibroblast phenotype and to express tenascin. The stromal microenvironment in human prostate cancer is altered compared with normal stroma and exhibits features of a wound repair stroma. Reactive stroma is composed of myofibroblasts and fibroblasts stimulated to express extracellular matrix components. Reactive stroma appears to be initiated during PIN and evolve with cancer progression to effectively displace the normal fibromuscular stroma. These studies and others suggest that TGF-beta1 is a candidate regulator of reactive stroma during prostate cancer progression.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Role of the TMPRSS2-ERG gene fusion in prostate cancer.

              TMPRSS2-ERG gene fusions are the predominant molecular subtype of prostate cancer. Here, we explored the role of TMPRSS2-ERG gene fusion product using in vitro and in vivo model systems. Transgenic mice expressing the ERG gene fusion product under androgen-regulation develop mouse prostatic intraepithelial neoplasia (PIN), a precursor lesion of prostate cancer. Introduction of the ERG gene fusion product into primary or immortalized benign prostate epithelial cells induced an invasion-associated transcriptional program but did not increase cellular proliferation or anchorage-independent growth. These results suggest that TMPRSS2-ERG may not be sufficient for transformation in the absence of secondary molecular lesions. Transcriptional profiling of ERG knockdown in the TMPPRSS2-ERG-positive prostate cancer cell line VCaP revealed decreased expression of genes over-expressed in prostate cancer versus PIN and genes overexpressed in ETS-positive versus -negative prostate cancers in addition to inhibiting invasion. ERG knockdown in VCaP cells also induced a transcriptional program consistent with prostate differentiation. Importantly, VCaP cells and benign prostate cells overexpressing ERG directly engage components of the plasminogen activation pathway to mediate cellular invasion, potentially representing a downstream ETS target susceptible to therapeutic intervention. Our results support previous work suggesting that TMPRSS2-ERG fusions mediate invasion, consistent with the defining histologic distinction between PIN and prostate cancer.
                Bookmark

                Author and article information

                Contributors
                natalie.sampson@i-med.ac.at
                Journal
                Int J Cancer
                Int. J. Cancer
                10.1002/(ISSN)1097-0215
                IJC
                International Journal of Cancer
                John Wiley and Sons Inc. (Hoboken )
                0020-7136
                1097-0215
                01 March 2018
                15 July 2018
                : 143
                : 2 ( doiID: 10.1002/ijc.v143.2 )
                : 383-395
                Affiliations
                [ 1 ] Department of Urology, Division of Experimental Urology Medical University of Innsbruck Innsbruck Austria
                [ 2 ] Division of Pathology Medical University of Innsbruck Innsbruck Austria
                [ 3 ] Genkyotex S.A. Geneva Switzerland
                Author notes
                [*] [* ] Correspondence to: Medical University of Innsbruck, Anichstrasse 35, A6020 Innsbruck, Austria, Tel.: +43‐512‐50‐504‐24818, Fax: +43‐512‐50‐504‐24819, E‐mail. natalie.sampson@ 123456i-med.ac.at
                Author information
                http://orcid.org/0000-0001-9326-8177
                Article
                IJC31316
                10.1002/ijc.31316
                6067067
                29441570
                a8b718f9-c208-487d-92c6-1d0b24a9aa4c
                © 2018 The Authors International Journal of Cancer published by John Wiley & Sons Ltd on behalf of UICC

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 22 August 2017
                : 18 December 2017
                : 17 January 2018
                Page count
                Figures: 6, Tables: 0, Pages: 13, Words: 8150
                Funding
                Funded by: Elise Richter fellowship from the Austrian Science Fund (FWF)
                Award ID: V216‐B13
                Funded by: Austrian Cancer Aid/Tirol
                Award ID: 15021
                Categories
                Tumor Immunology and Microenvironment
                Tumor Immunology and Microenvironment
                Custom metadata
                2.0
                ijc31316
                15 July 2018
                Converter:WILEY_ML3GV2_TO_NLMPMC version:version=5.4.4 mode:remove_FC converted:31.07.2018

                Oncology & Radiotherapy
                prostate cancer,cancer‐associated fibroblast,microenvironment,reactive oxygen species,transforming growth factor beta,in situ hybridization

                Comments

                Comment on this article