10
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Upregulation of mesothelial genes in ovarian carcinoma cells is associated with an unfavorable clinical outcome and the promotion of cancer cell adhesion

      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

          A hallmark of ovarian high‐grade serous carcinoma (HGSC) is its early and massive peritoneal dissemination via the peritoneal fluid. It is generally believed that tumor cells must breach the mesothelium of peritoneal organs to adhere to the underlying extracellular matrix (ECM) and initiate metastatic growth. However, the molecular mechanisms underlying these processes are only partially understood. Here, we have analyzed 52 matched samples of spheroids and solid tumor masses (suspected primary lesions and metastases) from 10 patients by targeted sequencing of 21 loci previously proposed as targets of HGSC driver mutations. This analysis revealed very similar patterns of genetic alterations in all samples. One exception was FAT3 with a strong enrichment of mutations in metastases compared with presumed primary lesions in two cases. FAT3 is a putative tumor suppressor gene that codes for an atypical cadherin, pointing a potential role in peritoneal dissemination in a subgroup of HGSC patients. By contrast, transcriptome data revealed clear and consistent differences between tumor cell spheroids from ascites and metastatic lesions, which were mirrored by the in vitro adherence of ascites‐derived spheroids. The adhesion‐induced transcriptional alterations in metastases and adherent cells resembled epithelial–mesenchymal transition, but surprisingly also included the upregulation of a specific subset of mesothelial genes, such as calretinin (CALB2) and podoplanin (PDPN). Consistent with this finding, calretinin staining was also observed in subsets of tumor cells in HGSC metastases, particularly at the invasive tumor edges. Intriguingly, a high expression of either CALB2 or PDPN was strongly associated with a poor clinical outcome. siRNA‐mediated CALB2 silencing triggered the detachment of adherent HGSC cells in vitro and inhibited the adhesion of detached HGSC cells to collagen type I. Our data suggest that the acquisition of a mesenchymal–mesothelial phenotype contributes to cancer cell adhesion to the ECM of peritoneal organs and HGSC progression.

          Abstract

          Attachment of ovarian cancer cells to damaged mesothelium is a critical step in metastatic spread. This study suggests that cancer cell adhesion to the extracellular matrix (ECM) is linked to mutations in the atypical cadherin and tumor suppressor FAT3 in a subset of patients, as well as to a mesenchymal–mesothelial phenotype involving the upregulation of calretinin and podoplanin.

          Related collections

          Most cited references65

          • Record: found
          • Abstract: found
          • Article: found
          Is Open Access

          Getting to Know Ovarian Cancer Ascites: Opportunities for Targeted Therapy-Based Translational Research

          More than one third of ovarian cancer patients present with ascites at diagnosis, and almost all have ascites at recurrence. The presence of ascites correlates with the peritoneal spread of ovarian cancer and is associated with poor disease prognosis. Malignant ascites acts as a reservoir of a complex mixture of soluble factors and cellular components which provide a pro-inflammatory and tumor-promoting microenvironment for the tumor cells. Subpopulations of these tumor cells exhibit cancer stem-like phenotypes, possess enhanced resistance to therapies and the capacity for distal metastatic spread and recurrent disease. Thus, ascites-derived malignant cells and the ascites microenvironment represent a major source of morbidity and mortality for ovarian cancer patients. This review focuses on recent advances in our understanding of the molecular, cellular, and functional characteristics of the cellular populations within ascites and discusses their contributions to ovarian cancer metastasis, chemoresistance, and recurrence. We highlight in particular recent translational findings which have used primary ascites-derived tumor cells as a tool to understand the pathogenesis of the disease, yielding new insights and targets for therapeutic manipulation.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Mesothelial cells promote early ovarian cancer metastasis through fibronectin secretion.

            Ovarian cancer (OvCa) metastasizes to organs in the abdominal cavity, such as the omentum, which are covered by a single layer of mesothelial cells. Mesothelial cells are generally thought to be "bystanders" to the metastatic process and simply displaced by OvCa cells to access the submesothelial extracellular matrix. Here, using organotypic 3D cultures, we found that primary human mesothelial cells secrete fibronectin in the presence of OvCa cells. Moreover, we evaluated the tumor stroma of 108 human omental metastases and determined that fibronectin was consistently overexpressed in these patients. Blocking fibronectin production in primary mesothelial cells in vitro or in murine models, either genetically (fibronectin 1 floxed mouse model) or via siRNA, decreased adhesion, invasion, proliferation, and metastasis of OvCa cells. Using a coculture model, we determined that OvCa cells secrete TGF-β1, which in turn activates a TGF-β receptor/RAC1/SMAD-dependent signaling pathway in the mesothelial cells that promotes a mesenchymal phenotype and transcriptional upregulation of fibronectin. Additionally, blocking α5 or β1 integrin function with antibodies reduced metastasis in an orthotopic preclinical model of OvCa metastasis. These findings indicate that cancer-associated mesothelial cells promote colonization during the initial steps of OvCa metastasis and suggest that mesothelial cells actively contribute to metastasis.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Podoplanin: emerging functions in development, the immune system, and cancer

              Podoplanin (PDPN) is a well-conserved, mucin-type transmembrane protein expressed in multiple tissues during ontogeny and in adult animals, including the brain, heart, kidney, lungs, osteoblasts, and lymphoid organs. Studies of PDPN-deficient mice have demonstrated that this molecule plays a critical role in development of the heart, lungs, and lymphatic system. PDPN is widely used as a marker for lymphatic endothelial cells and fibroblastic reticular cells of lymphoid organs and for lymphatics in the skin and tumor microenvironment. Much of the mechanistic insight into PDPN biology has been gleaned from studies of tumor cells; tumor cells often upregulate PDPN as they undergo epithelial-mesenchymal transition and this upregulation is correlated with increased motility and metastasis. The physiological role of PDPN that has been most studied is its ability to aggregate and activate CLEC-2-expressing platelets, as PDPN is the only known endogenous ligand for CLEC-2. However, more recent studies have revealed that PDPN also plays crucial roles in the biology of immune cells, including T cells and dendritic cells. This review will provide a comprehensive overview of the diverse roles of PDPN in development, immunology, and cancer.
                Bookmark

                Author and article information

                Contributors
                rmueller@imt.uni-marburg.de
                Journal
                Mol Oncol
                Mol Oncol
                10.1002/(ISSN)1878-0261
                MOL2
                Molecular Oncology
                John Wiley and Sons Inc. (Hoboken )
                1574-7891
                1878-0261
                25 June 2020
                September 2020
                : 14
                : 9 ( doiID: 10.1002/mol2.v14.9 )
                : 2142-2162
                Affiliations
                [ 1 ] Center for Tumor Biology and Immunology Philipps University Marburg Germany
                [ 2 ] Institute of Pathology Philipps University Marburg Germany
                [ 3 ] Institute of Pathology Justus‐Liebig University Giessen Germany
                [ 4 ] Genomics Core Facility Philipps University Marburg Germany
                [ 5 ] Institute of Molecular Oncology Member of the German Center of Lung Research (DZL) Philipps University Marburg Germany
                [ 6 ] Clinic for Gynecology Gynecological Oncology and Gynecological Endocrinology University Hospital Giessen and Marburg (UKGM) Marburg Germany
                [ 7 ]Present address: Institute of Pathology Aurich and Ammerland Aurich Germany
                Author notes
                [*] [* ] Correspondence

                R. Müller, Center for Tumor Biology and Immunology (ZTI), Philipps University, Marburg, Germany

                Tel: +49 (6421) 286 6236

                Fax: +49 (6421) 286 8923

                E‐mail: rmueller@ 123456imt.uni-marburg.de

                Author information
                https://orcid.org/0000-0003-3339-4248
                Article
                MOL212749
                10.1002/1878-0261.12749
                7463315
                32533757
                33e905f6-b577-4896-956f-9717517fe5f8
                © 2020 The Authors. Published by FEBS Press and John Wiley & Sons Ltd.

                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
                : 19 March 2020
                : 19 May 2020
                : 25 May 2020
                Page count
                Figures: 9, Tables: 2, Pages: 21, Words: 12505
                Funding
                Funded by: Deutsche Krebshilfe , open-funder-registry 10.13039/501100005972;
                Award ID: 70113255
                Funded by: Deutsche Forschungsgemeinschaft , open-funder-registry 10.13039/501100001659;
                Award ID: MU601‐25/1
                Categories
                Research Article
                Research Articles
                Custom metadata
                2.0
                September 2020
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.8.8 mode:remove_FC converted:02.09.2020

                Oncology & Radiotherapy
                adhesion,calretinin,driver mutations,mesothelial cells,metastasis,ovarian cancer,podoplanin,transcriptomics

                Comments

                Comment on this article