5
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Myeloid‐derived suppressor cells promote epithelial ovarian cancer cell stemness by inducing the CSF2/p‐STAT3 signalling pathway

      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

          Myeloid‐derived suppressor cells (MDSCs) are known to contribute to tumour immune evasion, and studies have verified that MDSCs can induce cancer stem cells (CSCs) and promote tumour immune evasion in breast cancers, cervical cancers and glioblastoma. However, the potential function of MDSCs in regulating CSCs in epithelial ovarian cancer (EOC) progression is unknown. Our results indicated that compared to nonmalignant ovarian patients, EOC patients showed a significantly increased proportion of MDSCs in the peripheral blood. In addition, MDSCs dramatically promoted tumour sphere formation, cell colony formation and CSC accumulation, and MDSCs enhanced the expression of the stemness biomarkers NANOG and c‐MYC in EOC cells during coculture. Moreover, the mechanisms by which MDSCs enhance EOC stemness were further explored, and 586 differentially expressed genes were found in EOC cells cocultured with or without MDSCs; during coculture, the expression level of colony‐stimulating factor 2 (CSF2) was significantly increased in EOC cells cocultured with MDSCs. Furthermore, the depletion of CSF2 in EOC cells was successfully performed, the promotive effects of MDSCs on EOC cell stemness could be markedly reversed by downregulating CSF2 expression, p‐STAT3 signalling pathway molecules were also altered, and the p‐STAT3 inhibitor could markedly reverse the promotive effects of MDSCs on EOC cell stemness. In addition, the CSF2 expression level was correlated with EOC clinical staging. Therefore, MDSCs enhance the stemness of EOC cells by inducing the CSF2/p‐STAT3 signalling pathway. Targeting MDSCs or CSF2 may be a reasonable strategy for enhancing the efficacy of conventional treatments.

          Database

          Gene expression data files are available in the GEO databases under the accession number(s) GSE145374.

          Abstract

          Previous studies have verified that myeloid‐derived suppressor cells (MDSCs) can promote tumour immune evasion. We found that epithelial ovarian cancer (EOC) patients showed a significantly increased proportion of MDSCs in the peripheral blood. MDSCs dramatically promoted EOC cell stemness, and the promotive effects of MDSCs on EOC cell stemness could be markedly reversed by downregulating CSF2/p‐STAT3 expression. In addition, the CSF2 expression level was correlated with EOC clinical staging.

          Related collections

          Most cited references61

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

          The blockade of immune checkpoints in cancer immunotherapy.

          Among the most promising approaches to activating therapeutic antitumour immunity is the blockade of immune checkpoints. Immune checkpoints refer to a plethora of inhibitory pathways hardwired into the immune system that are crucial for maintaining self-tolerance and modulating the duration and amplitude of physiological immune responses in peripheral tissues in order to minimize collateral tissue damage. It is now clear that tumours co-opt certain immune-checkpoint pathways as a major mechanism of immune resistance, particularly against T cells that are specific for tumour antigens. Because many of the immune checkpoints are initiated by ligand-receptor interactions, they can be readily blocked by antibodies or modulated by recombinant forms of ligands or receptors. Cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) antibodies were the first of this class of immunotherapeutics to achieve US Food and Drug Administration (FDA) approval. Preliminary clinical findings with blockers of additional immune-checkpoint proteins, such as programmed cell death protein 1 (PD1), indicate broad and diverse opportunities to enhance antitumour immunity with the potential to produce durable clinical responses.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Cancer immunoediting: integrating immunity's roles in cancer suppression and promotion.

            Understanding how the immune system affects cancer development and progression has been one of the most challenging questions in immunology. Research over the past two decades has helped explain why the answer to this question has evaded us for so long. We now appreciate that the immune system plays a dual role in cancer: It can not only suppress tumor growth by destroying cancer cells or inhibiting their outgrowth but also promote tumor progression either by selecting for tumor cells that are more fit to survive in an immunocompetent host or by establishing conditions within the tumor microenvironment that facilitate tumor outgrowth. Here, we discuss a unifying conceptual framework called "cancer immunoediting," which integrates the immune system's dual host-protective and tumor-promoting roles.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Myeloid-derived suppressor cells as regulators of the immune system.

              Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of cells that expand during cancer, inflammation and infection, and that have a remarkable ability to suppress T-cell responses. These cells constitute a unique component of the immune system that regulates immune responses in healthy individuals and in the context of various diseases. In this Review, we discuss the origin, mechanisms of expansion and suppressive functions of MDSCs, as well as the potential to target these cells for therapeutic benefit.
                Bookmark

                Author and article information

                Contributors
                lingfeihan@126.com
                Xiaoqing_Guo@tongji.edu.cn
                Journal
                FEBS J
                FEBS J
                10.1111/(ISSN)1742-4658
                FEBS
                The Febs Journal
                John Wiley and Sons Inc. (Hoboken )
                1742-464X
                1742-4658
                17 April 2020
                December 2020
                : 287
                : 23 ( doiID: 10.1111/febs.v287.23 )
                : 5218-5235
                Affiliations
                [ 1 ] Department of Obstetrics and Gynecology Shanghai First Maternity and Infant Hospital Tongji University School of Medicine Shanghai China
                Author notes
                [*] [* ] Correspondence

                X. Guo and L. Han, Department of Obstetrics and Gynecology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, 2699 West Gaoke Road, Shanghai 201204, China

                Tel: +86 02120261072

                E‐mails: Xiaoqing_Guo@ 123456tongji.edu.cn (XG); lingfeihan@ 123456126.com (LH)

                Author information
                https://orcid.org/0000-0002-2493-5156
                Article
                FEBS15311
                10.1111/febs.15311
                7754107
                32239647
                7e6503d1-99f0-4744-91bd-5af52ef4f3d2
                © 2020 The Authors. The FEBS Journal published by John Wiley & Sons Ltd on behalf of Federation of European Biochemical Societies

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.

                History
                : 12 August 2019
                : 17 February 2020
                : 25 March 2020
                Page count
                Figures: 9, Tables: 3, Pages: 18, Words: 8086
                Funding
                Funded by: National Natural Science Foundation of China , open-funder-registry 10.13039/501100001809;
                Award ID: 81572546
                Award ID: 81601309
                Award ID: 81972422
                Funded by: Shanghai First Maternity and Infant Hospital
                Award ID: 2018C06
                Categories
                Original Article
                Original Articles
                Custom metadata
                2.0
                December 2020
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.9.6 mode:remove_FC converted:22.12.2020

                Molecular biology
                cancer stem cells,colony‐stimulating factor 2,epithelial ovarian cancer,myeloid‐derived suppressor cells

                Comments

                Comment on this article