13
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      hUMSCs regulate the differentiation of ovarian stromal cells via TGF-β 1/Smad3 signaling pathway to inhibit ovarian fibrosis to repair ovarian function in POI rats

      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

          Objective

          The basic pathological changes of primary ovarian insufficiency (POI) include ovarian tissue fibrosis and follicular development disorders. The human umbilical cord mesenchymal stem cell (hUMSC) transplantation has been shown an effective method to improve the ovarian function in POI rat model; however, the exact mechanisms are still unclear. The purpose of this study is to investigate whether the recovery of ovarian function in POI rats is related to the inhibition of tissue fibrosis following hUMSC transplantation. Furthermore, the transforming growth factor-β 1 (TGF-β 1) signaling pathway is explored to determine the mechanisms of ovarian function recovery through its inhibition of tissue fibrosis.

          Methods

          The primary ovarian insufficiency (POI) rat model was established by intraperitoneal injection of chemotherapy drug cisplatin (CDDP) for 7 days. The levels of serum sex hormones were measured using enzyme-linked immunosorbent assay (ELISA). The tissue fibrosis in the ovary was examined using Masson staining and Sirius red staining. The collagen fibers in the ovarian tissues were detected by Western blot analysis. To investigate the mechanisms of ovarian function recovery following hUMSC transplantation, ovarian stromal cells were isolated from the ovarian cortex of immature rats. The expression of Cytochrome P450 17A1 (Cyp17a1) and fibrosis marker of alpha smooth muscle actin (α-SMA) in ovarian stromal cells was examined using immunofluorescence analysis. Also, the protein levels of Cyp17a1 and α-SMA in ovarian stromal cells were examined by Western blot analysis. The expression of TGF-β 1 and Smad3 signals was measured by Western blot and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) analysis.

          Results

          The results show that the function of the ovary in POI rats was significantly improved after hUMSC transplantation. The expression of fibrosis markers (α-SMA) and production of Collagen Type I (Collagen I) and Collagen Type III (Collagen III) in POI rats were significantly inhibited in POI rats following hUMSC transplantation. In the cultured ovarian stromal cells, the decrease of TGF-β 1 and p-Smad3 protein expression was observed in hUMSC-treated POI rats. The treatment with TGF-β 1 inhibitor of SB431542 further confirmed this signal pathway was involved in the process.

          Conclusion

          Our study demonstrated that the TGF-β 1/Smad3 signaling pathway was involved in the inhibition of ovarian tissue fibrosis, which contributed to the restoration of ovarian function in POI rats following hUMSC transplantation.

          Related collections

          Most cited references40

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

          Targeted disruption of TGF-beta1/Smad3 signaling protects against renal tubulointerstitial fibrosis induced by unilateral ureteral obstruction.

          Tubulointerstitial fibrosis is the final common result of a variety of progressive injuries leading to chronic renal failure. Transforming growth factor-beta (TGF-beta) is reportedly upregulated in response to injurious stimuli such as unilateral ureteral obstruction (UUO), causing renal fibrosis associated with epithelial-mesenchymal transition (EMT) of the renal tubules and synthesis of extracellular matrix. We now show that mice lacking Smad3 (Smad3ex8/ex8), a key signaling intermediate downstream of the TGF-beta receptors, are protected against tubulointerstitial fibrosis following UUO as evidenced by blocking of EMT and abrogation of monocyte influx and collagen accumulation. Culture of primary renal tubular epithelial cells from wild-type or Smad3-null mice confirms that the Smad3 pathway is essential for TGF-beta1-induced EMT and autoinduction of TGF-beta1. Moreover, mechanical stretch of the cultured epithelial cells, mimicking renal tubular distention due to accumulation of urine after UUO, induces EMT following Smad3-mediated upregulation of TGF-beta1. Exogenous bone marrow monocytes accelerate EMT of the cultured epithelial cells and renal tubules in the obstructed kidney after UUO dependent on Smad3 signaling. Together the data demonstrate that the Smad3 pathway is central to the pathogenesis of interstitial fibrosis and suggest that inhibitors of this pathway may have clinical application in the treatment of obstructive nephropathy.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Mesenchymal stromal cells: facilitators of successful transplantation?

            Mesenchymal stromal/stem cells (MSCs) possess immunomodulatory and reparative properties. Through specific interactions with immune cells that participate in both innate and adaptive responses, MSCs exposed to an inflammatory microenvironment can downregulate many immune effector functions. Clinical trials focusing on MSCs to treat graft-versus-host disease (GvHD) and autoimmune diseases are underway. Current analyses suggest that MSCs will improve cell and solid organ transplantation by ameliorating rejection and possibly eliminating the requirement for prolonged regimens of conventional immunosuppressive drugs. This review examines the in vitro and in vivo evidence for the clinical use of bone marrow derived MSCs. Copyright © 2010 Elsevier Inc. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Human endometrial mesenchymal stem cells restore ovarian function through improving the renewal of germline stem cells in a mouse model of premature ovarian failure

              Background Human endometrial mesenchymal stem cells (EnSCs) derived from menstrual blood have mesenchymal stem/stromal cells (MSCs) characteristics and can differentiate into cell types that arise from all three germ layers. We hypothesized that EnSCs may offer promise for restoration of ovarian dysfunction associated with premature ovarian failure/insufficiency (POF/POI). Methods Mouse ovaries were injured with busulfan and cyclophosphamide (B/C) to create a damaged ovary mouse model. Transplanted EnSCs were injected into the tail vein of sterilized mice (Chemoablated with EnSCs group; n = 80), or culture medium was injected into the sterilized mice via the tail vein as chemoablated group (n = 80). Non-sterilized mice were untreated controls (n = 80). Overall ovarian function was measured using vaginal smears, live imaging, mating trials and immunohistochemical techniques. Results EnSCs transplantation increased body weight and improved estrous cyclicity as well as restored fertility in sterilized mice. Migration and localization of GFP-labeled EnSCs as measured by live imaging and immunofluorescent methods indicated that GFP-labeled cells were undetectable 48 h after cell transplantation, but were later detected in and localized to the ovarian stroma. 5’-bromodeoxyuridine (BrdU) and mouse vasa homologue (MVH) protein double-positive cells were immunohistochemically detected in mouse ovaries, and EnSC transplantation reduced depletion of the germline stem cell (GSCs) pool induced by chemotherapy. Conclusion EnSCs derived from menstrual blood, as autologous stem cells, may restore damaged ovarian function and offer a suitable clinical strategy for regenerative medicine. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0516-y) contains supplementary material, which is available to authorized users.
                Bookmark

                Author and article information

                Contributors
                qiangfu11@fudan.edu.cn
                byzhhq@bzmc.edu.cn
                Journal
                Stem Cell Res Ther
                Stem Cell Res Ther
                Stem Cell Research & Therapy
                BioMed Central (London )
                1757-6512
                7 September 2020
                7 September 2020
                2020
                : 11
                : 386
                Affiliations
                [1 ]GRID grid.440653.0, ISNI 0000 0000 9588 091X, College of Basic Medicine & Institute of Reproductive Diseases, , Binzhou Medical University, ; Yantai, 264003 Shandong China
                [2 ]GRID grid.440653.0, ISNI 0000 0000 9588 091X, College of Basic Medicine, , Binzhou Medical University, ; Yantai, 264003 Shandong China
                [3 ]GRID grid.440323.2, Department of Clinical Medicine, , Yantai Yuhuangding Hospital, ; Yantai, 264000 Shandong China
                [4 ]GRID grid.452240.5, Yantai Affiliated Hospital of Binzhou Medical University, ; Yantai, 264100 Shandong China
                [5 ]GRID grid.440653.0, ISNI 0000 0000 9588 091X, Clinical Medical School, , Binzhou Medical University, ; Yantai, Shandong China
                [6 ]GRID grid.440653.0, ISNI 0000 0000 9588 091X, School of pharmacy, , Binzhou Medical University, ; Yantai, Shandong China
                Author information
                http://orcid.org/0000-0003-1440-7365
                Article
                1904
                10.1186/s13287-020-01904-3
                7487655
                32894203
                cd2d8283-609e-40a2-95c7-57e73ceac443
                © The Author(s) 2020

                Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

                History
                : 22 June 2020
                : 17 August 2020
                : 26 August 2020
                Funding
                Funded by: the Xu Rongxiang Fund
                Award ID: NO. BY2019XRX01
                Award Recipient :
                Funded by: the Major Science and Technology Innovation Plan of Shandong Province
                Award ID: NO.2019JZZY020902
                Award Recipient :
                Funded by: the Science and Technology Plan of Yantai city government
                Award ID: NO. 2019MSGY134
                Award Recipient :
                Funded by: Medical Health Science and Technology Project of Shandong Provincial Health Commission
                Award ID: NO. 2018WS552
                Award Recipient :
                Categories
                Research
                Custom metadata
                © The Author(s) 2020

                Molecular medicine
                primary ovarian insufficiency,human umbilical cord-derived mesenchymal stem cells,ovarian stromal cells,tgf-β1 signaling pathway,fibrosis

                Comments

                Comment on this article