21
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Human Amniotic Fluid Stem Cell-Derived Exosomes as a Novel Cell-Free Therapy for Cutaneous Regeneration

      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

          Adult wound healing often results in fibrotic scarring that is caused by myofibroblast aggregation. Human amniotic fluid stem cells (hAFSCs) exhibit significantly anti-fibrotic scarring properties during wound healing. However, it is little known whether hAFSCs directly or indirectly (paracrine) contribute to this process. Using the full-thickness skin-wounded rats, we investigated the therapeutic potential of hAFSC-derived exosomes (hAFSC-exo). Our results showed that hAFSC-exo accelerated the wound healing rate and improved the regeneration of hair follicles, nerves, and vessels, as well as increased proliferation of cutaneous cells and the natural distribution of collagen during wound healing. Additionally, hAFSC-exo suppressed the excessive aggregation of myofibroblasts and the extracellular matrix. We identified several miRNAs, including let-7-5p, miR-22-3p, miR-27a-3p, miR-21-5p, and miR-23a-3p, that were presented in hAFSC-exo. The functional analysis demonstrated that these hAFSC-exo-miRNAs contribute to the inhibition of the transforming growth factor-β (TGF-β) signaling pathway by targeting the TGF-β receptor type I (TGF-βR1) and TGF-β receptor type II (TGF-βR2). The reduction of TGF-βR1 and TGF-βR2 expression induced by hAFSC-exo was also confirmed in the healing tissue. Finally, using mimics of miRNAs, we found that hAFSC-exo-miRNAs were essential for myofibroblast suppression during the TGF-β1-induced human dermal fibroblast-to-myofibroblast transition in vitro. In summary, this study is the first to show that exosomal miRNAs used in hAFSC-based therapy inhibit myofibroblast differentiation. Our study suggests that hAFSC-exo may represent a strategic tool for suppressing fibrotic scarring during wound healing.

          Related collections

          Most cited references44

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

          Mechanisms of fibrosis: therapeutic translation for fibrotic disease.

          Fibrosis is a pathological feature of most chronic inflammatory diseases. Fibrosis, or scarring, is defined by the accumulation of excess extracellular matrix components. If highly progressive, the fibrotic process eventually leads to organ malfunction and death. Fibrosis affects nearly every tissue in the body. Here we discuss how key components of the innate and adaptive immune response contribute to the pathogenesis of fibrosis. We also describe how cell-intrinsic changes in important structural cells can perpetuate the fibrotic response by regulating the differentiation, recruitment, proliferation and activation of extracellular matrix-producing myofibroblasts. Finally, we highlight some of the key mechanisms and pathways of fibrosis that are being targeted as potential therapies for a variety of important human diseases.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Using exosomes, naturally-equipped nanocarriers, for drug delivery.

            Exosomes offer distinct advantages that uniquely position them as highly effective drug carriers. Comprised of cellular membranes with multiple adhesive proteins on their surface, exosomes are known to specialize in cell-cell communications and provide an exclusive approach for the delivery of various therapeutic agents to target cells. In addition, exosomes can be amended through their parental cells to express a targeting moiety on their surface, or supplemented with desired biological activity. Development and validation of exosome-based drug delivery systems are the focus of this review. Different techniques of exosome isolation, characterization, drug loading, and applications in experimental disease models and clinic are discussed. Exosome-based drug formulations may be applied to a wide variety of disorders such as cancer, various infectious, cardiovascular, and neurodegenerative disorders. Overall, exosomes combine benefits of both synthetic nanocarriers and cell-mediated drug delivery systems while avoiding their limitations.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Mesenchymal Stem Cells for Regenerative Medicine

              In recent decades, the biomedical applications of mesenchymal stem cells (MSCs) have attracted increasing attention. MSCs are easily extracted from the bone marrow, fat, and synovium, and differentiate into various cell lineages according to the requirements of specific biomedical applications. As MSCs do not express significant histocompatibility complexes and immune stimulating molecules, they are not detected by immune surveillance and do not lead to graft rejection after transplantation. These properties make them competent biomedical candidates, especially in tissue engineering. We present a brief overview of MSC extraction methods and subsequent potential for differentiation, and a comprehensive overview of their preclinical and clinical applications in regenerative medicine, and discuss future challenges.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Cell Dev Biol
                Front Cell Dev Biol
                Front. Cell Dev. Biol.
                Frontiers in Cell and Developmental Biology
                Frontiers Media S.A.
                2296-634X
                21 June 2021
                2021
                : 9
                : 685873
                Affiliations
                [1] 1Hospital of Stomatology, Jilin University , Changchun, China
                [2] 2Jilin Provincial Laboratory of Biomedical Engineering, Jilin University , Changchun, China
                [3] 3Center for Reproductive Medicine, Center for Prenatal Diagnosis, The First Hospital of Jilin University , Changchun, China
                [4] 4Chengnan Branch, Foshan Stomatology Hospital, School of Stomatology and Medicine, Foshan University , Foshan, China
                [5] 5Affiliated Stomatology Hospital, Guangzhou Medical University , Guangzhou, China
                [6] 6Institute of Antler Science and Product Technology, Changchun Sci-Tech University , Changchun, China
                [7] 7Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences , Changchun, China
                Author notes

                Edited by: Vicente Herranz-Pérez, University of Valencia, Spain

                Reviewed by: Ulf Anderegg, Leipzig University, Germany; Lina Dagnino, Western University, Canada

                *Correspondence: Jiang Li, ljiang@ 123456gzhmu.edu.cn

                This article was submitted to Stem Cell Research, a section of the journal Frontiers in Cell and Developmental Biology

                Article
                10.3389/fcell.2021.685873
                8255501
                34235150
                edc135dd-a41b-47ac-8a41-a58087b63480
                Copyright © 2021 Zhang, Yan, Liu, Chen, Li, Tang, Li, Duan and Zhang.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 25 March 2021
                : 27 May 2021
                Page count
                Figures: 9, Tables: 0, Equations: 0, References: 45, Pages: 14, Words: 0
                Categories
                Cell and Developmental Biology
                Original Research

                human amniotic fluid stem cells,exosomes,mirna,scarring,transforming growth factor,wound healing

                Comments

                Comment on this article