15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The potential and limitations of induced pluripotent stem cells to achieve wound healing

      review-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

          Wound healing is the physiologic response to a disruption in normal skin architecture and requires both spatial and temporal coordination of multiple cell types and cytokines. This complex process is prone to dysregulation secondary to local and systemic factors such as ischemia and diabetes that frequently lead to chronic wounds. Chronic wounds such as diabetic foot ulcers are epidemic with great cost to the healthcare system as they heal poorly and recur frequently, creating an urgent need for new and advanced therapies. Stem cell therapy is emerging as a potential treatment for chronic wounds, and adult-derived stem cells are currently employed in several commercially available products; however, stem cell therapy is limited by the need for invasive harvesting techniques, immunogenicity, and limited cell survival in vivo. Induced pluripotent stem cells (iPSC) are an exciting cell type with enhanced therapeutic and translational potential. iPSC are derived from adult cells by in vitro induction of pluripotency, obviating the ethical dilemmas surrounding the use of embryonic stem cells; they are harvested non-invasively and can be transplanted autologously, reducing immune rejection; and iPSC are the only cell type capable of being differentiated into all of the cell types in healthy skin. This review focuses on the use of iPSC in animal models of wound healing including limb ischemia, as well as their limitations and methods aimed at improving iPSC safety profile in an effort to hasten translation to human studies.

          Related collections

          Most cited references61

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

          Paracrine mechanisms of mesenchymal stem cell-based therapy: current status and perspectives.

          Mesenchymal stem cells (MSCs) are one of a few stem cell types to be applied in clinical practice as therapeutic agents for immunomodulation and ischemic tissue repair. In addition to their multipotent differentiation potential, a strong paracrine capacity has been proposed as the principal mechanism that contributes to tissue repair. Apart from cytokine/chemokine secretion, MSCs also display a strong capacity for mitochondrial transfer and microvesicle (exosomes) secretion in response to injury with subsequent promotion of tissue regeneration. These unique properties of MSCs make them an invaluable cell type to repair damaged tissues/organs. Although MSCs offer great promise in the treatment of degenerative diseases and inflammatory disorders, there are still many challenges to overcome prior to their widespread clinical application. Particularly, their in-depth paracrine mechanisms remain a matter for debate and exploration. This review will highlight the discovery of the paracrine mechanism of MSCs, regulation of the paracrine biology of MSCs, important paracrine factors of MSCs in modulation of tissue repair, exosome and mitochondrial transfer for tissue repair, and the future perspective for MSC-based therapy.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Regulation of wound healing by growth factors and cytokines.

            Cutaneous wound healing is a complex process involving blood clotting, inflammation, new tissue formation, and finally tissue remodeling. It is well described at the histological level, but the genes that regulate skin repair have only partially been identified. Many experimental and clinical studies have demonstrated varied, but in most cases beneficial, effects of exogenous growth factors on the healing process. However, the roles played by endogenous growth factors have remained largely unclear. Initial approaches at addressing this question focused on the expression analysis of various growth factors, cytokines, and their receptors in different wound models, with first functional data being obtained by applying neutralizing antibodies to wounds. During the past few years, the availability of genetically modified mice has allowed elucidation of the function of various genes in the healing process, and these studies have shed light onto the role of growth factors, cytokines, and their downstream effectors in wound repair. This review summarizes the results of expression studies that have been performed in rodents, pigs, and humans to localize growth factors and their receptors in skin wounds. Most importantly, we also report on genetic studies addressing the functions of endogenous growth factors in the wound repair process.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis

              Background Human induced pluripotent stem cell-derived mesenchymal stem cells (hiPSC-MSCs) have emerged as a promising alternative for stem cell transplantation therapy. Exosomes derived from mesenchymal stem cells (MSC-Exos) can play important roles in repairing injured tissues. However, to date, no reports have demonstrated the use of hiPSC-MSC-Exos in cutaneous wound healing, and little is known regarding their underlying mechanisms in tissue repair. Methods hiPSC-MSC-Exos were injected subcutaneously around wound sites in a rat model and the efficacy of hiPSC-MSC-Exos was assessed by measuring wound closure areas, by histological and immunofluorescence examinations. We also evaluated the in vitro effects of hiPSC-MSC-Exos on both the proliferation and migration of human dermal fibroblasts and human umbilical vein endothelial cells (HUVECs) by cell-counting and scratch assays, respectively. The effects of exosomes on fibroblast collagen and elastin secretion were studied in enzyme-linked immunosorbent assays and quantitative reverse-transcriptase–polymerase chain reaction (qRT-PCR). In vitro capillary network formation was determined in tube-formation assays. Results Transplanting hiPSC-MSC-Exos to wound sites resulted in accelerated re-epithelialization, reduced scar widths, and the promotion of collagen maturity. Moreover, hiPSC-MSC-Exos not only promoted the generation of newly formed vessels, but also accelerated their maturation in wound sites. We found that hiPSC-MSC-Exos stimulated the proliferation and migration of human dermal fibroblasts and HUVECs in a dose-dependent manner in vitro. Similarly, Type I, III collagen and elastin secretion and mRNA expression by fibroblasts and tube formation by HUVECs were also increased with increasing hiPSC-MSC-Exos concentrations. Conclusions Our findings suggest that hiPSC-MSC-Exos can facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. These data provide the first evidence for the potential of hiPSC-MSC-Exos in treating cutaneous wounds.
                Bookmark

                Author and article information

                Contributors
                jolanta.gorecka@yale.edu
                valentyna.kostiuk@yale.edu
                arash.fereydooni@yale.edu
                luis.gonzalez@yale.edu
                jiesi.luo@yale.edu
                biraja.dash@yale.edu
                toshihiko.isaji@yale.edu
                shu.ono@yale.edu
                shirley.liu@yale.edu
                shinrong.lee@yale.edu
                jx254@connect.yale.edu
                jia.liu.jl3365@yale.edu
                ryosuke.taniguchi@yale.edu
                bogdan.yatsula@yale.edu
                henry.hsia@yale.edu
                yibing.qyang@yale.edu
                alan.dardik@yale.edu
                Journal
                Stem Cell Res Ther
                Stem Cell Res Ther
                Stem Cell Research & Therapy
                BioMed Central (London )
                1757-6512
                12 March 2019
                12 March 2019
                2019
                : 10
                : 87
                Affiliations
                [1 ]ISNI 0000000419368710, GRID grid.47100.32, Vascular Biology and Therapeutics Program and the Department of Surgery, Yale School of Medicine, , Yale University, ; 10 Amistad Street, Room 437, PO Box 208089, New Haven, CT 06520-8089 USA
                [2 ]ISNI 0000000419368710, GRID grid.47100.32, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale Cardiovascular Research Center, , Yale School of Medicine, ; 300 George Street, Ste 773A, New Haven, CT 06511 USA
                [3 ]ISNI 0000000419368710, GRID grid.47100.32, Section of Plastic Surgery, Department of Surgery, , Yale School of Medicine, Yale University, ; PO Box 208062, New Haven, CT 06520-8062 USA
                [4 ]ISNI 0000000419368710, GRID grid.47100.32, Yale Stem Cell Center, , Yale University, ; New Haven, USA
                [5 ]ISNI 0000000419368710, GRID grid.47100.32, Vascular Biology and Therapeutics Program, , Yale School of Medicine, ; New Haven, USA
                [6 ]ISNI 0000000419368710, GRID grid.47100.32, Department of Pathology, , Yale University, ; New Haven, USA
                Article
                1185
                10.1186/s13287-019-1185-1
                6416973
                30867069
                604014a3-90d1-4ce8-833e-7634a769cbda
                © The Author(s). 2019

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

                History
                Funding
                Funded by: Association of VA Surgeons
                Award ID: Resident Research Award
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/100000002, National Institutes of Health;
                Award ID: R01HL128406
                Award ID: R01HL116705
                Award Recipient :
                Funded by: National Institutes of Health (US)
                Award ID: R01HL132130
                Award Recipient :
                Funded by: DOD
                Award ID: DOD11959515
                Award Recipient :
                Funded by: Connecticut Regenerative Medicine Research Fund
                Categories
                Review
                Custom metadata
                © The Author(s) 2019

                Molecular medicine
                induced pluripotent stem cell,stem cell,chronic wounds,wound healing,diabetic foot ulcer,peripheral arterial disease,angiogenesis,diabetes,teratoma

                Comments

                Comment on this article