14
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Exosomes from human umbilical cord mesenchymal stem cells enhance fracture healing through HIF‐1α‐mediated promotion of angiogenesis in a rat model of stabilized fracture

      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

          Objectives

          Exosomes, as important players in intercellular communication due to their ability to transfer certain molecules to target cells, are believed to take similar effects in promoting bone regeneration with their derived stem cells. Studies have suggested that umbilical cord mesenchymal stem cells (uMSCs) could promote angiogenesis. This study investigated whether exosomes derived from uMSCs (uMSC‐Exos) could enhance fracture healing as primary factors by promoting angiogenesis.

          Materials and Methods

          uMSCs were obtained to isolate uMSC‐Exos by ultrafiltration, with exosomes from human embryonic kidney 293 cells (HEK293) and phosphate‐buffered saline (PBS) being used as control groups. NanoSight, laser light scattering spectrometer, transmission electron microscopy and Western blotting were used to identify exosomes. Next, uMSC‐Exos combined with hydrogel were transplanted into the fracture site in a rat model of femoral fracture. Bone healing processes were monitored and evaluated by radiographic methods on days 7, 14, 21 and 31 after surgery; angiogenesis of the fracture sites was assessed by radiographic and histological strategies on post‐operative day 14. In vitro, the expression levels of osteogenesis‐ or angiogenesis‐related genes after being cultured with uMSC‐Exos were identified by qRT‐PCR. The internalization ability of exosomes was determined using the PKH67 assay. Cell cycle analysis, EdU incorporation and immunofluorescence staining, scratch wound assay and tube formation analysis were also used to determine the altered abilities of human umbilical vein endothelial cells (HUVECs) administered with uMSC‐Exos in proliferation, migration and angiogenesis. Finally, to further explore the underlying molecular mechanisms, specific RNA inhibitors or siRNAs were used, and the subsequent effects were observed.

          Results

          uMSC‐Exos had a diameter of approximately 100 nm, were spherical, meanwhile expressing CD9, CD63 and CD81. Transplantation of uMSC‐Exos markedly enhanced angiogenesis and bone healing processes in a rat model of femoral fracture. In vitro, other than enhancing osteogenic differentiation, uMSC‐Exos increased the expression of vascular endothelial growth factor (VEGF) and hypoxia inducible factor‐1α (HIF‐1α). uMSC‐Exos were taken up by HUVECs and enhanced their proliferation, migration and tube formation. Finally, by using specific RNA inhibitors or siRNAs, it has been confirmed that HIF‐1α played an important role in the uMSC‐Exos‐induced VEGF expression, pro‐angiogenesis and enhanced fracture repair, which may be one of the underlying mechanisms.

          Conclusions

          These results revealed a novel role of exosomes in uMSC‐mediated therapy and suggested that implanted uMSC‐Exos may represent a crucial clinical strategy to accelerate fracture healing via the promotion of angiogenesis. HIF‐1α played an important role in this process.

          Abstract

          Exosomes from human umbilical cord mesenchymal stem cells enhance fracture healing through HIF‐1α‐mediated promotion of angiogenesis in a rat model.

          Related collections

          Most cited references41

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

          Coupling of angiogenesis and osteogenesis by a specific vessel subtype in bone.

          The mammalian skeletal system harbours a hierarchical system of mesenchymal stem cells, osteoprogenitors and osteoblasts sustaining lifelong bone formation. Osteogenesis is indispensable for the homeostatic renewal of bone as well as regenerative fracture healing, but these processes frequently decline in ageing organisms, leading to loss of bone mass and increased fracture incidence. Evidence indicates that the growth of blood vessels in bone and osteogenesis are coupled, but relatively little is known about the underlying cellular and molecular mechanisms. Here we identify a new capillary subtype in the murine skeletal system with distinct morphological, molecular and functional properties. These vessels are found in specific locations, mediate growth of the bone vasculature, generate distinct metabolic and molecular microenvironments, maintain perivascular osteoprogenitors and couple angiogenesis to osteogenesis. The abundance of these vessels and associated osteoprogenitors was strongly reduced in bone from aged animals, and pharmacological reversal of this decline allowed the restoration of bone mass.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Exosome-mediated transfer of miR-133b from multipotent mesenchymal stromal cells to neural cells contributes to neurite outgrowth.

            Multipotent mesenchymal stromal cells (MSCs) have potential therapeutic benefit for the treatment of neurological diseases and injury. MSCs interact with and alter brain parenchymal cells by direct cell-cell communication and/or by indirect secretion of factors and thereby promote functional recovery. In this study, we found that MSC treatment of rats subjected to middle cerebral artery occlusion (MCAo) significantly increased microRNA 133b (miR-133b) level in the ipsilateral hemisphere. In vitro, miR-133b levels in MSCs and in their exosomes increased after MSCs were exposed to ipsilateral ischemic tissue extracts from rats subjected to MCAo. miR-133b levels were also increased in primary cultured neurons and astrocytes treated with the exosome-enriched fractions released from these MSCs. Knockdown of miR-133b in MSCs confirmed that the increased miR-133b level in astrocytes is attributed to their transfer from MSCs. Further verification of this exosome-mediated intercellular communication was performed using a cel-miR-67 luciferase reporter system and an MSC-astrocyte coculture model. Cel-miR-67 in MSCs was transferred to astrocytes via exosomes between 50 and 100 nm in diameter. Our data suggest that the cel-miR-67 released from MSCs was primarily contained in exosomes. A gap junction intercellular communication inhibitor arrested the exosomal microRNA communication by inhibiting exosome release. Cultured neurons treated with exosome-enriched fractions from MSCs exposed to 72 hours post-MCAo brain extracts significantly increased the neurite branch number and total neurite length. This study provides the first demonstration that MSCs communicate with brain parenchymal cells and may regulate neurite outgrowth by transfer of miR-133b to neural cells via exosomes. Copyright © 2012 AlphaMed Press.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Endothelial Notch activity promotes angiogenesis and osteogenesis in bone.

              Blood vessel growth in the skeletal system and osteogenesis seem to be coupled, suggesting the existence of molecular crosstalk between endothelial and osteoblastic cells. Understanding the nature of the mechanisms linking angiogenesis and bone formation should be of great relevance for improved fracture healing or prevention of bone mass loss. Here we show that vascular growth in bone involves a specialized, tissue-specific form of angiogenesis. Notch signalling promotes endothelial cell proliferation and vessel growth in postnatal long bone, which is the opposite of the well-established function of Notch and its ligand Dll4 in the endothelium of other organs and tumours. Endothelial-cell-specific and inducible genetic disruption of Notch signalling in mice not only impaired bone vessel morphology and growth, but also led to reduced osteogenesis, shortening of long bones, chondrocyte defects, loss of trabeculae and decreased bone mass. On the basis of a series of genetic experiments, we conclude that skeletal defects in these mutants involved defective angiocrine release of Noggin from endothelial cells, which is positively regulated by Notch. Administration of recombinant Noggin, a secreted antagonist of bone morphogenetic proteins, restored bone growth and mineralization, chondrocyte maturation, the formation of trabeculae and osteoprogenitor numbers in endothelial-cell-specific Notch pathway mutants. These findings establish a molecular framework coupling angiogenesis, angiocrine signals and osteogenesis, which may prove significant for the development of future therapeutic applications.
                Bookmark

                Author and article information

                Contributors
                fangshuo826@126.com
                shuogui_xu@smmu.edu.cn
                Journal
                Cell Prolif
                Cell Prolif
                10.1111/(ISSN)1365-2184
                CPR
                Cell Proliferation
                John Wiley and Sons Inc. (Hoboken )
                0960-7722
                1365-2184
                20 January 2019
                March 2019
                : 52
                : 2 ( doiID: 10.1111/cpr.2019.52.issue-2 )
                : e12570
                Affiliations
                [ 1 ] Department of Emergency and Trauma Shanghai Changhai Hospital Affiliated to the Second Military Medical University Shanghai China
                [ 2 ] Department of Orthopaedics and Rehabilitation, School of Medicine Yale University New Haven Connecticut
                [ 3 ] Department of Plastic and Reconstruction Shanghai Changhai Hospital Affiliated to the Second Military Medical University Shanghai China
                Author notes
                [*] [* ] Correspondence

                Shuo Fang, Department of Plastic and Reconstruction, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China.

                Email: fangshuo826@ 123456126.com

                and

                Shuogui Xu, Department of Emergency and Trauma, Shanghai Changhai Hospital Affiliated to the Second Military Medical University, Shanghai, China.

                Email: shuogui_xu@ 123456smmu.edu.cn

                Author information
                https://orcid.org/0000-0003-2713-2460
                Article
                CPR12570
                10.1111/cpr.12570
                6496165
                30663158
                b3598b3f-d48f-4e1c-92ec-11c0e3a9e931
                © 2019 The Authors. Cell Proliferation published by 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 October 2018
                : 24 November 2018
                : 06 December 2018
                Page count
                Figures: 5, Tables: 0, Pages: 12, Words: 6999
                Funding
                Funded by: National Natural Science Foundation of China , open-funder-registry 10.13039/501100001809;
                Award ID: 81601890
                Award ID: 81701923
                Categories
                Original Manuscript
                Original Manuscripts
                Custom metadata
                2.0
                March 2019
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.7.2 mode:remove_FC converted:05.12.2019

                Cell biology
                angiogenesis,exosomes,fracture healing,hif‐1α,umbilical cord mesenchymal stem cell
                Cell biology
                angiogenesis, exosomes, fracture healing, hif‐1α, umbilical cord mesenchymal stem cell

                Comments

                Comment on this article