Journal of Stem Cells Research Development & Therapy Category: Medical Type: Review Article

Mesenchymal Stem Cells, Exosomes and Cutaneous Wound Healing

Guanghao Zhu1, Haoyu Gu1 and Minjuan Wu1*
1 Department of histology and embryology, College of Basic Medicine, Second Military Medical University, Shanghai, 200433, China

*Corresponding Author(s):
Minjuan Wu
Department Of Histology And Embryology, College Of Basic Medicine, Second Military Medical University, Shanghai, 200433, China
Email:minjuanwu@163.com

Received Date: Feb 05, 2019
Accepted Date: Feb 20, 2020
Published Date: Feb 28, 2020

Abstract

Mesenchymal Stem Cells (MSCs) have been widely used in tissue regeneration due to the characteristic of multi-differentiation potential, strong proliferation ability, low immunogenicity, convenient material collection, no restrictions on ethical issues, and easy industrialization. A large number of studies on MSCs -based therapy for cutaneous wound have shown satisfactory results. However, the MSCs therapy is still facing many challenges, and there is still a long way to go for clinical applications. This article briefly reviewed the role and mechanism of MSCs in cutaneous wound healing and discussed issues that should be noticed in future studies. In particular, the research progress of exosomes derived from MSCs in cutaneous wound repair is reviewed.

INTRODUCTION

Skin is the largest organ of the human body. There are many factors that can cause cutaneous damage in the clinic, such as trauma, burns, diabetes, and surgery. The process of wound healing is generally described as three sequential phases: inflammation, proliferation, and remodeling [1,2]. The mechanism of wound healing is complicated, and the loss of control at any of these stages will lead to keloids, delayed wound healing of healing difficulty, therefore making the prognosis of the patient worse, and serious, even fatal infection [3]. In recent years, major breakthroughs in the clinical application of MSCs include hematological diseases, cardiovascular diseases, liver cirrhosis, neurological diseases, partial meniscus resection and repair of knee joints, and autoimmune diseases [4-9]. Studies have shown that MSCs can be used in the cutaneous wound repair [10,11]. MSCs therapy will be the most effective therapy in this field. However, there is still a long way to go before MSCs can be used to repair cutaneous injuries in clinic. So we will discuss the progress of MSCs therapy in cutaneous wound healing.

MESENCHYMAL STEM CELLS

MSCs was first discovered and reported by Friedenstein in the bone marrow in 1968 [12]. In 1999, Pittenger reported on its multi-differentiation potential [13]. In addition to bone marrow, various tissues of the body contain mesenchymal stem cells, such as adipose, muscle, liver, and umbilical cord [14]. MSCs have multi-directional differentiation potential. Under different induction conditions, they can differentiate into mesoderm cells, such as cartilage, bone, skeletal muscle, and fat, and neural cells of ectoderm and hepatic oval cells of endoderm [15]. Pluripotency of MSCs allows it to have a wide range of applications for repairing many different tissues. Under certain induction conditions, MSCs can also differentiate into cutaneous cells. Experiments in vivo confirmed that MSCs can differentiate into sebaceous ductal cells and epidermal cells in regenerated skin [16].

The current definition of MSCs is still unclear. At present, MSCs cells are considered to have at least the following three characteristics: 1) MSCs must be plastic-adherent when maintained in standard culture conditions; 2) MSCs must express CD105, CD73 and CD90, and lack expression of CD45, CD34, CD14 or CD11b, CD79alpha or CD19 and HLA-DR surface molecules; 3) MSCs must differentiate to osteoblasts, adipocytes and chondroblasts in vitro [17].

MSCs differentiate to form a variety of tissue cells, which are involved in tissue regeneration and wound repair

Although MSCs are derived from mesoderm, they can still be transformed into other germ layer cells through specific environment induction. Transplant adipose derived mesenchymal stem cells (ADMSCs) in the microenvironment of human epidermal-derived keratinocytes, and ADMSCs can differentiate into keratinocyte like cells and express keratinocyte-specific markers such as cytokeratin 14, cytokeratin 5, cytokeratin 19 [18]. By co-culturing ADMSCs with keratinocytes, cells expressing keratinocyte lineage markers i.e. cytokeratin 14, cytokeratin 5, cytokeratin 10, cytokeratin 18, cytokeratin 19 [19]. They can also directly differentiate into different kinds of cells and replace the lost tissue, such as fibroblasts and skin appendages [20-23]. Similar to in vitro studies, ADMSCs can directly differentiate into epidermal cell lines, fibroblast cell lines, and endothelial cell lines under the induction of cutaneous injury microenvironment, providing sufficient cell sources for cutaneous wound healing [24, 25].

MSCs secrete paracrine factors that play crucial roles during tissue regeneration

After being stimulated and activated in the injured local microenvironment, MSC can secrete active ingredients such as cytokines, inflammatory mediators, and antibacterial proteins, thereby exerting corresponding effects locally or throughout the body. MSCs possess powerful immune modulatory properties and are able to activate various genes that contribute to tissues repair [26,27]. Moreover, MSCs may regulate local reparatory responses by recruiting host cells, such as fibroblasts, keratinocytes, macrophages and progenitor cells migrate to the injured site [28-31]. Additionally, MSCs may also induce angiogenesis which is critical in wound healing through the secretion of all kinds of cellular factors [28,32,33].

Exosomes derived from MSCs play a critical role in tissue regeneration

In more recent findings, researchers have found that MSCs achieve the therapeutic effect in vivo mainly through paracrine signaling, they can release biologically active factors that affect the proliferation, migration and survival of the target cells [34-36]. Exosomes are the key bioactive vesicles responsible for the paracrine effects of MSCs, mimic the effects of parental MSCs. 

Exosomes are small (30-100 nm in diameter) extracellular membrane-enclosed vesicles released by different cells into the extracellular space or into biological fluids. Exosomes are released by exocytosis as result of fusion of intracellular multivesicular bodies with the plasma membrane, they can shuttle various effector proteins, DNA, small interfering RNA (siRNA), messenger RNA (mRNA) and microRNAs (miRNAs) to modulate the activity of recipient cells, playing important roles in wound healing. Several studies have reported that MSC-derived conditioned medium promotes cutaneous regeneration [30,37].

Exosomes derived from human umbilical cord MSCs (hucMSC-exosome), hADMSC-exosome, and human induced pluripotent stem cell-derived MSCs (hiPS-MSC-exosome) [38-40] can facilitate cutaneous wound healing by delivering various functional proteins, RNAs and soluble cytokines [41-43]. Most researchers believe that MSC-exosomes are the main effective paracrine component of MSCs and play biological effect almost equivalent to those of whole MSCs. 

Comparing with MSCs, MSC-exosomes have the following advantages: 1) MSC-exosomes exert intense biological effects because they directly fuse with target cells; 2) MSC-exosomes can be stored and transported at low degree for a long time; 3) The concentration, dose, route and time of use are easy to control; 4) There is no risk of immune rejection and tumorigenesis caused by cell transplantation therapy [44]. 

The process of cutaneous regeneration can be summarized as three important stages: inflammation stage, proliferation stage and remodeling stage [45]. MSC-exosomes are able to play an important role in all three stages (Table1). They lead to proliferation and re-epithelization by enhancing proliferation and migration of fibroblasts and keratinocytes through mediating activation of several factors, MSC-exosomes enhance wound healing by delivering Wnt4 [38]. MSC-exosomes can exhibit immunosuppressive effects by regulating proliferation and differentiation of lymphocytes, MSC-exosomes can repress T-lymphocyte proliferation and they exchange T lymphocytes into the T-regulatory phenotype. MSC-exosomes also enhance converting of macrophages toward the anti-inflammatory M2 phenotype in the inflammation stage [46]. MSC-exosomes can also exhibit angiogenic effects through several mechanism, they cause antivascular remodeling by suppress HIMF [47,48]. 

Stage

Effects

Molecular in Exosomes

References

Inflammation

Induce Tregs, suppress Th1 and Th17

miR-let7b, miR181c, miR-126, miR-130a, miR-125a, miR-131

[52,53]

[45,54,55]

Promote M2 macrophages, suppress M1 macrophages

Enhance IL-10 production?inhibit levels of TNF-α and IL-1β

dampen complement

CD59

[52]

Proliferation

Promote fibroblast, keratinocyte and epidermal cells proliferation, migration and re-epithelization

Wnt4, Wnt11, miR221/222

[38,56]

Promote angiogenesis

miR-126, miR-130a, miR-132, miR-125a, miR-31, Wnt4

[47,56-58]

Remodeling

Antivascular remodeling

suppress HIMF

[47,48]

Modulate collagen secretion and deposition, anti-scarring effect

miR-21, miR125b, miR-23a, miR-145, YAP

[41,51,59]

Table1: Effects of MSC-exosomes in different stages of cutaneous regeneration.

Composition of the MSC-exosomes can easily deliver the massage into target cells due to their lipid layer which can avoid proteolytic degradation [49]. Further, MSC-exosomes can activate some signaling pathways including Signal Transducer and Activator of Transcription 3 (STAT3), AKT, Wnt/β-catenin, and extracellular signal-regulated kinase (ERK) in target cells which play an important role in wound healing process [50]. Activation of these signaling pathways also can enhance the expression of several growth factors which involved in wound regeneration process by target cells, such as lnterleukin-6 (IL-6), STAT3, Hepatocyte Growth Factor (HGF), Insulin-like Growth Factor-1 (IGF-1), and Stromal Cell-Derived Factor-1 (SDF-1) [39,50], these growth factors can promote the angiogenesis, cell migration, cell proliferation, and re-epithelialization [50]. On the other hand, it has been revealed that MSC-exosomes in wound environment can transfer Wnt4 to stimulate Wnt/β-catenin pathway in skin cells, and subsequently active AKT pathway to inhibit epidermal cell apoptosis. β-catenin signaling pathway can stimulate pro-angiogenic effects in endothelial cells and enhance cutaneous wound healing [38,50]. 

Since MSC-exosomes are an ideal therapeutic tool for their plentiful advantages, such as easy to get and storage, increased safety and efficiency and lower immune rejection, they have been widely reported to support cutaneous regeneration during the proliferation stage. hADMSC-exosomes can promote fibroblast proliferation and collagen synthesis by up-regulating the gene expression of N-cadherin, cyclin-1, Proliferating Cell Nuclear Antigen (PCNA), collagen I and collagen III in vitro. In vivo experiments have also proven that hADMSC-exosomes could home to the skin incision site and significantly facilitate cutaneous wound healing [39]. 

In other studies, injecting hiPSC-MSC-exosomes to wound sites facilitated cutaneous wound healing by promoting human dermal fibroblast proliferation and migration and by enhancing collagen synthesis [40]. Moreover, transplanting human amniotic epithelial cell-derived exosomes (hAEC-exosomes) to wound sites accelerated wound closure and re-epithelization [51]. 

Previous study revealed that hucMSC-exosome promotes cell proliferation and accelerates re-epithelialization; in a rat deep second-degree burn injury model, hucMSC-exosomes reversed acute heat stress-induced skin cell apoptosis and increased the expression of cytokeratin 19, PCNA and collagen I by the parallel activation of Wnt4/β-catenin and AKT signaling [38].

CONCLUSION

Nowadays, more and more new methods have been developed for different types of wounds. Stem cell-based therapy is one of the promising methods which have been widely used in recent years. Variety of stem cells can be used in this era specifically for reducing scars following wound healing [60]. Beside these advantages of cell therapy, it also has some serious limitations like tumorigenicity and immunogenicity. To overcome these limitations, cell-free therapies have been developed by scientists in recent years which demonstrated interesting therapeutic effects. One of the considerable cell-free methods is using exosomes which can be extracted from different sources. Exosomes that contain DNA, protein, siRNA, mRNA and miRNA can moderate and regulate gene expression in target cells [41]. Moreover, using exosomes avoids many risks associated with cell transplantation. Therefore, MSC-exosomes may be safer and more efficient than whole cell, but more preclinical and clinical studies are still needed to reveal unknown aspects of exosomes and their therapeutic effects.

FUNDING

This work was supported by National Natural Science Foundation of China (No. 81772075).

COMPETING INTEREST

The authors declare that they have no competing interests.

REFERENCES

  1. Childs DR, Murthy AS (2017) Overview of Wound Healing and Management. The Surgical clinics of North America 97: 189-207.
  2. You H-J, Han SK (2014) Cell therapy for wound healing. J Korean Med Sci 29: 311-319.
  3. Baron JM, Glatz M, Proksch E (2020) Optimal Support of Wound Healing: New Insights. Dermatology 17: 1-8.
  4. Crippa S, Santi L, Bosotti R, Porro G, Bernardo ME (2019) Bone Marrow-Derived Mesenchymal Stromal Cells: A Novel Target to Optimize Hematopoietic Stem Cell Transplantation Protocols in Hematological Malignancies and Rare Genetic Disorders. Journal of clinical medicine 9: 2.
  5. Liao Y, Li G, Zhang X, Huang W, Xie D, et al. (2020) Cardiac Nestin+ Mesenchymal Stromal Cells Enhance Healing of Ischemic Heart through Periostin-Mediated M2 Macrophage Polarization. Mol Ther.
  6. Huang KC, Chuang MH, Lin ZS, Lin YC, Chen CH, et al. (2019) Transplantation with GXHPC1 for Liver Cirrhosis: Phase 1 Trial. Cell Transplant 28: 100-111.
  7. Forostyak S, Jendelova P, Sykova E (2013) The role of mesenchymal stromal cells in spinal cord injury, regenerative medicine and possible clinical applications. Biochimie 95: 2257-2270.
  8. Olivos-Meza A, Pérez Jiménez FJ, Granados-Montiel J, Landa-Solís C, Cortés González S, et al, (2019) First Clinical Application of Polyurethane Meniscal Scaffolds with Mesenchymal Stem Cells and Assessment of Cartilage Quality with T2 Mapping at 12 Months. Cartilage.
  9. Baharlooi H, Azimi M, Salehi Z, Izad M (2019) Mesenchymal Stem Cell-Derived Exosomes: A Promising Therapeutic Ace Card to Address Autoimmune Diseases. Int J Stem Cells.
  10. Wang X, Jiao Y, Pan Y, Zhang L, Gong H, et al. (2019) Fetal Dermal Mesenchymal Stem Cell-Derived Exosomes Accelerate Cutaneous Wound Healing by Activating Notch Signaling. Stem cells international 2019: 2402916.
  11. Zhang S, Chen L, Zhang G, Zhang B (2020) Umbilical cord-matrix stem cells induce the functional restoration of vascular endothelial cells and enhance skin wound healing in diabetic mice via the polarized macrophages. Stem cell research & therapy 11: 39.
  12. Friedenstein AJ, Petrakova KV, Kurolesova AI, Frolova GP (1968) Heterotopic of bone marrow. Analysis of precursor cells for osteogenic and hematopoietic tissues. Transplantation 6: 230-247.
  13. Pittenger MF, Mackay AM, Beck SC, Jaiswal RK, Douglas R, et al. (1999) Multilineage potential of adult human mesenchymal stem cells. Science 284: 143-147.
  14. da Silva Meirelles L, Chagastelles PC, Nardi NB (2006) Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J Cell Sci 119: 2204-2213.
  15. Jiang Y, Jahagirdar BN, Reinhardt RL, Schwartz RE, Keene CD, et al. (2002) Pluripotency of mesenchymal stem cells derived from adult marrow. Nature 418: 41-49.
  16. Fu X, Fang L, Li X, Cheng B, Sheng Z (2006) Enhanced wound-healing quality with bone marrow mesenchymal stem cells autografting after skin injury. Wound Repair Regen 14: 325-335.
  17. Dominici M, Le Blanc K, Mueller I, Slaper-Cortenbach I, Marini F, et al. (2006) Minimal criteria for defining multipotent mesenchymal stromal cells. The International Society for Cellular Therapy position statement. Cytotherapy 8: 315-317.
  18. Sivan U, Jayakumar K, Krishnan LK (2014) Constitution of fibrin-based niche for in vitro differentiation of adipose-derived mesenchymal stem cells to keratinocytes. Biores Open Access 3: 339-347.
  19. Irfan-Maqsood M, Matin MM, Heirani-Tabasi A, Bahrami M, Naderi-Meshkin H, et al. (2016) Adipose derived mesenchymal stem cells express keratinocyte lineage markers in a co-culture model. Cell Mol Biol (Noisy-le-grand) 62: 44-54.
  20. Li H, Fu X, Ouyang Y, Cai C, Wang J, et al. (2006) Adult bone-marrow-derived mesenchymal stem cells contribute to wound healing of skin appendages. Cell Tissue Res 326: 725-736.
  21. Sasaki M, Abe R, Fujita Y, Ando S, Inokuma D, et al. (2008) Mesenchymal stem cells are recruited into wounded skin and contribute to wound repair by transdifferentiation into multiple skin cell type. J Immunol 180: 2581-2587.
  22. Falanga V1, Iwamoto S, Chartier M, Yufit T, Butmarc J, et al. (2007) Autologous bone marrow-derived cultured mesenchymal stem cells delivered in a fibrin spray accelerate healing in murine and human cutaneous wounds. Tissue Eng 13: 1299-1312.
  23. Badiavas EV, Abedi M, Butmarc J, Falanga V, Quesenberry P (2003) Participation of bone marrow derived cells in cutaneous wound healing. J Cell Physiol 196: 245-250.
  24. Atalay S, Coruh A, Deniz K (2014) Stromal vascular fraction improves deep partial thickness burn wound healing. Burns 4: 1375-1383.
  25. Nie C, Yang D, Xu J, Si Z, Jin X, et al. (2011) Locally administered adipose-derived stem cells accelerate wound healing through differentiation and vasculogenesis. Cell transplant 20: 205-216.
  26. Brown JM, Nemeth K, Kushnir-Sukhov NM, Metcalfe DD, Mezey E (2011) Bone marrow stromal cells inhibit mast cell function via a COX2-dependent mechanism. Clin Exp Allergy 41: 526-534.
  27. Francois S, Mouiseddine M, Allenet-Lepage B, Voswinkel J, Douay L, et al. (2013) Human mesenchymal stem cells provide protection against radiation-induced liver injury by antioxidative process, vasculature protection, hepatocyte differentiation, and trophic effects. Biomed Res Int: 151679.
  28. Chen L, Tredget EE, Wu PYG, Wu Y (2008) Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One 3: 1886.
  29. Hocking AM, Gibran NS (2010) Mesenchymal stem cells: paracrine signaling and differentiation during cutaneous wound repair. Exp Cell Res 316: 2213-2219.
  30. Walter MN, Wright KT, Fuller HR, MacNeil S, Johnson WE (2010) Mesenchymal stem cell-conditioned medium accelerates skin wound healing: an in vitro study of fibroblast and keratinocyte scratch assays. Exp Cell Res 316: 1271-1281.
  31. Rodriguez-Menocal L, Salgado M, Ford D, Van Badiavas E (2012) Stimulation of skin and wound fibroblast migration by mesenchymal stem cells derived from normal donors and chronic wound patients. Stem Cells Transl Med 1: 221-229.
  32. Shabbir A, Cox A, Rodriguez-Menocal L, Salgado M, Van Badiavas E (2015) Mesenchymal Stem Cell Exosomes Induce Proliferation and Migration of Normal and Chronic Wound Fibroblasts, and Enhance Angiogenesis In Vitro. Stem Cells Dev 24: 1635-1647.
  33. Kniazeva E, Kachgal S, Putnam AJ (2011) Effects of extracellular matrix density and mesenchymal stem cells on neovascularization in vivo. Tissue Eng Part A 17: 905-914.
  34. Caplan AI, Correa D (2011) The MSC: an injury drugstore. Cell Stem Cell 9: 11-15.
  35. Pittenger M (2009) Sleuthing the source of regeneration by MSCs. Cell Stem Cell 5: 8-10.
  36. Ortiz LA, Dutreil M, Fattman C, Pandey AC, Torres G, et al. (2007) Interleukin 1 receptor antagonist mediates the antiinflammatory and antifibrotic effect of mesenchymal stem cells during lung injury. Proc Natl Acad Sci U S A 104: 11002-11007.
  37. Kusindarta DL, Wihadmadyatami H, Fibrianto YH, Nugroho WS, Susetya H, et al. (2016) Human umbilical mesenchymal stem cells conditioned medium promote primary wound healing regeneration. Vet World 9: 605-610.
  38. Zhang B, Wang M, Gong A, Zhang X, Wu X, et al., HucMSC-Exosome Mediated-Wnt4 Signaling Is Required for Cutaneous Wound Healing. Stem Cells 33: 2158-2168.
  39. Hu L, Wang J, Zhou X, Xiong Z, Zhao J, et al. (2016) Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci Rep 6: 32993.
  40. Zhang J, Guan J, Niu X, Hu G, Guo S, et al. (2015) Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. J Transl Med 13: 49.
  41. Fang S, Xu C, Zhang Y, Xue C, Yang C, et al. (2016) Umbilical Cord-Derived Mesenchymal Stem Cell-Derived Exosomal MicroRNAs Suppress Myofibroblast Differentiation by Inhibiting the Transforming Growth Factor-β/SMAD2 Pathway During Wound Healing. Stem Cells Transl Med 5: 1425-1439.
  42. Zhou Y, Zhou G, Tian C, Jiang W, Jin L, et al. (2016) Exosome-mediated small RNA delivery for gene therapy. Wiley Interdiscip Rev RNA 7: 758-771.
  43. Torreggiani E, Perut F, Roncuzzi L, Zini N, Baglìo SR, et al. (2014) Exosomes: novel effectors of human platelet lysate activity. Eur Cell Mater 28: 137-151.
  44. Lu K, Li HY, Yang K, Wu JL, Cai XW, et al. (2017) Exosomes as potential alternatives to stem cell therapy for intervertebral disc degeneration: in-vitro study on exosomes in interaction of nucleus pulposus cells and bone marrow mesenchymal stem cells. Stem Cell Res Ther 8: 108.
  45. Cabral J, Ryan AE, Griffin MD, Ritter T (2018) Extracellular vesicles as modulators of wound healing. Adv Drug Deliv Rev 129: 394-406.
  46. Silva AM, Teixeira JH, Almeida MI, Gonçalves RM, Barbosa MA, et al. (2017) Extracellular Vesicles: Immunomodulatory messengers in the context of tissue repair/regeneration. Eur J Pharm Sci 98: 86-95.
  47. Wu P, Zhang B, Shi H, Qian H, Xu W (2018) MSC-exosome: A novel cell-free therapy for cutaneous regeneration. Cytotherapy 20: 291-301.
  48. Huang L, Ma W, Ma Y, Feng D, Chen H, et al. (2015) Exosomes in mesenchymal stem cells, a new therapeutic strategy for cardiovascular diseases? Int J Biol Sci 11: 238-245.
  49. Schwab A, Meyering SS, Lepene B, Iordanskiy S, van Hoek ML, et al. (2015) Extracellular vesicles from infected cells: potential for direct pathogenesis. Front Microbiol 6: 1132.
  50. Rani S, Ritter T (2016) The Exosome - A Naturally Secreted Nanoparticle and its Application to Wound Healing. Adv Mater 28: 5542-5552.
  51. Zhao B, Zhang Y, Han S, Zhang W, Zhou Q, et al. (2017) Exosomes derived from human amniotic epithelial cells accelerate wound healing and inhibit scar formation. J Mol Histol 48: 121-132.
  52. Toh WS, Zhang B, Lai RC, Lim SK (2018) Immune regulatory targets of mesenchymal stromal cell exosomes/small extracellular vesicles in tissue regeneration. Cytotherapy 20: 1419-1426.
  53. He X, Dong Z, Cao Y, Wang H, Liu S, et al. (2019) MSC-Derived Exosome Promotes M2 Polarization and Enhances Cutaneous Wound Healing. Stem Cells Int: 7132708.
  54. Monguió-Tortajada M, Roura S, Gálvez-Montón C, Pujal JM, Aran G, et al. (2017) Nanosized UCMSC-derived extracellular vesicles but not conditioned medium exclusively inhibit the inflammatory response of stimulated T cells: implications for nanomedicine. Theranostics 7: 270-284.
  55. Zlotogorski-Hurvitz A, Dayan D, Chaushu G, Salo T3, Vered M (2016) Morphological and molecular features of oral fluid-derived exosomes: oral cancer patients versus healthy individuals. J Cancer Res Clin Oncol 142: 101-110.
  56. Sun L, Xu R, Sun X, Duan Y, Han Y, et al. (2016) Safety evaluation of exosomes derived from human umbilical cord mesenchymal stromal cell. Cytotherapy 18: 413-422.
  57. Liang X, Zhang L, Wang S, Han Q, Zhao RC (2016) Exosomes secreted by mesenchymal stem cells promote endothelial cell angiogenesis by transferring miR-125a. J Cell Sci 129: 2182-2189.
  58. Kang T, Jones TM, Naddell C, Bacanamwo M, Calvert JW, et al. (2016) Adipose-Derived Stem Cells Induce Angiogenesis via Microvesicle Transport of miRNA-31. Stem Cells Transl Med 5: 440-50.
  59. Zhang B, Shi Y, Gong A, Pan Z, Shi H, et al. (2016) HucMSC Exosome-Delivered 14-3-3ζ Orchestrates Self-Control of the Wnt Response via Modulation of YAP During Cutaneous Regeneration. Stem Cells 34: 2485-2500.
  60. Han G, Ceilley R (2017) Chronic Wound Healing: A Review of Current Management and Treatments. Adv Ther 34: 599-610.

Citation: Zhu G, Gu H, Wu M (2020) Mesenchymal Stem Cells, Exosomes and Cutaneous Wound Healing. J Stem Cell Res Dev Ther 6: 028.

Copyright: © 2020  Guanghao Zhu, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.


Herald Scholarly Open Access is a leading, internationally publishing house in the fields of Sciences. Our mission is to provide an access to knowledge globally.



© 2024, Copyrights Herald Scholarly Open Access. All Rights Reserved!