Minireviews Open Access
Copyright ©The Author(s) 2021. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Aug 6, 2021; 9(22): 6218-6233
Published online Aug 6, 2021. doi: 10.12998/wjcc.v9.i22.6218
Mesenchymal stem cell-derived exosomes: An emerging therapeutic strategy for normal and chronic wound healing
Qin-Lu Zeng, De-Wu Liu, Burns Institute, The First Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi Province, China
Qin-Lu Zeng, First Clinical Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, China
ORCID number: Qin-Lu Zeng (0000-0001-9602-9622); De-Wu Liu (0000-0001-6617-8715).
Author contributions: Zeng QL contributed to the investigation, data curation, and original draft preparation; Zeng QL and Liu DW contributed to manuscript writing, review, and editing; Liu DW supervised this study.
Supported by National Natural Science Foundation of China, No. 81460293; and Innovation and Entrepreneurship Training Program of Nanchang University, China, No. 20190402184.
Conflict-of-interest statement: The authors declare that there is no conflict of interest related to this manuscript.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: De-Wu Liu, MD, PhD, Chief Doctor, Professor, Burns Institute, The First Affiliated Hospital of Nanchang University, No. 17 Yongwaizheng Street, Donghu District, Nanchang 330006, Jiangxi Province, China. dewuliu@126.com
Received: February 8, 2021
Peer-review started: February 8, 2021
First decision: March 30, 2021
Revised: April 8, 2021
Accepted: June 22, 2021
Article in press: June 22, 2021
Published online: August 6, 2021

Abstract

Skin wound healing is a complex biological process. Mesenchymal stem cells (MSCs) play an important role in skin wound repair due to their multidirectional differentiation potential, hematopoietic support, promotion of stem cell implantation, self-replication, and immune regulation. Exosomes are vesicles with diameters of 40-100 nm that contain nucleic acids, proteins, and lipids and often act as mediators of cell-to-cell communication. Currently, many clinical scientists have carried out cell-free therapy for skin wounds, especially chronic wounds, using exosomes derived from MSCs. This review focuses on the latest research progress on the mechanisms of action associated with the treatment of wound healing with exosomes derived from different MSCs, the latest research progress on the combination of exosomes and other biological or nonbiological factors for the treatment of chronic skin wounds, and the new prospects and development goals of cell-free therapy.

Key Words: Mesenchymal stem cells, Exosomes, Mesenchymal stem cell-exosomes, Wound healing, Therapeutics

Core Tip: We have mainly sorted out the reported mechanisms by which different mesenchymal stem cells (MSC)-derived exosomes play a promoting role in the hemostatic stage, inflammatory stage, proliferative stage, and remodeling stage of skin wound healing. The clinical prospect of MSC-exosomes as a cell-free therapy, such as the addition of carriers, combination of drugs, combined physical treatment, and development as an immunosuppressive agent, is also summarized.



INTRODUCTION

Skin wound healing is a complex biological process that includes cell proliferation, differentiation, epithelization, migration, and matrix synthesis and deposition. Mesenchymal stem cells (MSCs) are important members of the stem cell family and come from many sources, such as adipose tissue, dental pulp, placenta, amniotic fluid, and umbilical cord blood[1]; these cells have the common characteristics of self-replication and multidirectional differentiation. The mechanism of action of stem cell therapy has been demonstrated to primarily involve paracrine actions mediated by stem cell secretion factors[2,3]. It has been reported that exosomes play a major paracrine role among certain secretory bodies associated with stem cells[3-5]. The small vesicles formed by reverse budding are called exosomes after they are released from cells[6], and these vesicles contain nucleic acids, proteins, and lipids and often serve as mediators of communication between cells. Currently, many clinical scientists have carried out research on these small exosomes. The commercial use of cell-free therapy based on exosomes has already begun[7-9]. In fact, in various disease models, including cardiovascular disease, respiratory disease, liver disease, kidney disease, nerve disease, musculoskeletal disease, eye disease, skin diseases, and cancer, MSC-derived exosomes have been used as cell-free substitutes for MSCs[4,10-13]. In the field of skin wound healing, researchers have attempted to explore the mechanism by which exosomes promote wound healing while developing cell-free therapy to promote the healing of normal skin wounds and chronic refractory wounds. Researchers have used different exosomes from MSCs to study the four stages of wound healing (hemostasis, inflammation, proliferation, and remodeling) from different angles and have made good progress.

BIOGENESIS AND BIOLOGICAL CHARACTERISTICS OF MSC-EXOSOMES

MSCs are described as pluripotent nonhematopoietic adult stem cells that express the surface markers CD73, CD90, and CD105 but do not express CD14, CD34, or CD45[1]. MSCs were first discovered by Freeden Stein in the 1960s through studies of bone marrow[14]. MSCs can also be isolated from other adult tissues, such as adipose tissue, dental pulp, placenta, amniotic fluid, umbilical cord blood, Wharton’s jelly[1], and even the brain, spleen, liver, kidney, lung, thymus, and pancreas.

In 1983, the secretion of extracellular vesicles (EVs) was identified during the maturation of reticulocytes[15]. EVs are a general term that is currently used to refer to all secreted membrane vesicles. However, these vesicles are highly heterogeneous and are cell-derived membrane-bound structures secreted by various cell types, including T cells, B cells, dendritic cells, platelets, mast cells, epithelial cells, endothelial cells, neurons, cancer cells, oligodendrocytes, Schwann cells, embryonic cells, and MSCs[16].

EVs, including exosomes, microvesicles, and apoptotic bodies[17-19], can be found in physiological fluids such as normal urine, blood, bronchial lavage fluid, breast milk, saliva, cerebrospinal fluid, amniotic fluid, synovial fluid, and malignant ascites. Exosomes play a relatively major role among EVs[15,16].

Exosomes are 40-100 nm diameter membranous vesicles of endocytic origin that are released by a variety of cell types into the extracellular space[20]. Exosomes were first reported in 1983 by Johnstone and colleagues while culturing reticulocytes. Inward budding of endosomal membranes results in the progressive accumulation of intraluminal vesicles (ILVs) within large multivesicular bodies (MVBs). Transmembrane proteins are incorporated into the invaginating membrane, while cytosolic components are engulfed within ILVs[21].

Part of the MVB is degraded after entering the lysosome; the remaining part sags inward again to form small granular vesicles by fusing with the plasma membrane and then releasing these vesicles into the extracellular environment. These vesicles that are released from the cell are called exosomes[6].

The biological characteristics of exosomes are mainly reflected in three aspects. First, exosomes function to transport material and transmit information. As a component of the intercellular microenvironment, exosomes regulate cell-to-cell communication by carrying a diverse array of signaling molecules, including lipids, proteins, and nucleic acids. Studies have shown that intercellular communication appeared during the early stage of evolution and could influence the behavior of target cells in many ways[22]. After the exosomes are absorbed by the target cells, the components such as lipids, proteins, mRNA, and microRNAs in the exosomes can affect the modification and positioning of proteins by changing the transcription and translation procedures, and ultimately regulate the cell phenotype and function of the receptor cells by regulating signal cascade pathways and key enzyme reactions to affect cell self-regulation, as well as directly participating in various physicochemical reactions in cells. Second, exosomes act as tools for transporting and removing components from cells. In addition, exosomes are also involved in immunoregulation. In vitro experiments have shown that exosomes can inhibit the proliferation and differentiation of T cells and reduce the release of IFN-γ from T cells[23].

PHYSIOLOGICAL AND PATHOLOGICAL PROCESSES OF WOUND HEALING
Normal cutaneous wound healing

The normal cutaneous wound healing process includes four stages: Hemostasis, inflammation, proliferation, and remodeling[24].

Hemostasis occurs immediately after an injury. Vasoconstriction and platelet aggregation proceed simultaneously. A blood clot covering the wound will form at the wound site. This clot not only reduces blood loss but also provides a scaffold-like structure for the migration of resident skin cells and immune cells[25]. Additionally, platelets in blood clots secrete or release various platelet-specific proteins; growth factors; adhesion molecules; fibrinogen and thrombin; pro- and antiangiogenic factors; and cytokines/chemokines such as NAP-2 and platelet-derived SDF-1α[26-28]. Subsequently, neutrophils, macrophages, fibroblasts, endothelial cells, smooth muscle cells, and circulating BMSCs can be induced by these secreted molecules to migrate to the wound site and become activated.

Inflammation is driven by secreted chemokines/cytokines, bacterial byproducts, and platelet-derived mediators. After vascular permeability increased, monocytes/ macrophages and neutrophils gradually infiltrated the wound surface. The cells not only kill bacteria but also remove debris and damaged matrix proteins[29]. After reaching the wound, neutrophils release proteases such as MMP, ROS, growth factors, and antimicrobial peptides, while monocytes arrive within 24 hours and transform into M1 macrophages. Lymphocytes are the last inflammatory cells that are attracted to the wound site. Among them, γδ+ T cells can participate in the growth and survival of fibroblasts, immune cells, and keratinocytes by producing mediators such as IGF-1, FGFs, or KGFs. αβ+ T cells are also present in this inflammatory stage and have an important effect on pathogenic microorganisms[26,30,31].

In the later stage of inflammation, M2 macrophages transform from M1 macrophages or monocytes. On the one hand, the release of IL-10, VEGF, PDGF, FGFs, and IGF-1 by M2 macrophages plays a role in inducing proliferation, cell migration, and matrix formation. On the other hand, these cells also produce TIMP1 to counteract MMPs[32].

The focus of the proliferation phase is the development of granulation tissue, which covering the exposed wound surface while helps to restore the vascular network[33]. At this stage, fibroblasts can play a role in reducing the interstitial space by depositing a large amount of ECM. This process is affected by cytokines and growth factors, and the regulation of these factors can also induce the release of additional cytokines such as VEGF, FGFs, IGFs, IFNs, and HGF[24,28,34,35]. In addition, keratinocytes also release growth factors. Keratinocytes proliferate and migrate to facilitate wound coverage, form layers and differentiate, ultimately achieving the effect of rebuilding the epidermal barrier of the skin[24].

Remodeling is the final stage and is characterized by a gradual decrease in cells and blood vessels[29]. Fibroblasts are the main factors at this stage, and their main role is to increase the amount of collagen I and other extracellular matrix (ECM) components, while the role of MMPs is to decompose the disordered collagen that was used as a template, similar to the more common collagen III[24,36,37]. The decomposition of granulation tissue and the formation of scars are driven by a variety of MMPs and their inhibitors. Any disturbance in the balance of matrix metalloproteinases and their inhibitors can trigger the development of hypertrophic scars or the formation of chronic wounds.

Chronic wound healing

When tissue repair is ineffective, the skin may suffer from ulcerative injuries, such as venous ulcers in the lower limbs, pressure ulcers, and diabetic foot ulcers[30]. Although there is widespread controversy, wounds lasting more than 3 mo are generally considered chronic wounds[38].

Persistent inflammation is an important feature of chronic wounds, which manifests as dysregulated cytokine/growth factor levels and/or increased protease activity, impaired angiogenesis, and difficult re-epithelialization of the wound[38]. At the cellular level, many disorders occur in neutrophils and macrophages in chronic wounds. Neutrophils show phenotypic changes, reduced infiltration and migration across endothelial cells, and stay in the wound for longer periods of time. In addition, chronic wounds also showed reduced induction of the M2 macrophage profile and reduced antibacterial activity[39,40]. The function of macrophages in chronic wounds is also disturbed and may cause abnormal repair. This effect is specifically reflected in the uncontrolled production of inflammatory mediators and growth factors by macrophages, as well as the imbalance in the M1:M2 profile ratio. There is also communication failure between macrophages and other crucial participants (including fibroblasts, epithelial cells, endothelial cells, and stem or tissue progenitor cells)[41].

CUTANEOUS WOUND HEALING BY MSC-EXOSOMES

According to the existing literature, MSC-exosomes or MSC-EVs can accelerate wound healing at various phases of wound healing, and even have the ability of improving scars (Table 1).

Table 1 Effects of mesenchymal stem cell-exosomes on cutaneous wound healing.
Phase
Exosome source
Nomenclature
Related exosomal cargo
Secreted factors or expressed genes affected
Outcome
Ref.
Hemostasis PhaseHuman mesenchymal stem cells (MSCs) from the umbilical cordEVs-Phosphatidylserine(+)Umbilical MSCs and extracellular vesicles derived from them have a reasonably high procoagulant potential[42]
Inflammatory PhaseHuman jaw bone marrow-derived MSCs and bone marrow MSCsExosomesmiR-223TNF-α ↓ IL-10 ↑Accelerated wound healing in mice[46]
Induced M2 macrophage polarization (CD206+ macrophage ↑)
Human umbilical cord (UC)-MSCsExosomeslet-7bTLR4, p-p65, iNOS ↓ p-STAT3, p-AKT, ARG1 ↑Alleviated inflammation and enhanced diabetic cutaneous wound healing in rats[47]
Induced M2 macrophage polarization Inhibited TLR4 signaling pathway
Human UC-MSCsExosomesmiR-181cTNF-α, IL-1β, TLR4, p65, p-p65↓ IL-10 ↑Reduced burn-induced inflammation in rats [48]
Reduced neutrophil and macrophage infiltration (MPO+ cell,CD68+ cell↓) Inhibited TLR4 signaling pathway
Human menstrual blood derived MSCs (MenSCs)Exosomes-iNOS ↓ ARG1, VEGF ↑Resolved inflammation and ameliorate cutaneous non healing wounds in diabetic mice[49]
Induced M2 macrophage polarization
Proliferative PhaseHuman bone marrow MSC-derived exosomesExosomesTGF-β/SmadTGF-β1, Smad2, Smad3, Smad4 ↓TGF-β3, Smad7↑Effectively promoted the cutaneous wound healing by inhibiting the TGF-β/Smad signal pathway[59]
Human adipose MSCs (ASCs)Exosomes-N-cadherin, cyclin 1, PCNA, collagen I/III, elastin ↑Facilitated cutaneous wound healing via optimizing the characteristics of fibroblasts[62]
Human ASCsExosomes-Collagen I/II, TGF-β1/3, MMP1/3 α-SMA ↓Promoted ECM reconstruction in cutaneous wound repair by regulating the ratios of collagen type III: type I, TGF-β3:TGF-β1, and MMP3:TIMP1, and by regulating fibroblast differentiation to mitigate scar formation[63]
Human fetal dermal MSCsExosomesJagged 1Collagen I/III, elastin, fibronectin mRNA ↑Promoted wound healing by activating the ADF cell motility and secretion ability via the Notch signaling pathway[64]
Human UC-MSCsExosomesWnt4CK19, PCNA, collagen I ↑Stimulated the AKT pathway to protect immortalized keratinocytes from heat-induced apoptosis[65]
Stimulated the AKT pathway to protect immortalized keratinocytes from heat-induced apoptosis
Human UC-MSCsExosomesAkt, ERK, STAT3HGF, IGF1, NGF, SDF1↑Promoted the proliferation and migration of fibroblasts in normal and chronic wounds. This effect was positively correlated with the dose of exosomes[66]
Induced pluripotent stem cell-derived MSCs Exosomes-Collagen ↑Increased the secretion of collagen by HaCaT cells to accelerate skin cell proliferation[67]
Adipose mesenchymal stem cells (ADSCs)ExosomesAKT/HIF-1α-Promoted the proliferation and migration of HaCaT cells by regulating the activation of the AKT/HIF-1α signaling pathway, thus promoting wound healing[68]
Human UC-MSCsExosomes-PARP-1, PAR↑Suppressed HaCaT cell apoptosis induced by H2O2 by restraining the nuclear translocation of apoptosis-inducing factor (AIF) and promoting poly (ADP-ribose) (PAR) and poly ADP ribose polymerase 1 (PARP-1) expression[69]
Human adipose-derived MSCs (adMSC-Exo)ExosomesmiR-125aAngiogenic inhibitor delta-like 4 (DLL4)↓Transferred miR-125a to endothelial cells and promoted angiogenesis by repressing DLL4[70]
Mouse BM- MSCsExosomesmiR-17 miR-23a miR-125bTNF-α, IL-1β, iNOS, TLR4, IRAK1, p65↓ ARG1, IL-10, TGF-β↑Decreased the threshold for thermal and mechanical stimuli in mice[71]
Increased nerve conduction velocity, the number of intraepidermal nerve fibers, myelin thickness, and axonal diameters
Rat BM-MSCsExosomes-MDA, HIF1α, NOX2, Caspase 3, BAX, PARP1, MPO, ICAM1, IL-1β, NF-κB↓SOD, CAT, GPX, HO-1, BCL2, IL-10, bFGF, HGF, SOX9, VEGF↑Decreased histopathological score of kidney injury in rats[72]
Reduced the levels of blood urea nitrogen (BUN) and creatinineReduced the level of oxidative stress
Increased anti-oxidant status
Reduced apoptosis and inflammation
Improved regeneration and enhanced angiogenesis
Human endometrial MSCsExosomes-Tie2, VEGF, Ang1, Ang2↑Increased the expression of angiogenesis markers, including Tie2, VEGF, Ang1, and Ang2, and increased the proliferation, migration, and angiogenesis of HUVECs[73]
Human umbilical cord mesenchymal stem cells (hUCMSCs)Exosomes-Ang2↑hucMSC-Ex-derived Ang-2 plays a significant role in tube formation of HUVECs and promotion of angiogenesis[74]
Human UC blood-MSCsExosomes-Ang, Ang1, HFG, VEGF↑Human umbilical cord blood (UCB)-MSC-derived exosomes pretreated with thrombin could accelerate skin wound healing in rats with full-thickness wounds. Exosomes from human UCB-MSCs increased angiogenesis factors, such as VEGF, HGF, and Ang1, and decreased TNFα and IL-6[75]
Human UC-MSCsExosomesWnt4β-catenin, N-cadherin, PCNA, Cyclin D3↑Enhanced angiogenesis in rats through the Wnt4/β-catenin pathway. When the expression of Wnt4 was knocked out by shRNA, the proangiogenic effect of hUC-MSC-derived exosomes was eliminated[76]
Human UC-MSCsExosomes-α-SMA, collagen I↓Increased the formation and maturation of new blood vessels at the wound site, although the mechanism is still unclear[77]
Human UC-MSCsExosomesGSK3β-Wnt/β-catenin-Alleviated hepatic IRI by transporting miR-1246 via regulating GSK3β-mediated Wnt/β-catenin pathway[78]
Remodeling PhaseHuman gingival MSCsExosomes-Collagen↑Reduced the formation of scars by inhibiting the accumulation of mouse myofibroblasts[79]
Adipose mesenchymal stem cells (ASCs)Exosomes-N-cadherin, cyclin-1, PCNA collagen I, III↑Facilitates cutaneous wound healing via optimizing the characteristics of fibroblasts[62]
ASCsExosomesERK/MAPKMatrix metalloproteinases-3 (MMP3)↑APromoted ECM reconstruction in cutaneous wound repair by regulating the ratios of collagen type III: type I, TGF-β3:TGF-β1, and MMP3:TIMP1, and by regulating fibroblast differentiation to mitigate scar formation[63]
MenSCsExosomes-iNOS↓ ARG1, VEGF↑Resolved inflammation and ameliorated cutaneous non-healing wounds in diabetic mice[49]
Induced M2 macrophage polarization
Hemostasis phase

During the initial hemostatic stage of wound healing, the physiological mechanism for restoring skin barrier function mainly involves reducing blood loss through vasoconstriction and platelet aggregation in blood clots. As mentioned previously, platelets in blood clots can induce the migration of a series of cells to the wound site by releasing specific proteins. To date, there is no direct evidence that MSC-derived exosomes are involved in hemostasis or blood clotting. According to reports, human umbilical cord (hUC)-MSC-EVs can induce blood coagulation in vitro[42]. This result indicates that MSC-derived exosomes may have potential benefits in the coagulation process of wound healing, but further studies are needed to analyze the role of MSC-EVs or MSC-derived exosomes in the coagulation process under healthy or diseased conditions.

Inflammatory phase

The inflammatory stage is very important in the process of skin repair. As mentioned previously, this stage mainly relies on the infiltration of neutrophils and monocytes/ macrophages at the wound site to complete tasks such as killing bacteria and removing debris. In late inflammation, with the release of M2-type macrophages and other mediators, wound healing transitions from the inflammatory phase to the proliferative phase, which is a critical step during normal wound healing[43]. However, studies have shown that prolonged inflammation can lead to excessive scarring of the wound[37], mainly in burns and other chronic wounds[44,45]. Based on this mechanism in the inflammatory phase of wound healing, we believe that macrophages play a crucial role in the later stages of inflammation, especially through proper transformation of M1-type macrophages into M2-type macrophages. A series of studies have shown that MSC-derived exosomes are involved in the promotion of macrophage polarization through a variety of regulatory mechanisms. This phenomenon is critical for transitioning from the inflammatory phase to the proliferative phase during the wound healing process, especially for the promotion of chronic wound healing: (1) MiR-223 produced by human bone marrow MSC (hBM-MSC)-derived exosomes and jaw bone marrow MSC-derived exosomes regulates the polarization of macrophages by targeting pknox1, thereby promoting the healing and metastasis of skin wounds in mice[46]; (2) Lipopolysaccharide (LPS)-preconditioned exosomes may mediate the regulation of macrophage polarization and chronic inflammation regression by shuttling let-7b, thus promoting the healing of diabetic skin wounds[47]; (3) Burns significantly increased the inflammatory response in rats or macrophages exposed to LPS, while hUC-MSC-derived exosomes overexpressing miR-181c inhibited the TLR4 signaling pathway effectively, thus reducing inflammation in burned rats[48]; (4) The miRNA let-7b carried by MSC-EXOs pretreated with LPS can regulate the polarization of macrophages by inhibiting the TLR4/NF-κB pathway and activating the STAT3/AKT signaling pathway, thereby promoting wound healing[47]; and (5) Menstrual blood-derived MSC exosomes can resolve inflammation by inducing M1–M2 macrophage polarization[49].

In fact, there have been a large number of reports on the anti-inflammatory and immunoregulatory effects of MSC-derived exosomes[18,50-58].

Proliferative phase

In the proliferation stage of wound healing, whether MSC-derived exosomes can promote the development of granulation tissue and the recovery of the vascular network is a concern. Studies have shown that hBM-MSC-derived exosomes could effectively promote the proliferation of two types of skin cells in vitro: Human dermal fibroblasts (HDFs) and human keratinocytes (HaCaTs). In vivo, hBM-MSC-derived exosomes could accelerate skin wound healing by inhibiting the TGF-β/Smad signaling pathway[59]. This pathway is critical in the pathogenesis of wound healing[60]. TGF-β1 seems to be the key mediator of tissue scarring and mainly antagonizes acellular (Smad) signal transduction by activating its downstream factors[61].

According to related studies, MSC-derived exosomes are able to promote the migration and proliferation of dermal fibroblasts and can produce collagen, elastin, and fibronectin: (1) Human adipose MSC (ASC)-derived EVs or ASC-derived exosomes induce the migration and proliferation of dermal fibroblasts or keratinocytes in vitro[62,63]; (2) Human ASC-derived exosomes induce type I/III collagen and elastin production in HDFs to promote the healing of skin wounds in mice[62,63]; (3) Human fetal dermis (FD)-MSC-derived exosomes activate the Notch pathway by transmitting Jagged1 protein and induce the mRNA expression of collagen I/III, elastin, and fibronectin[64]; (4) hUC-MSC-derived exosomes containing Wnt4 accelerated the re-epithelialization of burned skin in rats. The wound healing effect was inhibited when the expression of Wnt4 in hUC-MSC-derived exosomes was knocked out with siRNA[65]; and (5) hUC-MSC-exosomes can promote the proliferation and migration of fibroblasts in normal and chronic wounds. This effect was positively correlated with the dose of exosomes[66].

The positive effects of MSC-derived exosomes on keratinocytes have also been reported: (1) hUC-MSC-derived exosomes can stimulate the AKT pathway to protect immortalized keratinocytes from heat-induced apoptosis[65]; (2) Induced pluripotent stem cell (iPSC)-derived MSC (iMSC) exosomes could increase the secretion of collagen by HaCaT cells to accelerate skin cell proliferation[67]; (3) ASC-derived exosomes activate the AKT/HIF-1α axis to accelerate the migration and proliferation of keratinocytes to promote wound healing[68]; and (4) Human umbilical cord MSC (hUCMSC)-derived exosome treatment suppressed HaCaT cell apoptosis induced by H2O2 by restraining the nuclear translocation of apoptosis-inducing factor (AIF) and promoting poly (ADP-ribose) (PAR) and poly ADP ribose polymerase 1 (PARP-1) expression[69].

Regarding the recovery of the vascular network, it has also been reported that MSC-derived exosomes can induce angiogenic activity in endothelial cells: (1) Human ASC-derived exosomes induce HUVEC tube formation by delivering miR-125a, thereby inhibiting the expression of the angiogenesis inhibitor delta-like 4[70]; (2) Human BM-MSC-EVs or rat BM-MSC-derived exosomes enhance angiogenesis in stroke mice[71] or in rats with renal ischemia-reperfusion (IR) injury[72]; (3) Exosomes from human endometrial MSCs can increase the expression of angiogenesis markers, including Tie2, vascular endothelial growth factor (VEGF), angiopoietin 1 (Ang1), and Ang2, and increase the proliferation, migration, and angiogenesis of human umbilical vein endothelial cells (HUVECs)[73]; and (4) In a rat burn model, treatment with hUCMSC-derived exosomes promoted Ang-2 protein expression in wounds and HUVECs through exosome-mediated Ang-2 transfer. The overexpression of Ang-2 in hUCMSC-derived exosomes further promoted the migration and tube formation of HUVECs, while knockout of Ang-2 in hUCMSC-derived exosomes eliminated these therapeutic and proangiogenic effects[74].

In addition, there have been reports confirming the proangiogenic effect of MSC-derived exosomes in vivo: (1) Human umbilical cord blood (UCB)-MSC-derived exosomes pretreated with thrombin could accelerate skin wound healing in rats with full-thickness wounds. Exosomes from human UCB-MSCs increased angiogenesis factors, such as VEGF, HGF, and Ang1, and decreased TNFα and IL-6[75]; (2) hUC-MSC-derived exosomes enhanced angiogenesis in rats through the Wnt4/β-catenin pathway. When the expression of Wnt4 was knocked out with shRNA, the proangiogenic effect of hUC-MSC-derived exosomes was eliminated[76]; (3) Human iMSC-derived exosomes increased the formation and maturation of new blood vessels at the wound site, although the mechanism is still unclear[77]; and (4) hUCB-MSC-derived exosomes alleviated hepatic ischemia/reperfusion injury by transporting miR-1246 by regulating the glycogen synthase kinase 3β (GSK3β)-mediated Wnt/β-catenin pathway[78].

Remodeling phase

MSC-derived exosomes may help to further reduce scar formation. Typically, uncontrolled accumulation of myofibroblasts in the wound leads to scar formation. Recently, it has been reported that hUC-MSC-derived exosomes reduced the formation of scars by inhibiting the accumulation of mouse myofibroblasts[79]. It has also been reported that human ASC-derived exosomes accelerate skin healing by optimizing the characteristics of fibroblasts. In the early stage of wound healing, ASC-derived exosomes increased the production of collagen I and III, and exosomes could inhibit the expression of collagen to reduce the formation of scars in the later stage[62]. It is well known that various proteases, such as MMPs, are necessary for all stages of skin wound healing[80]. In the remodeling phase, macrophages, fibroblasts, endothelial cells, and epidermal cells control the release of MMPs to degrade the majority of type III collagen fibers[81]. However, the levels of MMPs and their inhibitors should be kept in a relatively stable balance; otherwise, hypertrophic scars or chronic wound outcomes may occur. Studies have shown that ASC-derived exosomes promote ECM reconstruction in cutaneous wound repair by regulating the collagen III:collagen I, MMP3:TIMP1, and TGF-β3: TGF-β1 ratios and by regulating fibroblast differentiation to mitigate scar formation[63]. However, whether ASC-derived exosomes prevent excessive ECM degradation in chronic wounds through the same regulatory mechanism remains to be investigated. Another study also showed that exosomes derived from human menstrual blood mesenchymal cells can reduce scar formation by affecting the Col1:Col3 ratio[49].

PERSPECTIVES ON CLINICAL APPLICATIONS OF MSC-EXOSOMES
Advantages and disadvantages

In the past few years, MSC-derived exosomes have been used as a new cell-free treatment for wound healing and regeneration. This new treatment method has unique advantages. First, exosomes with diameters of 40-100 nm are more likely to participate in blood circulation than MSCs. Some MSCs cannot circulate easily through capillaries, but exosomes can. Second, the treatment also benefits from the small sizes of exosomes. Compared with injection of MSCs, injection of exosomes can achieve higher efficacy with a lower dose. In addition, MSC-derived exosomes can be produced and sterilized as a commercial product, while MSCs cannot be produced. Last but not least, the use of exosomes for cell-free therapy can largely avoid the transfer cells whose DNA may have been damaged or mutated. In cell-based therapy, MSC-derived exosomes are considered to have no safety issues, such as the tumorigenicity potential of cell-mediated drug delivery[11,82]. However, this method of treatment also has some shortcomings. The exosomes injected into the body are static and cannot produce more exosomes. In addition, compared with using MSCs, exosome treatment has increased manufacturing costs and difficulty associated with laboratory preparation.

New prospects of MSC-EXOs in treating wounds

Adding carriers and combined stents to treat wounds: It is well known that uncontrolled protease activity can hinder wound healing[63]. In addition, some studies have shown that the delayed healing of chronic wounds is related to prolonged, high levels of protease activity[80,81,83,84]. The proteolytic environment of chronic wounds may also affect the therapeutic efficacy of MSC-derived exosomes because MSC-derived exosome surface proteins are susceptible to proteolysis, which in turn affects the interaction between exosomes and recipient cells[85]. Based on the natural biocompatibility and cell targeting ability of exosomes, many clinical researchers have focused their attention on using exosomes as new drug delivery systems[86]. Some studies have mixed exosomes with hydrogel or chitosan dressings, allowing the hydrogel or chitosan dressing to act as a stent for sustained release. This strategy can stabilize the local concentration of exosomes, thereby enhancing the ability of exosomes to heal chronic wounds. For example, the FHE@exo hydrogel is prepared by mixing FHE hydrogel (F127/OHA-EPL) and AMSC-derived exosomes to treat diabetic full-thickness skin wounds and can increase the wound closure rate and accelerate angiogenesis, re-epithelialization, and collagen deposition at the wound site. This treatment can significantly improve the healing of chronic diabetic wounds[87]. Similarly, Pluronic F-127 (PF-127) hydrogel containing hUCMSC-derived exosomes can also promote the healing of chronic diabetes wounds and complete skin regeneration[88]. In addition, gingival MSC-derived exosomes combined with a chitosan/silk hydrogel sponge are also helpful for healing chronic wounds in diabetic patients[79]. Furthermore, studies have shown that exosomes derived from microRNA-126-overexpressing synovial MSCs mixed with chitosan dressing can also promote the healing of chronic diabetic wounds[89,90].

Combined with other medications: It has been reported that the exosomes from BMSCs pretreated with atorvastatin could promote angiogenesis through the AKT/eNOS pathway, thereby accelerating the repair of diabetic wounds[91]. Melatonin-stimulated MSC-derived exosomes regulate the polarization of M1 and M2 macrophages by targeting the PTEN/AKT pathway to improve wound healing in diabetes[92].

Combined treatment with physical factors: It has been reported that the exosomes derived from hUC-MSCs exposed to blue light at 455 nm have a robust ability to promote angiogenesis[93]. Labeling human BMSC-derived exosomes with iron oxide nanoparticles and guiding this treatment with a magnetic field could significantly promote the proliferation and migration of endothelial cells and the formation of angiogenic tubules in vivo. Moreover, this treatment could also reduce scar formation and PCNA, CK19, and collagen expression in vivo[94].

Development as an immunosuppressant: Based on the inhibitory effect of exosomes on immune regulation, exosomes can be used as immunosuppressive therapeutic agents in clinical practice. However, because exosomes carry a small amount of allogeneic protein, which can stimulate an autoimmune response, their application in immune regulation remains to be verified[95].

Quality control of extracellular vesicle therapy

Cell-free therapy using exosomes has great development prospects, but the quality control and production specifications should be controlled to some extent for clinical use. The existing preparation guide is minimal information for studies of extracellular vesicles 2018 (MISEV2018), which was proposed by the International Society of Extracellular Vesicles in 2018 and is a series of minimal information on extracellular vesicle research (MISEV)[96-98]. The Korean Ministry of Food and Drug Safety also issued guidelines for EV therapeutic products, entitled "Guidelines for the Quality, Nonclinical and Clinical Evaluation of Extracellular Vesicle Therapeutic Products"[99]. There are certain specifications on the identity, quantity, size, and purity of EVs. Unfortunately, there is currently no evaluation standard for exosomal therapeutic products.

CONCLUSION

In the field of traditional regenerative medicine, although high-dose single factors or cells are used to treat diseases, which can take effect to a certain extent, their regulation mechanism is single and they carry a certain potential risk, such as the formation of ectopic tissue and cell rejection. The clinical application of stem cells in wound repair is greatly limited. Compared with direct reaction of stem cells, exosomes can be extracted on a large scale, with high activity, safe use, more suitable for the internal environment, and no ethical controversy involved, thus having good application potential in wound repair. With the recent explosion in the number of studies on MSC-derived exosomes, various studies have shown anti-inflammatory, anti-aging, and wound healing effects of MSC-exosomes in vivo and in vitro models. MSC-exosome factors are now widely accepted as a new generation of drugs for cell-free therapy. Undeniably, this treatment can obtain good therapeutic effects and has many advantages with practical significance. However, transforming this treatment from a clinical experiment into a mature medical commodity still faces many challenges. For example, we need to set certain standards for exosomal therapeutic products in terms of the identity, quantity, size, purity, and the content of exosomes derived from MSCs.

Footnotes

Manuscript source: Invited manuscript

Specialty type: Research and experimental medicine

Country/Territory of origin: China

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B, B

Grade C (Good): 0

Grade D (Fair): 0

Grade E (Poor): 0

P-Reviewer: Zhang Q S-Editor: Gong ZM L-Editor: Wang TQ P-Editor: Xing YX

References
1.  Andrzejewska A, Lukomska B, Janowski M. Concise Review: Mesenchymal Stem Cells: From Roots to Boost. Stem Cells. 2019;37:855-864.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 303]  [Article Influence: 60.6]  [Reference Citation Analysis (0)]
2.  Timmers L, Lim SK, Arslan F, Armstrong JS, Hoefer IE, Doevendans PA, Piek JJ, El Oakley RM, Choo A, Lee CN, Pasterkamp G, de Kleijn DP. Reduction of myocardial infarct size by human mesenchymal stem cell conditioned medium. Stem Cell Res. 2007;1:129-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 417]  [Cited by in F6Publishing: 436]  [Article Influence: 27.3]  [Reference Citation Analysis (0)]
3.  Liang X, Ding Y, Zhang Y, Tse HF, Lian Q. Paracrine mechanisms of mesenchymal stem cell-based therapy: current status and perspectives. Cell Transplant. 2014;23:1045-1059.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 513]  [Cited by in F6Publishing: 608]  [Article Influence: 67.6]  [Reference Citation Analysis (0)]
4.  Phinney DG, Pittenger MF. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells. 2017;35:851-858.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 831]  [Cited by in F6Publishing: 1033]  [Article Influence: 147.6]  [Reference Citation Analysis (0)]
5.  Lai RC, Arslan F, Lee MM, Sze NS, Choo A, Chen TS, Salto-Tellez M, Timmers L, Lee CN, El Oakley RM, Pasterkamp G, de Kleijn DP, Lim SK. Exosome secreted by MSC reduces myocardial ischemia/reperfusion injury. Stem Cell Res. 2010;4:214-222.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1435]  [Cited by in F6Publishing: 1559]  [Article Influence: 111.4]  [Reference Citation Analysis (0)]
6.  Keller S, Sanderson MP, Stoeck A, Altevogt P. Exosomes: from biogenesis and secretion to biological function. Immunol Lett. 2006;107:102-108.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 592]  [Cited by in F6Publishing: 643]  [Article Influence: 35.7]  [Reference Citation Analysis (0)]
7.  Zipkin M. Exosome redux. Nat Biotechnol. 2019;37:1395-1400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 52]  [Article Influence: 10.4]  [Reference Citation Analysis (0)]
8.  Cross R  Meet the exosome, the rising star in delivery. Chemical & Engineering News. 30 July 2018. [cited 13 December 2019]. Available from: https://cen.acs.org/business/start‐ups/Meet‐exosome‐rising‐star‐drug/96/i31.  [PubMed]  [DOI]  [Cited in This Article: ]
9.  Plieth J, Armstrong M.   Exosomes start to deliver deals. Vantage. 28 Jan 2019. [cited 13 December 2019]. Available from: https://www.evaluate.com/vantage/articles/news/snippets/exosomes‐start‐deliver‐deals.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Han C, Sun X, Liu L, Jiang H, Shen Y, Xu X, Li J, Zhang G, Huang J, Lin Z, Xiong N, Wang T. Exosomes and Their Therapeutic Potentials of Stem Cells. Stem Cells Int. 2016;2016:7653489.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 86]  [Cited by in F6Publishing: 129]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
11.  Lou G, Chen Z, Zheng M, Liu Y. Mesenchymal stem cell-derived exosomes as a new therapeutic strategy for liver diseases. Exp Mol Med. 2017;49:e346.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 264]  [Cited by in F6Publishing: 357]  [Article Influence: 51.0]  [Reference Citation Analysis (0)]
12.  Nooshabadi VT, Mardpour S, Yousefi-Ahmadipour A, Allahverdi A, Izadpanah M, Daneshimehr F, Ai J, Banafshe HR, Ebrahimi-Barough S. The extracellular vesicles-derived from mesenchymal stromal cells: A new therapeutic option in regenerative medicine. J Cell Biochem. 2018;119:8048-8073.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 74]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
13.  Mendt M, Rezvani K, Shpall E. Mesenchymal stem cell-derived exosomes for clinical use. Bone Marrow Transplant. 2019;54:789-792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 193]  [Cited by in F6Publishing: 274]  [Article Influence: 68.5]  [Reference Citation Analysis (0)]
14.  Teixeira FG, Carvalho MM, Sousa N, Salgado AJ. Mesenchymal stem cells secretome: a new paradigm for central nervous system regeneration? Cell Mol Life Sci. 2013;70:3871-3882.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 218]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
15.  Konala VB, Mamidi MK, Bhonde R, Das AK, Pochampally R, Pal R. The current landscape of the mesenchymal stromal cell secretome: A new paradigm for cell-free regeneration. Cytotherapy. 2016;18:13-24.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 286]  [Cited by in F6Publishing: 305]  [Article Influence: 33.9]  [Reference Citation Analysis (0)]
16.  Raposo G, Stoorvogel W. Extracellular vesicles: exosomes, microvesicles, and friends. J Cell Biol. 2013;200:373-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4900]  [Cited by in F6Publishing: 5527]  [Article Influence: 502.5]  [Reference Citation Analysis (0)]
17.  Cunnane EM, Weinbaum JS, O'Brien FJ, Vorp DA. Future Perspectives on the Role of Stem Cells and Extracellular Vesicles in Vascular Tissue Regeneration. Front Cardiovasc Med. 2018;5:86.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 28]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
18.  Koniusz S, Andrzejewska A, Muraca M, Srivastava AK, Janowski M, Lukomska B. Extracellular Vesicles in Physiology, Pathology, and Therapy of the Immune and Central Nervous System, with Focus on Extracellular Vesicles Derived from Mesenchymal Stem Cells as Therapeutic Tools. Front Cell Neurosci. 2016;10:109.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 114]  [Cited by in F6Publishing: 122]  [Article Influence: 15.3]  [Reference Citation Analysis (0)]
19.  Corso G, Mäger I, Lee Y, Görgens A, Bultema J, Giebel B, Wood MJA, Nordin JZ, Andaloussi SE. Reproducible and scalable purification of extracellular vesicles using combined bind-elute and size exclusion chromatography. Sci Rep. 2017;7:11561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 118]  [Cited by in F6Publishing: 118]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
20.  Simpson RJ, Jensen SS, Lim JW. Proteomic profiling of exosomes: current perspectives. Proteomics. 2008;8:4083-4099.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 598]  [Cited by in F6Publishing: 641]  [Article Influence: 40.1]  [Reference Citation Analysis (0)]
21.  van Niel G, Porto-Carreiro I, Simoes S, Raposo G. Exosomes: a common pathway for a specialized function. J Biochem. 2006;140:13-21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 592]  [Cited by in F6Publishing: 651]  [Article Influence: 36.2]  [Reference Citation Analysis (0)]
22.  Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A, Ratajczak MZ. Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication. Leukemia. 2006;20:1487-1495.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 960]  [Cited by in F6Publishing: 1005]  [Article Influence: 55.8]  [Reference Citation Analysis (0)]
23.  Blazquez R, Sanchez-Margallo FM, de la Rosa O, Dalemans W, Alvarez V, Tarazona R, Casado JG. Immunomodulatory Potential of Human Adipose Mesenchymal Stem Cells Derived Exosomes on in vitro Stimulated T Cells. Front Immunol. 2014;5:556.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 218]  [Cited by in F6Publishing: 257]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
24.  Sun BK, Siprashvili Z, Khavari PA. Advances in skin grafting and treatment of cutaneous wounds. Science. 2014;346:941-945.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 388]  [Cited by in F6Publishing: 463]  [Article Influence: 46.3]  [Reference Citation Analysis (1)]
25.  Reish RG, Eriksson E. Scars: a review of emerging and currently available therapies. Plast Reconstr Surg. 2008;122:1068-1078.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 69]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
26.  Rodrigues M, Kosaric N, Bonham CA, Gurtner GC. Wound Healing: A Cellular Perspective. Physiol Rev. 2019;99:665-706.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1040]  [Cited by in F6Publishing: 1064]  [Article Influence: 212.8]  [Reference Citation Analysis (0)]
27.  Golebiewska EM, Poole AW. Platelet secretion: From haemostasis to wound healing and beyond. Blood Rev. 2015;29:153-162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 408]  [Cited by in F6Publishing: 477]  [Article Influence: 47.7]  [Reference Citation Analysis (0)]
28.  Hardwicke J, Schmaljohann D, Boyce D, Thomas D. Epidermal growth factor therapy and wound healing--past, present and future perspectives. Surgeon. 2008;6:172-177.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 154]  [Cited by in F6Publishing: 162]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
29.  Gurtner GC, Werner S, Barrandon Y, Longaker MT. Wound repair and regeneration. Nature. 2008;453:314-321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3609]  [Cited by in F6Publishing: 3814]  [Article Influence: 238.4]  [Reference Citation Analysis (0)]
30.  Eming SA, Martin P, Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci Transl Med. 2014;6:265sr6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1467]  [Cited by in F6Publishing: 1723]  [Article Influence: 191.4]  [Reference Citation Analysis (0)]
31.  Havran WL, Jameson JM. Epidermal T cells and wound healing. J Immunol. 2010;184:5423-5428.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 132]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
32.  Krzyszczyk P, Schloss R, Palmer A, Berthiaume F. The Role of Macrophages in Acute and Chronic Wound Healing and Interventions to Promote Pro-wound Healing Phenotypes. Front Physiol. 2018;9:419.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 487]  [Cited by in F6Publishing: 671]  [Article Influence: 111.8]  [Reference Citation Analysis (0)]
33.  Aukhil I. Biology of wound healing. Periodontol 2000. 2000;22:44-50.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 101]  [Cited by in F6Publishing: 103]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
34.  Efron PA, Moldawer LL. Cytokines and wound healing: the role of cytokine and anticytokine therapy in the repair response. J Burn Care Rehabil. 2004;25:149-160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 100]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
35.  Barrientos S, Stojadinovic O, Golinko MS, Brem H, Tomic-Canic M. Growth factors and cytokines in wound healing. Wound Repair Regen. 2008;16:585-601.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2237]  [Cited by in F6Publishing: 2302]  [Article Influence: 143.9]  [Reference Citation Analysis (0)]
36.  Reinke JM, Sorg H. Wound repair and regeneration. Eur Surg Res. 2012;49:35-43.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 937]  [Cited by in F6Publishing: 1041]  [Article Influence: 86.8]  [Reference Citation Analysis (0)]
37.  Xue M, Jackson CJ. Extracellular Matrix Reorganization During Wound Healing and Its Impact on Abnormal Scarring. Adv Wound Care (New Rochelle). 2015;4:119-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 650]  [Cited by in F6Publishing: 762]  [Article Influence: 84.7]  [Reference Citation Analysis (0)]
38.  Stojadinovic A, Carlson JW, Schultz GS, Davis TA, Elster EA. Topical advances in wound care. Gynecol Oncol. 2008;111:S70-S80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 78]  [Cited by in F6Publishing: 78]  [Article Influence: 4.9]  [Reference Citation Analysis (0)]
39.  Soehnlein O, Steffens S, Hidalgo A, Weber C. Neutrophils as protagonists and targets in chronic inflammation. Nat Rev Immunol. 2017;17:248-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 292]  [Cited by in F6Publishing: 354]  [Article Influence: 50.6]  [Reference Citation Analysis (0)]
40.  Kolaczkowska E, Kubes P. Neutrophil recruitment and function in health and inflammation. Nat Rev Immunol. 2013;13:159-175.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2954]  [Cited by in F6Publishing: 3353]  [Article Influence: 304.8]  [Reference Citation Analysis (0)]
41.  Wynn TA, Vannella KM. Macrophages in Tissue Repair, Regeneration, and Fibrosis. Immunity. 2016;44:450-462.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1758]  [Cited by in F6Publishing: 2310]  [Article Influence: 288.8]  [Reference Citation Analysis (0)]
42.  Silachev DN, Goryunov KV, Shpilyuk MA, Beznoschenko OS, Morozova NY, Kraevaya EE, Popkov VA, Pevzner IB, Zorova LD, Evtushenko EA, Starodubtseva NL, Kononikhin AS, Bugrova AE, Evtushenko EG, Plotnikov EY, Zorov DB, Sukhikh GT. Effect of MSCs and MSC-Derived Extracellular Vesicles on Human Blood Coagulation. Cells. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 65]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
43.  Landén NX, Li D, Ståhle M. Transition from inflammation to proliferation: a critical step during wound healing. Cell Mol Life Sci. 2016;73:3861-3885.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 613]  [Cited by in F6Publishing: 775]  [Article Influence: 96.9]  [Reference Citation Analysis (0)]
44.  Ferreira ADF, Gomes DA. Stem Cell Extracellular Vesicles in Skin Repair. Bioengineering (Basel). 2018;6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 58]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
45.  Eming SA, Krieg T, Davidson JM. Inflammation in wound repair: molecular and cellular mechanisms. J Invest Dermatol. 2007;127:514-525.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1356]  [Cited by in F6Publishing: 1328]  [Article Influence: 78.1]  [Reference Citation Analysis (0)]
46.  He X, Dong Z, Cao Y, Wang H, Liu S, Liao L, Jin Y, Yuan L, Li B. MSC-Derived Exosome Promotes M2 Polarization and Enhances Cutaneous Wound Healing. Stem Cells Int. 2019;2019:7132708.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 99]  [Cited by in F6Publishing: 201]  [Article Influence: 40.2]  [Reference Citation Analysis (0)]
47.  Ti D, Hao H, Tong C, Liu J, Dong L, Zheng J, Zhao Y, Liu H, Fu X, Han W. LPS-preconditioned mesenchymal stromal cells modify macrophage polarization for resolution of chronic inflammation via exosome-shuttled let-7b. J Transl Med. 2015;13:308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 328]  [Cited by in F6Publishing: 427]  [Article Influence: 47.4]  [Reference Citation Analysis (0)]
48.  Li X, Liu L, Yang J, Yu Y, Chai J, Wang L, Ma L, Yin H. Exosome Derived From Human Umbilical Cord Mesenchymal Stem Cell Mediates MiR-181c Attenuating Burn-induced Excessive Inflammation. EBioMedicine. 2016;8:72-82.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 222]  [Cited by in F6Publishing: 289]  [Article Influence: 36.1]  [Reference Citation Analysis (0)]
49.  Dalirfardouei R, Jamialahmadi K, Jafarian AH, Mahdipour E. Promising effects of exosomes isolated from menstrual blood-derived mesenchymal stem cell on wound-healing process in diabetic mouse model. J Tissue Eng Regen Med. 2019;13:555-568.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 73]  [Cited by in F6Publishing: 117]  [Article Influence: 23.4]  [Reference Citation Analysis (0)]
50.  Heo JS, Choi Y, Kim HO. Adipose-Derived Mesenchymal Stem Cells Promote M2 Macrophage Phenotype through Exosomes. Stem Cells Int. 2019;2019:7921760.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 78]  [Article Influence: 15.6]  [Reference Citation Analysis (1)]
51.  Zhao H, Shang Q, Pan Z, Bai Y, Li Z, Zhang H, Zhang Q, Guo C, Zhang L, Wang Q. Exosomes From Adipose-Derived Stem Cells Attenuate Adipose Inflammation and Obesity Through Polarizing M2 Macrophages and Beiging in White Adipose Tissue. Diabetes. 2018;67:235-247.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 326]  [Cited by in F6Publishing: 387]  [Article Influence: 64.5]  [Reference Citation Analysis (0)]
52.  Chen W, Huang Y, Han J, Yu L, Li Y, Lu Z, Li H, Liu Z, Shi C, Duan F, Xiao Y. Immunomodulatory effects of mesenchymal stromal cells-derived exosome. Immunol Res. 2016;64:831-840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 184]  [Cited by in F6Publishing: 220]  [Article Influence: 31.4]  [Reference Citation Analysis (0)]
53.  Du YM, Zhuansun YX, Chen R, Lin L, Lin Y, Li JG. Mesenchymal stem cell exosomes promote immunosuppression of regulatory T cells in asthma. Exp Cell Res. 2018;363:114-120.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 124]  [Cited by in F6Publishing: 142]  [Article Influence: 20.3]  [Reference Citation Analysis (0)]
54.  Zhang Q, Fu L, Liang Y, Guo Z, Wang L, Ma C, Wang H. Exosomes originating from MSCs stimulated with TGF-β and IFN-γ promote Treg differentiation. J Cell Physiol. 2018;233:6832-6840.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 110]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
55.  Zhang B, Yin Y, Lai RC, Tan SS, Choo AB, Lim SK. Mesenchymal stem cells secrete immunologically active exosomes. Stem Cells Dev. 2014;23:1233-1244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 431]  [Cited by in F6Publishing: 473]  [Article Influence: 47.3]  [Reference Citation Analysis (0)]
56.  Nojehdehi S, Soudi S, Hesampour A, Rasouli S, Soleimani M, Hashemi SM. Immunomodulatory effects of mesenchymal stem cell-derived exosomes on experimental type-1 autoimmune diabetes. J Cell Biochem. 2018;119:9433-9443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 119]  [Cited by in F6Publishing: 154]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
57.  Riazifar M, Mohammadi MR, Pone EJ, Yeri A, Lässer C, Segaliny AI, McIntyre LL, Shelke GV, Hutchins E, Hamamoto A, Calle EN, Crescitelli R, Liao W, Pham V, Yin Y, Jayaraman J, Lakey JRT, Walsh CM, Van Keuren-Jensen K, Lotvall J, Zhao W. Stem Cell-Derived Exosomes as Nanotherapeutics for Autoimmune and Neurodegenerative Disorders. ACS Nano. 2019;13:6670-6688.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 252]  [Cited by in F6Publishing: 305]  [Article Influence: 61.0]  [Reference Citation Analysis (0)]
58.  Tamura R, Uemoto S, Tabata Y. Immunosuppressive effect of mesenchymal stem cell-derived exosomes on a concanavalin A-induced liver injury model. Inflamm Regen. 2016;36:26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 97]  [Article Influence: 12.1]  [Reference Citation Analysis (0)]
59.  Jiang T, Wang Z, Sun J. Human bone marrow mesenchymal stem cell-derived exosomes stimulate cutaneous wound healing mediates through TGF-β/Smad signaling pathway. Stem Cell Res Ther. 2020;11:198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 74]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
60.  Hu HH, Chen DQ, Wang YN, Feng YL, Cao G, Vaziri ND, Zhao YY. New insights into TGF-β/Smad signaling in tissue fibrosis. Chem Biol Interact. 2018;292:76-83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 363]  [Cited by in F6Publishing: 594]  [Article Influence: 99.0]  [Reference Citation Analysis (0)]
61.  Mokoena D, Dhilip Kumar SS, Houreld NN, Abrahamse H. Role of photobiomodulation on the activation of the Smad pathway via TGF-β in wound healing. J Photochem Photobiol B. 2018;189:138-144.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 39]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
62.  Hu L, Wang J, Zhou X, Xiong Z, Zhao J, Yu R, Huang F, Zhang H, Chen L. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci Rep. 2016;6:32993.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 296]  [Cited by in F6Publishing: 375]  [Article Influence: 46.9]  [Reference Citation Analysis (0)]
63.  Wang L, Hu L, Zhou X, Xiong Z, Zhang C, Shehada HMA, Hu B, Song J, Chen L. Exosomes secreted by human adipose mesenchymal stem cells promote scarless cutaneous repair by regulating extracellular matrix remodelling. Sci Rep. 2017;7:13321.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 134]  [Cited by in F6Publishing: 184]  [Article Influence: 26.3]  [Reference Citation Analysis (0)]
64.  Wang X, Jiao Y, Pan Y, Zhang L, Gong H, Qi Y, Wang M, Shao M, Wang X, Jiang D. Fetal Dermal Mesenchymal Stem Cell-Derived Exosomes Accelerate Cutaneous Wound Healing by Activating Notch Signaling. Stem Cells Int. 2019;2019:2402916.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 64]  [Article Influence: 12.8]  [Reference Citation Analysis (0)]
65.  Zhang B, Wang M, Gong A, Zhang X, Wu X, Zhu Y, Shi H, Wu L, Zhu W, Qian H, Xu W. HucMSC-Exosome Mediated-Wnt4 Signaling Is Required for Cutaneous Wound Healing. Stem Cells. 2015;33:2158-2168.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 406]  [Cited by in F6Publishing: 514]  [Article Influence: 57.1]  [Reference Citation Analysis (0)]
66.  Shabbir A, Cox A, Rodriguez-Menocal L, Salgado M, Van Badiavas E. Mesenchymal Stem Cell Exosomes Induce Proliferation and Migration of Normal and Chronic Wound Fibroblasts, and Enhance Angiogenesis In Vitro. Stem Cells Dev. 2015;24:1635-1647.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 371]  [Cited by in F6Publishing: 430]  [Article Influence: 47.8]  [Reference Citation Analysis (0)]
67.  Kim S, Lee SK, Kim H, Kim TM. Exosomes Secreted from Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Accelerate Skin Cell Proliferation. Int J Mol Sci. 2018;19.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 120]  [Article Influence: 20.0]  [Reference Citation Analysis (0)]
68.  Zhang Y, Han F, Gu L, Ji P, Yang X, Liu M, Tao K, Hu D. Adipose mesenchymal stem cell exosomes promote wound healing through accelerated keratinocyte migration and proliferation by activating the AKT/HIF-1α axis. J Mol Histol. 2020;51:375-383.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 28]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
69.  Zhao G, Liu F, Liu Z, Zuo K, Wang B, Zhang Y, Han X, Lian A, Wang Y, Liu M, Zou F, Li P, Liu X, Jin M, Liu JY. MSC-derived exosomes attenuate cell death through suppressing AIF nucleus translocation and enhance cutaneous wound healing. Stem Cell Res Ther. 2020;11:174.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 52]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
70.  Liang X, Zhang L, Wang S, Han Q, Zhao RC. Exosomes secreted by mesenchymal stem cells promote endothelial cell angiogenesis by transferring miR-125a. J Cell Sci. 2016;129:2182-2189.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 306]  [Cited by in F6Publishing: 372]  [Article Influence: 53.1]  [Reference Citation Analysis (0)]
71.  Fan B, Li C, Szalad A, Wang L, Pan W, Zhang R, Chopp M, Zhang ZG, Liu XS. Mesenchymal stromal cell-derived exosomes ameliorate peripheral neuropathy in a mouse model of diabetes. Diabetologia. 2020;63:431-443.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 80]  [Cited by in F6Publishing: 104]  [Article Influence: 26.0]  [Reference Citation Analysis (0)]
72.  Alzahrani FA. Melatonin improves therapeutic potential of mesenchymal stem cells-derived exosomes against renal ischemia-reperfusion injury in rats. Am J Transl Res. 2019;11:2887-2907.  [PubMed]  [DOI]  [Cited in This Article: ]
73.  Ha DH, Kim HK, Lee J, Kwon HH, Park GH, Yang SH, Jung JY, Choi H, Lee JH, Sung S, Yi YW, Cho BS. Mesenchymal Stem/Stromal Cell-Derived Exosomes for Immunomodulatory Therapeutics and Skin Regeneration. Cells. 2020;9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 264]  [Cited by in F6Publishing: 223]  [Article Influence: 55.8]  [Reference Citation Analysis (0)]
74.  Liu J, Yan Z, Yang F, Huang Y, Yu Y, Zhou L, Sun Z, Cui D, Yan Y. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Accelerate Cutaneous Wound Healing by Enhancing Angiogenesis through Delivering Angiopoietin-2. Stem Cell Rev Rep. 2020;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 49]  [Article Influence: 12.3]  [Reference Citation Analysis (0)]
75.  Sung DK, Chang YS, Sung SI, Ahn SY, Park WS. Thrombin Preconditioning of Extracellular Vesicles Derived from Mesenchymal Stem Cells Accelerates Cutaneous Wound Healing by Boosting Their Biogenesis and Enriching Cargo Content. J Clin Med. 2019;8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 41]  [Article Influence: 8.2]  [Reference Citation Analysis (0)]
76.  Zhang B, Wu X, Zhang X, Sun Y, Yan Y, Shi H, Zhu Y, Wu L, Pan Z, Zhu W, Qian H, Xu W. Human umbilical cord mesenchymal stem cell exosomes enhance angiogenesis through the Wnt4/β-catenin pathway. Stem Cells Transl Med. 2015;4:513-522.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 315]  [Article Influence: 35.0]  [Reference Citation Analysis (0)]
77.  Fang S, Xu C, Zhang Y, Xue C, Yang C, Bi H, Qian X, Wu M, Ji K, Zhao Y, Wang Y, Liu H, Xing X. 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. 2016;5:1425-1439.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 281]  [Cited by in F6Publishing: 355]  [Article Influence: 44.4]  [Reference Citation Analysis (0)]
78.  Xie K, Liu L, Chen J, Liu F. Exosomes derived from human umbilical cord blood mesenchymal stem cells improve hepatic ischemia reperfusion injury via delivering miR-1246. Cell Cycle. 2019;18:3491-3501.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 34]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
79.  Shi Q, Qian Z, Liu D, Sun J, Wang X, Liu H, Xu J, Guo X. GMSC-Derived Exosomes Combined with a Chitosan/Silk Hydrogel Sponge Accelerates Wound Healing in a Diabetic Rat Skin Defect Model. Front Physiol. 2017;8:904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 151]  [Cited by in F6Publishing: 223]  [Article Influence: 31.9]  [Reference Citation Analysis (0)]
80.  McCarty SM, Percival SL. Proteases and Delayed Wound Healing. Adv Wound Care (New Rochelle). 2013;2:438-447.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 255]  [Cited by in F6Publishing: 272]  [Article Influence: 24.7]  [Reference Citation Analysis (0)]
81.  Sabino F, auf dem Keller U. Matrix metalloproteinases in impaired wound healing. Metalloproteinases Med. 2015;2:1-8.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 13]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
82.  Cho BS, Kim JO, Ha DH, Yi YW. Exosomes derived from human adipose tissue-derived mesenchymal stem cells alleviate atopic dermatitis. Stem Cell Res Ther. 2018;9:187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 166]  [Article Influence: 27.7]  [Reference Citation Analysis (0)]
83.  Westby MJ, Dumville JC, Stubbs N, Norman G, Wong JK, Cullum N, Riley RD. Protease activity as a prognostic factor for wound healing in venous leg ulcers. Cochrane Database Syst Rev. 2018;9:CD012841.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 20]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]
84.  International Consensus  The role of proteases in wound healing diagnostics. An expert working group review. Wounds International: London, UK, 2011.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Longatti A, Schindler C, Collinson A, Jenkinson L, Matthews C, Fitzpatrick L, Blundy M, Minter R, Vaughan T, Shaw M, Tigue N. High affinity single-chain variable fragments are specific and versatile targeting motifs for extracellular vesicles. Nanoscale. 2018;10:14230-14244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 52]  [Article Influence: 8.7]  [Reference Citation Analysis (0)]
86.  Taverna S, Pucci M, Alessandro R. Extracellular vesicles: small bricks for tissue repair/regeneration. Ann Transl Med. 2017;5:83.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 41]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
87.  Wang C, Wang M, Xu T, Zhang X, Lin C, Gao W, Xu H, Lei B, Mao C. Engineering Bioactive Self-Healing Antibacterial Exosomes Hydrogel for Promoting Chronic Diabetic Wound Healing and Complete Skin Regeneration. Theranostics. 2019;9:65-76.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 441]  [Cited by in F6Publishing: 424]  [Article Influence: 84.8]  [Reference Citation Analysis (0)]
88.  Yang J, Chen Z, Pan D, Li H, Shen J. Umbilical Cord-Derived Mesenchymal Stem Cell-Derived Exosomes Combined Pluronic F127 Hydrogel Promote Chronic Diabetic Wound Healing and Complete Skin Regeneration. Int J Nanomedicine. 2020;15:5911-5926.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 203]  [Article Influence: 50.8]  [Reference Citation Analysis (0)]
89.  Tao SC, Guo SC, Li M, Ke QF, Guo YP, Zhang CQ. Chitosan Wound Dressings Incorporating Exosomes Derived from MicroRNA-126-Overexpressing Synovium Mesenchymal Stem Cells Provide Sustained Release of Exosomes and Heal Full-Thickness Skin Defects in a Diabetic Rat Model. Stem Cells Transl Med. 2017;6:736-747.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 183]  [Cited by in F6Publishing: 235]  [Article Influence: 29.4]  [Reference Citation Analysis (0)]
90.  Li M, Ke QF, Tao SC, Guo SC, Rui BY, Guo YP. Fabrication of hydroxyapatite/chitosan composite hydrogels loaded with exosomes derived from miR-126-3p overexpressed synovial mesenchymal stem cells for diabetic chronic wound healing. J Mater Chem B. 2016;4:6830-6841.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 72]  [Article Influence: 9.0]  [Reference Citation Analysis (0)]
91.  Yu M, Liu W, Li J, Lu J, Lu H, Jia W, Liu F. Exosomes derived from atorvastatin-pretreated MSC accelerate diabetic wound repair by enhancing angiogenesis via AKT/eNOS pathway. Stem Cell Res Ther. 2020;11:350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 63]  [Cited by in F6Publishing: 154]  [Article Influence: 38.5]  [Reference Citation Analysis (0)]
92.  Liu W, Yu M, Xie D, Wang L, Ye C, Zhu Q, Liu F, Yang L. Melatonin-stimulated MSC-derived exosomes improve diabetic wound healing through regulating macrophage M1 and M2 polarization by targeting the PTEN/AKT pathway. Stem Cell Res Ther. 2020;11:259.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 200]  [Article Influence: 50.0]  [Reference Citation Analysis (0)]
93.  Yang K, Li D, Wang M, Xu Z, Chen X, Liu Q, Sun W, Li J, Gong Y, Liu D, Shao C, Li X. Exposure to blue light stimulates the proangiogenic capability of exosomes derived from human umbilical cord mesenchymal stem cells. Stem Cell Res Ther. 2019;10:358.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 48]  [Article Influence: 9.6]  [Reference Citation Analysis (0)]
94.  Li X, Wang Y, Shi L, Li B, Li J, Wei Z, Lv H, Wu L, Zhang H, Yang B, Xu X, Jiang J. Magnetic targeting enhances the cutaneous wound healing effects of human mesenchymal stem cell-derived iron oxide exosomes. J Nanobiotechnology. 2020;18:113.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 70]  [Article Influence: 17.5]  [Reference Citation Analysis (0)]
95.  Théry C, Duban L, Segura E, Véron P, Lantz O, Amigorena S. Indirect activation of naïve CD4+ T cells by dendritic cell-derived exosomes. Nat Immunol. 2002;3:1156-1162.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 666]  [Cited by in F6Publishing: 691]  [Article Influence: 31.4]  [Reference Citation Analysis (0)]
96.  Lötvall J, Hill AF, Hochberg F, Buzás EI, Di Vizio D, Gardiner C, Gho YS, Kurochkin IV, Mathivanan S, Quesenberry P, Sahoo S, Tahara H, Wauben MH, Witwer KW, Théry C. Minimal experimental requirements for definition of extracellular vesicles and their functions: a position statement from the International Society for Extracellular Vesicles. J Extracell Vesicles. 2014;3:26913.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1880]  [Cited by in F6Publishing: 1846]  [Article Influence: 184.6]  [Reference Citation Analysis (0)]
97.  Witwer KW, Soekmadji C, Hill AF, Wauben MH, Buzás EI, Di Vizio D, Falcon-Perez JM, Gardiner C, Hochberg F, Kurochkin IV, Lötvall J, Mathivanan S, Nieuwland R, Sahoo S, Tahara H, Torrecilhas AC, Weaver AM, Yin H, Zheng L, Gho YS, Quesenberry P, Théry C. Updating the MISEV minimal requirements for extracellular vesicle studies: building bridges to reproducibility. J Extracell Vesicles. 2017;6:1396823.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 165]  [Article Influence: 23.6]  [Reference Citation Analysis (0)]
98.  Théry C, Witwer KW, Aikawa E, Alcaraz MJ, Anderson JD, Andriantsitohaina R, Antoniou A, Arab T, Archer F, Atkin-Smith GK, Ayre DC, Bach JM, Bachurski D, Baharvand H, Balaj L, Baldacchino S, Bauer NN, Baxter AA, Bebawy M, Beckham C, Bedina Zavec A, Benmoussa A, Berardi AC, Bergese P, Bielska E, Blenkiron C, Bobis-Wozowicz S, Boilard E, Boireau W, Bongiovanni A, Borràs FE, Bosch S, Boulanger CM, Breakefield X, Breglio AM, Brennan MÁ, Brigstock DR, Brisson A, Broekman ML, Bromberg JF, Bryl-Górecka P, Buch S, Buck AH, Burger D, Busatto S, Buschmann D, Bussolati B, Buzás EI, Byrd JB, Camussi G, Carter DR, Caruso S, Chamley LW, Chang YT, Chen C, Chen S, Cheng L, Chin AR, Clayton A, Clerici SP, Cocks A, Cocucci E, Coffey RJ, Cordeiro-da-Silva A, Couch Y, Coumans FA, Coyle B, Crescitelli R, Criado MF, D'Souza-Schorey C, Das S, Datta Chaudhuri A, de Candia P, De Santana EF, De Wever O, Del Portillo HA, Demaret T, Deville S, Devitt A, Dhondt B, Di Vizio D, Dieterich LC, Dolo V, Dominguez Rubio AP, Dominici M, Dourado MR, Driedonks TA, Duarte FV, Duncan HM, Eichenberger RM, Ekström K, El Andaloussi S, Elie-Caille C, Erdbrügger U, Falcón-Pérez JM, Fatima F, Fish JE, Flores-Bellver M, Försönits A, Frelet-Barrand A, Fricke F, Fuhrmann G, Gabrielsson S, Gámez-Valero A, Gardiner C, Gärtner K, Gaudin R, Gho YS, Giebel B, Gilbert C, Gimona M, Giusti I, Goberdhan DC, Görgens A, Gorski SM, Greening DW, Gross JC, Gualerzi A, Gupta GN, Gustafson D, Handberg A, Haraszti RA, Harrison P, Hegyesi H, Hendrix A, Hill AF, Hochberg FH, Hoffmann KF, Holder B, Holthofer H, Hosseinkhani B, Hu G, Huang Y, Huber V, Hunt S, Ibrahim AG, Ikezu T, Inal JM, Isin M, Ivanova A, Jackson HK, Jacobsen S, Jay SM, Jayachandran M, Jenster G, Jiang L, Johnson SM, Jones JC, Jong A, Jovanovic-Talisman T, Jung S, Kalluri R, Kano SI, Kaur S, Kawamura Y, Keller ET, Khamari D, Khomyakova E, Khvorova A, Kierulf P, Kim KP, Kislinger T, Klingeborn M, Klinke DJ 2nd, Kornek M, Kosanović MM, Kovács ÁF, Krämer-Albers EM, Krasemann S, Krause M, Kurochkin IV, Kusuma GD, Kuypers S, Laitinen S, Langevin SM, Languino LR, Lannigan J, Lässer C, Laurent LC, Lavieu G, Lázaro-Ibáñez E, Le Lay S, Lee MS, Lee YXF, Lemos DS, Lenassi M, Leszczynska A, Li IT, Liao K, Libregts SF, Ligeti E, Lim R, Lim SK, Linē A, Linnemannstöns K, Llorente A, Lombard CA, Lorenowicz MJ, Lörincz ÁM, Lötvall J, Lovett J, Lowry MC, Loyer X, Lu Q, Lukomska B, Lunavat TR, Maas SL, Malhi H, Marcilla A, Mariani J, Mariscal J, Martens-Uzunova ES, Martin-Jaular L, Martinez MC, Martins VR, Mathieu M, Mathivanan S, Maugeri M, McGinnis LK, McVey MJ, Meckes DG Jr, Meehan KL, Mertens I, Minciacchi VR, Möller A, Møller Jørgensen M, Morales-Kastresana A, Morhayim J, Mullier F, Muraca M, Musante L, Mussack V, Muth DC, Myburgh KH, Najrana T, Nawaz M, Nazarenko I, Nejsum P, Neri C, Neri T, Nieuwland R, Nimrichter L, Nolan JP, Nolte-'t Hoen EN, Noren Hooten N, O'Driscoll L, O'Grady T, O'Loghlen A, Ochiya T, Olivier M, Ortiz A, Ortiz LA, Osteikoetxea X, Østergaard O, Ostrowski M, Park J, Pegtel DM, Peinado H, Perut F, Pfaffl MW, Phinney DG, Pieters BC, Pink RC, Pisetsky DS, Pogge von Strandmann E, Polakovicova I, Poon IK, Powell BH, Prada I, Pulliam L, Quesenberry P, Radeghieri A, Raffai RL, Raimondo S, Rak J, Ramirez MI, Raposo G, Rayyan MS, Regev-Rudzki N, Ricklefs FL, Robbins PD, Roberts DD, Rodrigues SC, Rohde E, Rome S, Rouschop KM, Rughetti A, Russell AE, Saá P, Sahoo S, Salas-Huenuleo E, Sánchez C, Saugstad JA, Saul MJ, Schiffelers RM, Schneider R, Schøyen TH, Scott A, Shahaj E, Sharma S, Shatnyeva O, Shekari F, Shelke GV, Shetty AK, Shiba K, Siljander PR, Silva AM, Skowronek A, Snyder OL 2nd, Soares RP, Sódar BW, Soekmadji C, Sotillo J, Stahl PD, Stoorvogel W, Stott SL, Strasser EF, Swift S, Tahara H, Tewari M, Timms K, Tiwari S, Tixeira R, Tkach M, Toh WS, Tomasini R, Torrecilhas AC, Tosar JP, Toxavidis V, Urbanelli L, Vader P, van Balkom BW, van der Grein SG, Van Deun J, van Herwijnen MJ, Van Keuren-Jensen K, van Niel G, van Royen ME, van Wijnen AJ, Vasconcelos MH, Vechetti IJ Jr, Veit TD, Vella LJ, Velot É, Verweij FJ, Vestad B, Viñas JL, Visnovitz T, Vukman KV, Wahlgren J, Watson DC, Wauben MH, Weaver A, Webber JP, Weber V, Wehman AM, Weiss DJ, Welsh JA, Wendt S, Wheelock AM, Wiener Z, Witte L, Wolfram J, Xagorari A, Xander P, Xu J, Yan X, Yáñez-Mó M, Yin H, Yuana Y, Zappulli V, Zarubova J, Žėkas V, Zhang JY, Zhao Z, Zheng L, Zheutlin AR, Zickler AM, Zimmermann P, Zivkovic AM, Zocco D, Zuba-Surma EK. Minimal information for studies of extracellular vesicles 2018 (MISEV2018): a position statement of the International Society for Extracellular Vesicles and update of the MISEV2014 guidelines. J Extracell Vesicles. 2018;7:1535750.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6453]  [Cited by in F6Publishing: 6146]  [Article Influence: 1024.3]  [Reference Citation Analysis (0)]
99.  Cell and Gene Therapy Products Division Biopharmaceutical and Herbal Medicine Evaluation Department National Institute of Food and Drug Safety Evaluation Ministry of Food and Drug Safety  Guideline on quality non‐clinical and clinical assessment of extracellular vesicles therapy products. 2018. [cited 13 December 2019]. Available from: www.nifds.go.kr/brd/m_15/down.do?brd_id=167&seq=12625&data_tp=A&file_seq=1.  [PubMed]  [DOI]  [Cited in This Article: ]