Back to Journals » International Journal of Nanomedicine » Volume 20

MSC-Derived Extracellular Vesicles: Roles and Molecular Mechanisms for Tissue Repair

Authors Wu T, Liu Y, Wang S, Shi C

Received 27 February 2025

Accepted for publication 27 May 2025

Published 21 June 2025 Volume 2025:20 Pages 7953—7974

DOI https://doi.org/10.2147/IJN.S525394

Checked for plagiarism Yes

Review by Single anonymous peer review

Peer reviewer comments 2

Editor who approved publication: Professor Eng San Thian



Tingting Wu,1,2,* Yajing Liu,1,2,* Shuman Wang,1,2 Chen Shi1– 3

1Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China; 2Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, Hubei, People’s Republic of China; 3Hubei Key Laboratory of Natural Active Polysaccharides, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China

*These authors contributed equally to this work

Correspondence: Chen Shi, Email [email protected]

Abstract: Mesenchymal stem cells (MSCs) are pluripotent stem cells that have great potential in the field of regenerative medicine. Extracellular vesicles that derived from MSCs inherit the bioactive molecules, including lipids, proteins, nucleic acids, from the donate cells. With similar biological functions as MSCs, MSC-EVs show lower toxicity, better biocompatibility, stability, and less immune rejection. Therefore, MSC-EVs are emerging as a cell-free alternative therapy for MSC-based cell therapy. Here, we reviewed the characteristics of different subtype of MSC-EVs, the functional molecules in MSC-EVs for tissue repair, the potential applications of MSC-EVs for different tissue injuries in recent three years, and the clinical progress as well as the existing challenges in MSC-EV based therapy. This review would provide some insights for the future researches and clinical translation of MSC-EVs in regenerative medicine.

Keywords: mesenchymal stem cells, extracellular vesicles, cell-free therapy, tissue repair, regenerative medicine

Introduction

Tissue repair and regeneration remain as a great challenge in regenerative medicine. Tissue damages occur in cells, tissues, or organs due to various factors such as degenerative diseases, organ failure, or mechanical injury.1 Degenerative diseases such as osteoarthritis and Alzheimer’s disease (AD), are one of the major driving factors for tissue damage. As of 2020, over 500 million people are affected by osteoarthritis, accounting for approximately 7.6% of the global population.2 In the entire AD continuum, the overall prevalence among people aged >50 is 22%. Especially in the elderly and advanced disease stages, the rate accounts for 54%. Furthermore, multiple organ dysfunction syndrome is often triggered by acute sepsis. Its main manifestations include high vascular permeability, release of inflammatory mediators, microcirculation disorders, and self-reactive endothelial damage. All of these would directly lead to organ damage.3,4 In addition, there are also extensive traumatic injuries caused by fractures or complex surgeries. The incidence of related conditions such as infection, hematoma, and nerve damage reach 8.5%–20%.5 Therefore, there is an urgent need for regenerative therapies in tissue damage.

Generally, the tissue repair processes undergo three stages: acute inflammatory phase, tissue repair phase, and remodeling phase.6–8 After tissue damage, blood vessels rapidly dilate, the permeability of blood vessel walls increase, and plasma proteins and white blood cells rapidly infiltrate into the interstitium of injured tissue. Then various immune cells, such as macrophages, will be activated and migrate to damaged tissue, releasing signaling molecules to recruit more immune cells to participate in the inflammatory response.9 Afterwards, the damaged tissue and immune cells release various inflammatory mediators, such as prostaglandins (PGE), interleukins (ILs), and tumor necrosis factor (TNF),10 which can trigger inflammatory reactions such as local congestion and exudation. When the inflammation subsides, the damaged tissue enters the repair stage. In the early stage of repair, fibroblasts rapidly proliferate in the damaged area and synthesize collagen. At the same time, epithelial cells will migrate and proliferate to cover the wound surface, preventing infection and loss of moisture. Then endothelial cells in blood vessels form new blood vessels to restore blood supply to the damaged cells and provide oxygen, nutrients, and immune cells for wound healing.11 In the middle stage of repair, fibroblasts continue to synthesize collagen, and the deposition and arrangement of these collagens support the repair area, providing a structural basis for subsequent repair and functional recovery, also preventing further damage. In the late stage of tissue repair, fibroblasts and myoblasts synthesize and deposit extracellular matrix (ECM) proteins to form scars.12 Following by the repair phase, the tissue enters the remodeling phase.13 During this period, the inflammatory response gradually weakens, the number of immune cells decreases, and stem cells differentiate into various cell types, promoting the restoration of tissue structure and function. Afterwards, the newly formed tissues gradually interact with the surrounding environment, and form a new physiological balance to maintain the stability and function of the body (Figure 1).

Figure 1 Schematic illustration of the process of tissue repair.

Mesenchymal stem cells (MSCs), also referred as mesenchymal stromal cells or drug signaling cells, are multipotent cells that originate from mesodermal tissues14 and have the potential for multidirectional differentiation15 immune regulation,16 and paracrine secretion.17 According to the statement from the International Society for Cell and Gene Therapy (ISCT)®,18 MSCs can be characterized by the expression of surface antigens CD73, CD90 and CD105, the lack of expression of CD45, CD34, CD14 or CD11b, CD79α or CD19 and HLA-DR, as well as plastic adherence during in vitro growth. MSCs mainly exist in the bone marrow,19 but they can also be isolated from peripheral tissues, including umbilical cord,20 placenta,21 adipose tissue22, amniotic fluid,23 Wharton’s jelly,24 and skin,25 etc. Furthermore, oral tissues are also a major source of MSCs.26 The oral tissue-derived MSCs include dental pulp stem cells, stem cells from human exfoliated deciduous teeth, periodontal ligament stem cells, gingival mesenchymal stem cells, dental follicle stem cells, alveolar bone-derived MSCs, and stem cells from apical papilla.27–30 MSCs derived from different tissues exhibit heterogeneous characteristics and regulate various biological functions, including multi-lineage differentiation, angiogenesis, and anti-inflammatory effects, etc. Moreover, MSCs lack the expression of MHC-II on their surface, making them less immunogenic and advantageous for allogeneic transplantation.31

Extracellular vesicles (EVs) are nano-sized membrane particles secreted by cells. Almost all type of cells can secrete membrane-bound vesicles. According to the size or origin, EVs can be roughly classified into microvesicles, exosomes, and apoptotic bodies.32 Notably, they can mediate the intracellular communication and thereby play important roles in immune responses, anti-inflammatory processes, angiogenesis, and anti-tumor activities.33 When compared with MSC, MSC derived-EVs (MSC-EVs) show better stability, biosafety and storability.34 MSC-EVs are promising candidates in treating various diseases, such as autoimmune diseases and diabetes, etc.

This review summarized the various types of MSC-EVs and their bioactive molecules for tissue repair, as well as the application of MSC-EVs in various disease models in recent three years. This could provide a theoretical basis for clinical translation of MSC-EVs.

Characteristics of MSC-EVs

As a kind of multipotent cells, MSCs are derived from various tissue sources and possess the capacity for self-renewal and can differentiate into various types of cells, encompassing mesodermal lineages such as chondrocytes, osteocytes, adipocytes, myocytes, and fibroblasts; ectodermal lineages like neurons; and endodermal lineages including hepatocytes.35 Derived from natural MSCs, MSC-EVs showed innate biomimetic-properties and high biocompatibility. They also inherited the various biomolecules from the donate MSCs, thereby possessing similar functions as the parental MSCs. They can transfer the bioactive substances, such as proteins, lipids, DNA, and microRNA (miRNA), to the recipient cells, which endow them with the capability to participate in intercellular signal transduction and regulate multiple functions (Figure 2). Notably, as a cell-free therapy, MSC-EVs have lower toxicity, better biocompatibility, stability, and a lower risk of immune rejection compared to cell-based therapies.36,37 Moreover, as a kind of top-bottom approach, MSC-EVs can directly act as nanosized-drug carrier and utilize the self-carrying biomolecules as therapeutic agents while avoid complicated and costive drug loading or engineering processes. Therefore, MSC-EV could be a promising candidate in treating various diseases.

Figure 2 The biogenesis of MSC-EVs and their communicated roles between MSCs and recipient cells.

Abbreviations: MSC, Mesenchymal stem cell; MVB, Multivesicular Body.

According to the size and origin, MSC-EVs can be broadly categorized into three subtypes: MSC-derived exosome (MSC-Exo), MSC-derived microvesicles (MSC-MVs), and MSC-derived apoptotic body.38 MSC-Exo typically ranges in size from 30 to 100 nm and is released through fusion of endosomal multivesicular bodies (MVB) structures with the plasma membrane. In detail, early sorting endosomes (ESEs) are firstly formed through plasma membrane invagination. Then, late sorting endosomes are generated by material exchange following acidification and maturation of ESEs. Subsequently, MVBs form via double plasma membrane invagination, containing numerous intraluminal vesicles (ILVs). Ultimately, ILVs are released as exosomes.39 MSC-MVs are generated by shedding from the plasma membrane and have a diameter ranging from 100 nm to 1000 nm. MSC-derived apoptotic bodies, which usually form through apoptotic blebs during cell death, are the largest in size, ranging from 500 nm to 2000 nm. For the isolation of different MSC-EVs, the commonly used methods include ultracentrifugation, ultrafiltration, size-exclusion chromatography, polymer precipitation, immunoaffinity capture, microfluidic technology, and commercial EV isolation kits.40 According to the actual demand, a suitable separation method could be chosen for the isolation of MSC-EVs.

Functional Molecules in MSC-EVs for Tissue Repair

MSC-EVs have shown great potential in tissue repair. The bioactive molecules including miRNA, long non-coding RNA (lncRNA), circular RNA (circRNA), proteins and lipids inherited from the donor cells are highlighted for the pro-regenerative effects.

MiRNA in MSC-EVs for Tissue Repair

MiRNA is a class of endogenous non-coding RNA molecules with a length of about 19–25 nucleotides, which play an important role in the differentiation, proliferation and apoptosis of cells. Inherited from the donate MSCs, MSC-EVs contain abundant miRNAs, which play important roles in many aspects of tissue repair (Figure 3).

Figure 3 Roles and mechanisms of miRNAs in MSC-EVs to promote tissue repair.

Abbreviations: TGF-β, Transforming Growth Factor-β; APAF1, Apoptotic Protease Activating Factor 1; PI3K, Phosphoinositide 3-Kinase; AKT, Rac-alpha Serine/Threonine-Protein Kinase; MMP, Matrix Metalloproteinase; ERK, Extracellular Signal-Related Kinase; KEAP1, Kelch-like ECH-associated Protein 1; Nrf2, Nuclear Factor Erythroid-Related Factor 2; HO-1, Heme Oxygenase-1; STAT1, Signal Transducer and Activator of Transcription 1; TRAF6, TNF Receptor-Associated Factor 6; IRF5, Interferon Regulatory Factor 5; eNOS, Endothelial Nitric Oxide Synthase; PARP9, Poly [ADP -ribose] polymerase 9; pSTAT1, Phosphorylated Signal Transducer and Activator of Transcription 1; Src, Rous Sarcoma Virus Oncogene Homolog; VE-cadherin, Vascular Endothelial Cadherin; IL-1β, Interleukin-1β; IL-6, Interleukin-6; TNF-α, Tumor Necrosis Factor-α; CXCR4, C-X-C Chemokine Receptor Type 4; COX-2, Cyclooxygenase-2; YAP1, Yes-associated Protein 1; HDAC7, Histone Deacetylase 7; cAMP, Cyclic Adenosine Monophosphate; PKA, Protein Kinase A; CREB, cAMP-response Element-Binding Protein; GPX4, Glutathione Peroxidase 4; MAP3K12, Mitogen-Activated Protein Kinase 12; Caspase-3, Cysteine-aspartic Acid Protease 3.

Typically, miRNA in MSC-EV can regulate cell proliferation and differentiation by regulating gene expression in target cells, thus accelerating the repair process of damaged tissues. It was reported that miR-21-5p and miR-125b-5p in human umbilical cord blood MSCs derived exosomes were correlated with the inhibition of transforming growth factor-β (TGF-β) signaling pathway for myofibroblast differentiation, thereby reducing scar formation while accelerating wound healing.41 MiR-21-5p was also engineered to adipose stem-cell-derived exosomes (ADSC-Exo) to promote the proliferation and migration of keratinocytes via Wnt/β-catenin signaling, promoting diabetic wound healing.42 MiR-93-3p in bone marrow MSC-Exo promoted the proliferation and migration while reduced the apoptosis of HaCaT cells (human keratinocytes) by suppressing apoptotic peptidase activating factor 1 (APAF1) for skin wound healing.43 Similarly, miR-21 in ADSC-Exo could also promote the proliferation and migration of HaCaT cells, which was mediated by the enhanced expression of MMPs via PI3K/AKT signaling pathway.44 MiR-29c and miR-129 in MSC-EVs were found to induce dedifferentiation of myofibroblasts and thereby exerted antifibrotic effect in pulmonary fibrosis.45 MiR-145-5p was highly enriched in placental MSC-EVs, which promoted the proliferation, migration, and antiapoptotic abilities of high glucose-induced senescent fibroblasts by suppressing the expression of cyclin dependent kinase inhibitor 1A and activating ERK/AKT signaling pathway. As a result, miR-145-5p mimics could improve the delayed skin wound healing caused by these abnormal fibroblasts.46 MiR-24 in human umbilical cord MSC-EVs was found with ability to shift macrophage polarization from M1 towards M2 type by targeting kelch like ECH associated protein 1 (KEAP1) and disrupting the downstream Nrf2/HO-1 signaling, providing protective effect on myocardial ischemia–reperfusion injury47. MiR-139-3p and miR-125a-5p in MSC-EVs showed similar effects on macrophage polarization. Mechanically, miR-139-3p realized this effect by suppressing signal transducer and activator of transcription 1 (STAT1)48 while miR-125a-5p acted via inhibiting TNF receptor associated factor 6/Interferon regulatory factor 5 (TRAF6/IRF5) signaling pathway.49

Besides regulating cell proliferation and differentiation, miRNAs in MSC-EV can also regulate angiogenesis, thereby improving blood supply to damaged tissues and promoting tissue repair. For example, miR-125a-3p in ADSC derived exosome can be transferred to endothelial cells to promote the angiogenesis for wound healing by inhibiting phosphatase and tensin homolog (PTEN) and activating phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway.50 MiR-221-3p was found to be upregulated in atorvastatin pretreated MSC-Exo. It can facilitate the diabetic wound regeneration by activating AKT/eNOS pathway to enhance angiogenesis.51 MiR-186-5p from MSC-EVs can be transferred to damaged human cardiac microvascular endothelial cells to develop new blood vessels by down regulating PARP9 and STAT1/pSTAT1 signaling pathway.52 Rather than directly promoting angiogenesis, MSC-EVS carried miRNA could also regulate the structure of vascular. For instance, miR-5119 was identified to be enriched in hypoxia stimulated MSC-EVs, which could improve the vascular remodeling via regulating calcium signaling pathway.53 MiR-146a, which was highly expressed in MSC-EVs, could mitigate the senescence of endothelial cell to rescue angiogenesis as these senescent cells could disrupt the integrity of normal blood vessel endothelium. The mechanism indicated that miR-146a inhibited the phosphorylation of Src and downstream VE-cadherin and Caveolin-1 to realize the stimulatory effect on senescence.54

MiRNAs in MSC-EV also have anti-inflammatory effects, helping to create a microenvironment conducive to tissue repair. MiR-155 inhibition has been demonstrated with modulating inflammatory profile in diabetic wound healing.55 On the basis of this role, miR-155 inhibitor was loaded in MSC-Exo, which showed potent anti-inflammatory effect by decreasing the level of pro-inflammatory factors (TGF-β1, IL-1β, IL-6, and TNF-α) in diabetic wounds.56 MiRNA-146a-5p in amniotic fluid derived MSC-Exo could also reduce the secretion of pro-inflammatory cytokines including IL-1β, TNF-α, and COX-2 by downregulating the expression of C-X-C chemokine receptor type 4 (CXCR4)57. Moreover, miR-146a-5p could inhibit inflammation response in a different way by ameliorating NOD-, LRR- and pyrin domain-containing protein 3 (NLRP3)-inflammasome.58,59 MiR-125a-3p in MSC-Exo was reported to improve the inflammatory environment caused by neutrophils in spinal cord injury by diminishing the formation of neutrophil extracellular traps (NETs)60. In another study, miR-199a in MSC-Exo showed similar effects on NETs formation, thereby alleviating the inflammation after myocardial ischemia/reperfusion injury.61 Another miRNA, miR-21-5p in MSC-Exo, could modulate multiple signaling molecules to inhibit inflammatory responses. On one hand, it can inhibit yes-associated protein 1 (YAP1) to attenuate the inflammation after myocardial infarction.62 On the other hand, its upregulation resulted in the downregulation of programmed cell death 4 (PDCD4) to decrease the secretion of inflammatory cytokines after retinal ischemia-reperfusion injury.63 In addition to the miRNAs mentioned above, miR-140-5p in MSC-EVs was reported to mitigate subarachnoid hemorrhage triggered neuroinflammation and brain injury through the inhibition of histone deacetylases 7 (HDAC7) for activating cAMP/PKA/CREB axis.64

Studies have shown that MSC-EV can protect damaged tissues by inhibiting cell apoptosis through the miRNA it carries. MiR-214-3p in human umbilical cord MSC-EVs was identified to increase the expression of glutathione peroxidase 4 (GPX4, an enzyme protecting against ferroptosis marker), attenuating the ferroptosis of HT22 neuronal cell and improving neuronal injury.65 MiR22 in MSC-EVs could target mitogen-activated protein kinase kinase kinase 12 (MAP3K12) to decrease caspase-3 expression, inhibiting apoptosis of retinal ganglion cells after damage.66 Collectively, as the most well-characterized contents in MSC-EVs, miRNAs play pivotal roles in the communication between MSCs and recipient cells for tissue repair.

lncRNA in MSC-EVs for Tissue Repair

LncRNA is a class of non-coding RNA with a length >200 nt, which can interact with transcription factors and RNA-binding proteins and regulate the expression of target genes in transcriptional regulation, post-transcriptional regulation and post-translational modification.67 MSC-EVs entrap a variety of lncRNAs to regulate the tissue repair through various ways (Figure 4). The most typical acting way for lncRNA is to bind with miRNA as a competitive endogenous RNA to inhibit the function of miRNA through molecular sponging effect, then regulating the miRNA-related signaling pathways. For example, lncRNA fetal-lethal non-coding developmental regulatory RNA (FENDRR) released from MSC-Exo bound to miR-28, which inhibited miR-28 to bind with TEA domain transcription factor 1 (TEAD1). As a result, the expression of TEAD1 was increased after MSC-Exo treatment, reducing the injury to human vascular endothelial cells reflecting on the decreased cell apoptosis in atherosclerosis.68 Long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 (lncRNA-MALAT1) acted as a functional molecule in human umbilical cord MSC-Exo to suppress the apoptosis of epithelial cells and improve spinal cord injury. Mechanically, lncRNA-MALAT1 reduced the inhibitory effect of miR-22-3p on its targeted sirtuin 1 (SIRT1) via miRNA sponging. The downstream signaling of SIRT1 was then activated shown as the enhanced phosphorylation of AMP‑activated protein kinase (AMPK) and increased level of antiapoptotic Bcl-2.69 LncRNA-p21 in MSC-Exo shared similar target as lncRNA-MALAT1, which bound to miR-181 to suppress its negative effect on SIRT1. Therefore, lncRNA-p21 expressed MSC-Exo protected epithelial cells against apoptosis and alleviated acute lung injury.70 Another lncRNA, long non-coding RNA alpha-2-macroglobulin antisense RNA 1 (Lnc RNA A2MAS1) in MSC-Exo also acted as a sponge with miRNA to reduce cell apoptosis for the protective effect against myocardial ischemia-reperfusion injury. In detail, exosomal lnc A2M-AS1 bound to miR-556-5p, impairing the inhibition of miR-556-5p to its target, X-linked inhibitor of apoptosis protein (XIAP).71 Besides inhibiting apoptosis, lncRNAs also regulate other roles via miRNA sponging. MSC derived exosomal lncRNA H19 was demonstrated to bind with miR-152-3p to abolish the suppression of miR-152-3p on PTEN. The increased PTEN expression then inhibited PI3K/AKT signaling pathway to promote the proliferation and migration of fibroblasts, accelerating the wound healing of diabetic foot ulcers.72 LncRNA H19 in ADSC-Exo could bind to another miRNA, miR-19b, via molecular sponge. Therefore, it upregulated the target of miR-19 (SRY-related high-mobility-group box 9, SOX9) to activate Wnt/β-catenin signaling pathway, accelerating the proliferation and migration of HSF cells in skin wound73. LncRNA krüppel-like factor 3 antisense RNA 1 (KLF3-AS1) in MSC-Exo promoted diabetic cutaneous wound healing by stimulating angiogenesis, which was mediated by interaction between lncRNA KLF3-AS1 with miR-383 to boost vascular endothelial growth factor A (VEGFA).74

Figure 4 Roles and mechanisms of lncRNAs in MSC-EVs to promote tissue repair.

Abbreviations: TEAD1, TEA Domain Transcription Factor 1; SIRT1, Sirtuin 1; XIAP, X-linked Inhibitor of Apoptosis Protein; SOX9, SRY-box Transcription Factor 9; VEGFA, Vascular Endothelial Growth Factor A; TWIST1, Twist Family BHLH Transcription Factor 1.

Instead of miRNA binding, lncRNA can directly bind with functional molecules for tissue repair. Linc00511 in ADSC-Exo was demonstrated to inhibit the ubiquitination of Twist homolog 1 (TWIST1), promoting angiogenesis for diabetic foot ulcers healing.75 LncRNA KLF3-AS1 was found to directly bind with musashi-1 (MSI1) to attenuate the inflammatory response after cerebral ischemia reperfusion injury via sphingosine kinase 1 (Sphk1)/NF-κB signaling pathway.76

CircRNA in MSC-EVs for Tissue Repair

CircRNA is a recently discovered form of non-coding RNA. The 5′ and 3′ ends of circRNA are connected to form a stable integrated circular structure, which makes circRNA more resistant to being degraded by RNA exonucleases.77

CircRNA executes its function in tissue repair via multiple modalities (Figure 5). The principal action mode of circRNA is to serve as molecular sponge, sequestering miRNAs and thus functioning as competitive endogenous RNAs (ceRNAs) to modulate the expression of relevant genes. For example, in the context of treating wounds in diabetic mice, numerous circRNAs have demonstrated promising therapeutic implications. The circ-Erbb2ip originating from ADSC-Exo promoted angiogenesis by triggering the miR-670-5p/Nrf1 axis, thus expediting wound healing in diabetic mice.78 Meanwhile, circ-Astn1 in ADSC-Exos instigated wound healing in diabetic mice via the miR-138-5p/SIRT1/FOXO1 axis.79 Moreover, circ-Snhg11 in BMSC-Exo suppressed the expression of miR-144-5p, in turn upregulating the expression of SLC7A11/GPX4 to mediate the anti-ferroptosis signaling pathway for angiogenesis.80 In addition, circARHGAP12 can directly interact with miR-301b-3p, regulating the expression of ATG16L1 and ULK2, enhancing autophagy in MSCs, and therefore inhibiting MSC apoptosis. These effects thereby promoted blood vessel reconstruction and epithelialization of MSCs in diabetic wounds.81

Figure 5 Roles and mechanisms of circRNAs in MSC-EVs to promote tissue repair.

Abbreviations: SIRT1, Sirtuin 1; XIAP, X-linked Inhibitor of Apoptosis Protein; FOXO1, Forkhead Box O1; ATG16L1, Autophagy-related Protein 16-like 1; ULK2, Unc-51-like Autophagy-activating Kinase 2; GPX4, Glutathione Peroxidase 4; SLC7A11, Solute Carrier Family 7 Member 11.

CircRNA is also capable of repairing other tissue damages such as spinal cord injury and myocardial damage. Studies have shown that hypoxic pre-conditioning of ADSC-Exo (H-ADSC-Exo), which delivered circ-Wdfy3, downregulated the expression of miR-423-3p. Concomitantly, it upregulated the expression of GPX4, decreased the levels of reactive oxygen species (ROS) and inflammatory factors following spinal cord injury, suppressed ferroptosis, and ultimately facilitated spinal cord repair.82 Likewise, H-ADSC-Exo can also reduce ferroptosis through the circ-stt3b/miR-15a-5p/GPX4 signaling pathway to alleviate cardiac damage after myocardial infarction.83

Proteins in MSC-EVs for Tissue Repair

MSC-EVs contain a great amount of protein cargoes. The clinical samples of MSC-EVs were harvested for proteomic analysis from healthy donors. Detected by mass spectrometry, more than 3000 proteins were identified in MSC-EVs while around 600 proteins were shared among different donors. Regarding to wound healing, MSC-EVs contained important proteins, such as programmed cell death protein 10, disabled homolog 2-interacting protein, neuropilin-1, tumor necrosis factor alpha-induced protein 2, Sushi repeat-containing protein and fibronectin 1 to regulate cell proliferation and angiogenesis.84

Besides these proteins, there are several other functional proteins in MSC-EVs participating in wound healing. CD73, a kind of ectoenzyme on the surface of MSCs, can convert adenosine monophosphate into adenosine to mitigate inflammatory response. CD73-overexprsssing MSC-EVs showed significant anti-inflammatory effect against autoimmune uveitis, thereby reducing tissue damages. Mechanically, the treatment of EVs inhibited the proliferation of T cells and the differentiation of Th1 cells while increased the proportion of regulatory T cells through enhancing adenosine levels.84 TGF-β has been reported to mediated partial of anti-inflammation of MSCs. It was found that TGF-β was necessary in MSC-EVs to normalize the inflammatory factors for neuroprotection in hyperammonemic rats.85 Steroid receptor coactivator-3 (SRC-3) expressed MSC-Exo was also involved in neurological actions. It protected against neuronal damage through inhibiting ferroptosis of neurons in cerebral ischemia injury.86

Moreover, proteins in MSC-EVs possess pro-angiogenic potential for tissue repair. Hypoxia-inducible factor 2-alpha (HIF2α) overexpressing MSC-EVs remarkably stimulated the tube formation ability of human umbilical vein endothelial cells (HUVECs) via upregulating the expression of miR-146a to activate AKT/ERK signaling pathway.87 Integrin α5 (Itgα5) and neuropilin-1 in MSV-C-EVs were found to regulate angiogenesis as well as lymphangiogenesis through the VEGF-C–Itgα5 and VEGF-C-neuropilin-1 axes in ischemic tissues.88 Another integrin, integrin beta-1 (ITGB1) in MSC-EVs improved the proliferation and differentiation of MSCs by activating the TGF-β/Smad2/3 axis, thereby promoting cartilage regeneration89 (Figure 6).

Figure 6 Roles and mechanisms of proteins in MSC-EVs to promote tissue repair.

Abbreviations: CD73, Cluster of Differentiation 73; T cell, T lymphocyte; Th1, T helper cell 1; HIF2α, Hypoxia-Inducible Factor 2α; AKT, Rac-alpha Serine/Threonine-Protein Kinase; ERK, Extracellular Signal-Related Kinase; Itgα5, Integrin alpha 5; VEGF, Vascular Endothelial Growth Factor; TGF-β, Transforming Growth Factor-β; Smad2/3, Sm mothers against decapentaplegic homolog 2/3.

Other Molecules in MSC-EVs for Tissue Repair

In addition to acting as transporter for nucleic acids and proteins, MSC-EVs also act as transporters for other molecules. MSC-EVs have been reported to be a kind of lipid mediators, which would transmit the oxylipins in MSCs to cardiomyoblasts to improve the inflammation in cardiac microenvironment.90 The surface glycans in MSC-EVs could be a critical factor in EV-based cell communication. The roles of N-glycosylation in Wharton’s Jelly MSC-EVs were explored. The results showed that N-glycans in MSC-EVs are essential for their interaction with HUVECs, as well as the induction of cell migration and angiogenesis.91 MSC-EVs carry a lot of bioactive molecules, and in addition to these known molecules that have been studied, more molecular recognition patterns still need to be explored.

Application of MSC-EVs for Tissue Repair

MSC-EVs have been widely applied for different tissue repairs, which were briefly summarized in Table 1.

Table 1 The Application of MSC-EVs in Diseases

MSC-Exo for Tissue Repair

Exosomes, derived from multivesicular endosomes, are extracellular vesicles with a cup-shaped morphology and a diameter ranging from 30 to 100 nm. Their predominant surface markers include CD81, CD9, CD63, and heat shock protein 70 (HSP70).104 As an intracellular mediators, they play a crucial role in bio-communication among cells.39,105 Owing to the remarkable immune regulatory properties, antioxidative stress capabilities, and anti-apoptotic effects, MSC-Exo has been effectively employed in the treatment of tissue injuries and wound healing. As the most well-studied internal factors, miRNA in MSC-Exo has been extensively explored for tissue repair. miRNA-224-3p in BMSC-Exo regulated angiogenesis, and knockdown of miRNA-224-3p expression enhanced angiogenesis and prevented the progression of traumatic osteonecrosis of the femoral head.106 Another study has shown that miR-197-3p in dental pulp MSC-Exo inhibited FOXO3 expression and reduced NLRP3 inflammasome activation to alleviate brain edema and neuroinflammation after subarachnoid hemorrhage and inhibited microglial pyroptosis.92 Furthermore, placental MSC-Exo was enriched in miR-125b-5p, miR-30c-5p, and miR-23a-3p, which targeted the expression of the 3′-untranslated region of Smad2 and Smad3, thereby down-regulating the TGF-β/Smad signaling pathway and reversing uterine fibrosis. It eventually repaired the damaged endometrium.107

Moreover, some external factors facilitated MSC-Exo to play a better role in tissue repair and regeneration. It has been reported that exosomes derived from fucoidan pretreated MSCs increased the level of miR-146b-5p. This kind of MSC-Exo effectively inhibited TRAF6 activation, thereby suppressing inflammatory responses and preventing extracellular matrix degradation. At the same time, they promoted chondrocyte autophagy and provided protection for osteoarthritic chondrocytes.93 BMSC-Exo-loaded cell-derived extracellular matrix (ECM) mimics enhanced TGF-β1-mediated activation of the Nrf2 signaling pathway and simultaneously disrupted NET, which effectively alleviated cartilage degradation in osteoarthritis.108 Nerve growth factor (NGF)-stimulated MSC-Exo transferred multi-component miRNAs and activated MAPK and PI3K-AKT signaling pathways, resulting in the formation of massive nerve and vascular structures and significant innervation of bone regeneration.109

Owing to the destruction of blood supply and the generation of anoxic environment, the damaged tissues often produce a large amount of ROS, which would lead to oxidative stress and the destruction of mitochondrial homeostasis. MSC-Exo can restore and maintain mitochondrial homeostasis by promoting mitochondrial biogenesis, enhancing mitochondrial function, regulating mitophagy, and reducing mitochondrial damage. For instance, hypoxia-preconditioned hair follicle mesenchymal stem cells-derived exosomes can reduce inflammatory injury in ulcerative colitis by inhibiting the PI3K/AKT/mTOR signaling pathway, maintaining mitochondrial dynamic stability, reducing mitochondrial dysfunction, as well as enhancing mitophagy.94 Liu et al developed a bioenergetic exosome (Suc-Exo). The ATP content of Suc-Exo was 5.42-fold higher than that of ordinary exosomes, which could regulate cellular energy metabolism by providing fuel for the TCA cycle. Under continuous energy supply, Suc-Exo promoted chondrogenic differentiation of BMSC and improved chondrocyte anabolism and mitochondrial homeostasis.110 In another study, a kind of engineered exosome loaded with Sephin1 was constructed to activate mitophagy and eliminate mtROS to maintain ER homeostasis, promoting bone tissue repair, and shortening bone healing time in diabetic bone defects.95

In addition, MSC-Exo from different sources could exert various functions. Three-dimensional (3D) cell culture conditions of mesenchymal stem cell could potentiate the immune homeostasis and tissue regeneration ability of secreted exosomes. Wang et al found that 3D-cultured hUMSC-Exo enhanced Treg-mediated immunosuppression in vitiligo lesions and deliver miR-132-3p and miR-125b-5p to inhibit oxidative stress-induced melanoma cell damage.96 Xu et al reported that 3D cultured MSC-Exo promoted the proliferation of corneal-derived cells and reduced the release of inflammatory factors by targeting the PDCD4 gene through miR-150-5p. It restored corneal morphology and function, reduced inflammation, promoted corneal epithelium and limbus repair, and reduced scar formation in the stroma.97 The preconditioning of MSC under hypoxic environments has gained increasing attention for the potential to promote the secretion and functionality of exosome. For example, hypoxia-induced ADSC-Exo mediated miR-21-5p targeting sprouly1 in HUVEC to promote the activation of the PI3K/AKT signaling pathway, thereby enhancing HUVEC proliferation, migration, and angiogenesis and concomitant bone regeneration.111 And hypoxia-induced BMSC-Exo delivered Bcl-2/adenovirus E1B 19 kDa-interacting protein 3 (BNIP3)-rich vesicles to alleviate intervertebral disc degeneration by activating the mitochondrial BNIP3/ANXA2/TFEB axis, which may provide a new target for the treatment of intervertebral disc lesion.98

The effects of MSC-Exo administration alone are limited, but the therapeutic effects can be enhanced by engineering or drug combinations. For example, the modification of upper central nervous system-specific rabies virus glycoprotein peptide on MSC-Exo can rescue the memory defects of AD mice.112 MSC-Exo modified with lncRNA MIR155HG attenuated intimal hyperplasia of vein grafts following coronary artery bypass grafting.113 When combining MSC-Exo with metformin, MSC-Exo can act as an anti-inflammatory agent without altering the dosage of metformin, thereby reducing the production of pro-inflammatory cytokines (including IL-1, IL-6, and TNF-α) and enhancing the effectiveness of metformin.114 Lu et al developed a smart bilayer hydrogel loaded with diclofenac sodium (an anti-inflammatory drug) and BMSC-Exo, which reduced endogenous ROS production, promoted M2-type polarization of macrophages, regulated the inflammatory microenvironment, and promoted the recruitment and chondrogenic differentiation of late stem cell.115 Human endometrial MSC-Exo was incorporated into polypyrrole chitosan to obtain an injectable hydrogel. The hydrogel reduced H2O2-induced apoptosis and promoted renal tubule formation, improving cardiac function after myocardial infarction (MI).116

Besides, MSC-Exo can also inhibit the apoptosis of renal tubular epithelial cells to effectively treat acute kidney injury,117 repair diabetic lower limb ischemic injury,118 improve the liver fibrosis environment,119 reduce pulmonary vascular remodeling, successfully reverse pulmonary arterial hypertension,120 and prevent experimental autoimmune disease progression in uveoretinitis121 and repair damaged myocardium.122 In summary, MSC-Exo, as a small-molecule non-cellular therapeutic strategy, can be widely used in various diseases to promote tissue damage and wound repair due to its biological characteristics.

MSC Derived Microvesicles for Tissue Repair

The origin of MVs can be traced back to the cell membrane, with the diameter ranging from 100 nm to 1000 nm. These vesicles are fragments derived from cells that remain attached to both the nuclear and plasma membranes. Following shedding from the cell membrane, MVs exhibit targeting specificity and selectively interact with specific ligands to exert their effects.123 The MVs also serve as carriers of proteins, nucleic acids, lipids, and other bioactive substances.124 It has been demonstrated that MVs show anti-apoptosis, anti-inflammation, and angiogenesis effects for tissue repair and wound healing.125 The clinical application of MSC-MVs emerged relatively later compared to MSC-Exo. It was developed in the past decade and witnessed rapid advancements and extensive utilization in various medical fields, including orthopedics, hepatology, pulmonology, nephrology, neurology, and other disciplines.

MSC-MV exhibits the capability of promoting bone repair and protecting cartilage tissue. Liang et al utilized the carrier function of MSC-MVs to construct a gene-activated scaffold through layer-by-layer assembly, which significantly facilitated angiogenesis for bone repair.126 In collagen-induced arthritis mice, intravenous injection of MSC-MVs downregulated the expression of immune cell inflammatory factors (IL-1β, TNFα), MMP-13, and ADAMTS-5 in cartilage while maintained the stability of type I and II collagen, significantly reducing early cartilage destruction.99 It has been shown that electrostatic field enhanced the release of mitochondria containing-MSC-MV, which played a key role in delaying cell aging and mitochondrial dysfunction, providing a new possibility for the treatment of intervertebral disc degeneration.127

The application of MSC-MV has been demonstrated in the repair of kidney injury. A study on kidney injury revealed that human umbilical cord MSC-MVs effectively delivered miR-21 to target PDCD4 in the renal tubular epithelium, inhibiting cell apoptosis and improving renal ischemia-reperfusion injury.100 The administration of Wharton’s jelly MSC-MVs effectively induced M2 macrophage polarization to mitigate renal fibrosis.128

MSC-MVs can also be used for neurological injury. MPTP (1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine) induces neurotoxicity resembling Parkinson’s disease in mice. MSC-MV exhibited the potential to modulate the gut-microbiota-brain axis for ameliorating this neurotoxicity in both the brain and colon. In another study, MSC-MV can effectively down-regulate TNF-α and p-AKT levels to improving the neuroinflammatory symptoms in Alzheimer’s disease. Moreover, it also reduced the apoptosis and improved the hippocampal and cortical histopathological alterations.101

MSC-Derived Apoptotic Bodies for Tissue Repair

The apoptotic bodies are large extracellular vesicles, ranging in diameter from 500 to 2000 nm, that are generated during the process of cell apoptosis. Apoptotic MSCs dismantle organelles and other materials into fragments, which are then encapsulated within distinct membrane-bound vesicles. These apoptotic bodies also encompass a diverse array of cellular components, including proteins, lipids, DNA, mRNA, and miRNAs.129 They can act as an intercellular communication mediator, being involved in the regulation of immune response, cell proliferation, compensatory tissue regeneration, and other physiological processes.130 Compared to exosomes and microvesicles, the therapeutic application of apoptotic bodies is relatively limited, leaving ample room for exploration.

MSC-derived apoptotic bodies have the potential to enhance wound healing through inducing macrophage polarization towards the M2 phenotype, suppress proinflammatory cytokine expression, and facilitate skin wound healing.131 In another study, a kind of hyaluronic acid hydrogel loaded with MSC-derived apoptotic bodies was developed. The gel-system induced macrophage polarization towards the M2 phenotype and significantly suppressed the expression of proinflammatory cytokines, including IL-1β, TNFα, IL-6, and IFN-γ, facilitating endometrial regeneration.102

The application of MSC-derived apoptotic bodies in periodontitis treatment is also feasible. They enhanced the expression of dendritic cell-specific transmembrane protein on osteoclast membranes and facilitated the phagocytosis of apoptotic bodies by osteoclasts. Moreover, they effectively mitigated periodontitis-induced alveolar bone loss through miR-223-3p transferring to osteoclasts.132

In addition, apoptotic bodies released from transplanted MSCs could be engulfed by endothelial cells (ECs). The phagocytic apoptotic bodies stimulated the expression of transcription factor EB, which in turn enhanced the expression of autophagy-related genes in ECs and facilitated cardiac function recovery following myocardial infarction.103 A recent study showed that hsa-miR-4485-3p in MSC apoptotic bodies inhibited osteogenesis by down-regulating the AKT pathway, and apoptotic bodies customized by knocking down hsa-miR-4485-3p exhibited potent osteogenic induction.133 Apoptotic bodies have the potential for more comprehensive applications, and further researches and developments are needed to explore more application of apoptotic bodies generated by mesenchymal stem cells.

Clinical Progress in MSC-Derived Extracellular Vesicles for Tissue Regeneration

At present, extensive studies on MSC-EVs have focused on the basic biological functions and mechanisms of extracellular vesicles. The efficacy of MSC-EVs in various diseases is still restricted in the cellular and animal levels, and only a few MSC-EVs have entered the early clinical research stage. In a Phase I clinical trial, clinical-grade nebulized allogenic adipose mesenchymal stromal cell-derived extracellular vesicles were used to verify their safety in 24 healthy volunteers. The results showed that aerosolized inhalation of MSC-EVs was safe without serious side effects.134 In another single-arm, open-label, phase I clinical trial, nine patients with complete subacute spinal cord injury received intrathecal human umbilical cord MSCs-Exos and were followed up for 12 months. During this period, no early or late adverse events attributed by the study intervention were observed. At the same time, the patients also had significant improvements in ASIA pinprick (P  = 0.039) and light touch score (P = 0.038), SCIM III total score (P = 0.027), and NBD score (P = 0.042) at 12 months after the injection.135 In a Phase I clinical trial for the treatment of psoriasis, MSC-Exo ointment was applied topically to the forearm of 10 healthy subjects for 24 days. There were no drug-related adverse events during the study period, and the subjects showed no adverse reactions such as dryness, pruritus, exudation/crusting, redness, scratching, skin thickening, insomnia, or swelling in the application area.136 Additionally, a human adipose tissue stem cell-derived exosome-containing solution was enrolled in a Phase II clinical trial. Twenty-eight individuals participated in this study and were followed up for 12 weeks. At the last follow-up, the global aesthetic improvement scale score of the exosome-treated side was significantly higher than that of the control group (p = 0.005). The clinical improvement of skin wrinkles, elasticity, hydration, and pigmentation on the exosome-treated side was greater than that in the control group. No serious adverse events were observed.137

In 2019, the outbreak and rapid development of COVID-19 posed a significant threat to human life safety. The researches on MSC-EVs for the treatment of COVID-19 developed rapidly, and some studies have entered the Phase II clinical trial cycle. Human placental MSC-EVs were administered to two groups of patients with COVID-associated acute respiratory distress syndrome, noted as the control group and the intervention group. Compared with the control group (13/24 [54.16%]), MSC-EVs significantly reduced the mortality in the intervention group (4/21 [19.04%]) (p = 0.015). The mean time to death was 11.10 days in the control group compared with that of 28.06 days in the intervention group (p < 0.001).138 Bone marrow MSC-EVs were also investigated for the treatment of COVID-19 respiratory failure in a prospective phase II, multicenter, double-anonymous, randomized, placebo-controlled administration trial. The post hoc subgroup analyses showed lower 60-day mortality with MSC-EVs than that with placebo (relative risk, 0.385; 95% CI, 0.159–0.931; P = 0340; n = 50).139 Further, a phase IIa, single-arm, open-label, interventional trial (MEXCOVID trial) examined the safety and efficacy of aerosolized human adipose - derived MSC-derived exosomes (haMSC-Exos) in COVID-19 patients. Subsequently, the 5-day trial demonstrated that the COVID-19 patients had good tolerance, and all patients exhibited varying degrees of improvement in their lung lesions.140

As a cell-free therapy, MSC-EVs have shown the potential to treat various diseases. Although the clinical progress of MSC-EVs is still in the early stage (Table 2), a growing number of preliminary researches have laid a solid foundation for their subsequent development and will make continuous breakthroughs to bring insights for tissue repair.

Table 2 The Summary of Clinical Study About MSC-EVs for Tissue Repair

Perspective and Conclusion

MSC-EVs are nanoscale particles paracrine from MSCs. They contain many bioactive molecules, including miRNA, lncRNA, proteins, and lipids that came from donor cells. These biologically active molecules have great potential to promote tissue repair. In recent years, MSC-EVs have become the focus of research in the field of cell-free therapy and regenerative medicine due to their participation in intercellular communication, anti-inflammation, immune regulation, angiogenesis, and other functions. A small number of MSC-EVs have been put into clinical trials. MSC-EVs are expected to become a new cell-free therapy for various diseases.

Despite MSC-EVs having shown promising results in laboratory studies, there are still many challenges in how to translate them into clinical applications. Firstly, the standardization and scale-up production of MSCs still present formidable challenges. The manufacturing process for MSC- EVs entails intricate bioprocessing steps encompassing cell isolation, cultivation, collection, and purification. Secondly, it is difficult to control the quality of the final product of MSC-EVs. Both MSCs and EVs are biological products, and every step in their production process may affect the quality and effectiveness of the final product. Additionally, the clinical application of MSCs and EVs necessitates rigorous validation through comprehensive clinical trials to ensure their safety and efficacy. Therefore, continuous enhancement of the production and quality control processes for MSC-EVs will remain imperative in the future.

In conclusion, this review summarized the characteristics of three type of MSC-EVs including MSC-Exo, MSC-MV and MSC-derived apoptotic bodies. We detailly discussed the bioactive molecules involving miRNAs, lncRNAs, circRNAs and proteins in MSC-EVs for tissue repair. These functional molecules could regulate the cell proliferation, differentiation and apoptosis, angiogenesis as well as inflammatory response during the tissue repair process. We briefly described the application of different types of MSC-EVs in the treatment of various tissue injuries. We also reviewed the clinical progress and remained challenges of MSC-EVs for tissue repair. In the future, efforts should be focused on how to achieve the clinical translation and ensure safety and efficacy of MSC-EVs, with the ultimate goal of benefiting mankind.

Data Sharing Statement

Data sharing not applicable – no new data generated.

Author Contributions

All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

Funding

This work was supported by National Natural Science Foundation of China (82274026).

Disclosure

The authors declare that they have no competing interests in this work.

References

1. Schuster R, Younesi F, Ezzo M, Hinz B. The role of myofibroblasts in physiological and pathological tissue repair. Cold Spring Harb Perspect Biol. 2023;15(1):a041231. doi:10.1101/cshperspect.a041231

2. GBD 2021 Osteoarthritis Collaborators. Global, regional, and national burden of osteoarthritis, 1990-2020 and projections to 2050: a systematic analysis for the global burden of disease study 2021. Lancet Rheumatol. 2023;5(9):e508–e522. doi:10.1016/S2665-9913(23)00163-7

3. Tang F, Zhao XL, Xu LY, Zhang JN, Ao H, Peng C. Endothelial dysfunction: pathophysiology and therapeutic targets for sepsis-induced multiple organ dysfunction syndrome. Biomed Pharmacother. 2024;178:117180. doi:10.1016/j.biopha.2024.117180

4. Hu JM, Hsu CH, Lin YC, et al. Ethyl pyruvate ameliorates heat stroke-induced multiple organ dysfunction and inflammatory responses by induction of stress proteins and activation of autophagy in rats. Int J Hyperthermia. 2021;38(1):862–874. doi:10.1080/02656736.2021.1931479

5. Santoro A, Voto A, Fortino L, et al. Bone defect treatment in regenerative medicine: exploring natural and synthetic bone substitutes. Int J Mol Sci. 2025;26(7):3085. doi:10.3390/ijms26073085

6. Xiao T, Yan Z, Xiao S, Xia Y. Proinflammatory cytokines regulate epidermal stem cells in wound epithelialization. Stem Cell Res Ther. 2020;11(1):232. doi:10.1186/s13287-020-01755-y

7. Maruyama M, Rhee C, Utsunomiya T, et al. Modulation of the inflammatory response and bone healing. Front Endocrinol. 2020;11:386. doi:10.3389/fendo.2020.00386

8. Raziyeva K, Kim Y, Zharkinbekov Z, Kassymbek K, Jimi S, Saparov A. Immunology of acute and chronic wound healing. Biomolecules. 2021;11(5):700. doi:10.3390/biom11050700

9. Caratti G, Desgeorges T, Juban G, et al. Macrophagic AMPKα1 orchestrates regenerative inflammation induced by glucocorticoids. EMBO Rep. 2023;24(2):e55363. doi:10.15252/embr.202255363

10. Caballero-Sánchez N, Alonso-Alonso S, Nagy L. Regenerative inflammation: when immune cells help to re-build tissues. FEBS J. 2024;291(8):1597–1614. doi:10.1111/febs.16693

11. Diomede F, Marconi GD, Fonticoli L, et al. Functional relationship between osteogenesis and angiogenesis in tissue regeneration. Int J Mol Sci. 2020;21(9):3242. doi:10.3390/ijms21093242

12. Nourian Dehkordi A, Mirahmadi Babaheydari F, Chehelgerdi M, Raeisi Dehkordi S. Skin tissue engineering: wound healing based on stem-cell-based therapeutic strategies. Stem Cell Res Ther. 2019;10(1):111. doi:10.1186/s13287-019-1212-2

13. Mony MP, Harmon KA, Hess R, Dorafshar AH, Shafikhani SH. An updated review of hypertrophic scarring. Cells. 2023;12(5):678. doi:10.3390/cells12050678

14. Xiang E, Han B, Zhang Q, et al. Human umbilical cord-derived mesenchymal stem cells prevent the progression of early diabetic nephropathy through inhibiting inflammation and fibrosis. Stem Cell Res Ther. 2020;11(1):336. doi:10.1186/s13287-020-01852-y

15. Yan Z, Qin H, Ren J, Qu X. Photocontrolled multidirectional differentiation of mesenchymal stem cells on an upconversion substrate. Angew Chem Int Ed Engl. 2018;57(35):11182–11187. doi:10.1002/anie.201803939

16. Wang Y, Fang J, Liu B, Shao C, Shi Y. Reciprocal regulation of mesenchymal stem cells and immune responses. Cell Stem Cell. 2022;29(11):1515–1530. doi:10.1016/j.stem.2022.10.001

17. Viswanathan S, Shi Y, Galipeau J, et al. Mesenchymal stem versus stromal cells: international society for cell & gene therapy (ISCT®) mesenchymal stromal cell committee position statement on nomenclature. Cytotherapy. 2019;21(10):1019–1024. doi:10.1016/j.jcyt.2019.08.002

18. Mebarki M, Abadie C, Larghero J, Cras A. Human umbilical cord-derived mesenchymal stem/stromal cells: a promising candidate for the development of advanced therapy medicinal products. Stem Cell Res Ther. 2021;12(1):152. doi:10.1186/s13287-021-02222-y

19. Andrzejewska A, Lukomska B, Janowski M. Concise review: mesenchymal stem cells: from roots to boost. Stem Cells. 2019;37(7):855–864. doi:10.1002/stem.3016

20. Saleh M, Taher M, Sohrabpour AA, et al. Perspective of placenta derived mesenchymal stem cells in acute liver failure. Cell Biosci. 2020;10:71. doi:10.1186/s13578-020-00433-z

21. Bagdasarian FA, Yuan X, Athey J, Bunnell BA, Grant SC. NODDI highlights recovery mechanisms in white and gray matter in ischemic stroke following human stem cell treatment. Magn Reson Med. 2021;86(6):3211–3223. doi:10.1002/mrm.28929

22. Harrell CR, Gazdic M, Fellabaum C, et al. Therapeutic potential of amniotic fluid derived mesenchymal stem cells based on their differentiation capacity and immunomodulatory properties. Curr Stem Cell Res Ther. 2019;14(4):327–336. doi:10.2174/1574888X14666190222201749

23. Abbaszadeh H, Ghorbani F, Derakhshani M, et al. Regenerative potential of Wharton’s jelly-derived mesenchymal stem cells: a new horizon of stem cell therapy. J Cell Physiol. 2020;235(12):9230–9240. doi:10.1002/jcp.29810

24. Niebergall-Roth E, Frank NY, Ganss C, Frank MH, Kluth MA. Skin-derived ABCB5+ mesenchymal stem cells for high-medical-need inflammatory diseases: from discovery to entering clinical routine. Int J Mol Sci. 2022;24(1):66. doi:10.3390/ijms24010066

25. Li C, Zhao H, Cheng L, Wang B. Allogeneic vs. autologous mesenchymal stem/stromal cells in their medication practice. Cell Biosci. 2021;11(1):187. doi:10.1186/s13578-021-00698-y

26. Luo H, Birjandi AA, Ren F, et al. Advances in oral mesenchymal stem cell-derived extracellular vesicles in health and disease. Genes Dis. 2023;11(1):346–357. doi:10.1016/j.gendis.2023.03.015

27. Shi X, Mao J, Liu Y. Pulp stem cells derived from human permanent and deciduous teeth: biological characteristics and therapeutic applications. Stem Cells Transl Med. 2020;9(4):445–464. doi:10.1002/sctm.19-0398

28. Heng BC, Bai Y, Li X, Zhang X, Deng X. Extrapolating neurogenesis of mesenchymal stem/stromal cells on electroactive and electroconductive scaffolds to dental and oral-derived stem cells. Int J Oral Sci. 2022;14(1):13. doi:10.1038/s41368-022-00164-6

29. Alarcón-Apablaza J, Prieto R, Rojas M, Fuentes R. Potential of oral cavity stem cells for bone regeneration: a scoping review. Cells. 2023;12(10):1392. doi:10.3390/cells12101392

30. Shekatkar M, Kheur S, Deshpande S, et al. Angiogenic potential of various oral cavity-derived mesenchymal stem cells and cell-derived secretome: a systematic review and meta-analysis. Eur J Dent. 2024;18(3):712–742. doi:10.1055/s-0043-1776315

31. Cone AS, Yuan X, Sun L, et al. Mesenchymal stem cell-derived extracellular vesicles ameliorate Alzheimer’s disease-like phenotypes in a preclinical mouse model. Theranostics. 2021;11(17):8129–8142. doi:10.7150/thno.62069

32. Li A, Guo F, Pan Q, et al. Mesenchymal stem cell therapy: hope for patients with systemic lupus erythematosus. Front Immunol. 2021;12:728190. doi:10.3389/fimmu.2021.728190

33. Kalluri R. The biology and function of extracellular vesicles in immune response and immunity. Immunity. 2024;57(8):1752–1768. doi:10.1016/j.immuni.2024.07.009

34. Tieu A, Lalu MM, Slobodian M, et al. An analysis of mesenchymal stem cell-derived extracellular vesicles for preclinical use. ACS Nano. 2020;14(8):9728–9743. doi:10.1021/acsnano.0c01363

35. Ong WK, Chakraborty S, Sugii S. Adipose tissue: understanding the heterogeneity of stem cells for regenerative medicine. Biomolecules. 2021;11(7):918. doi:10.3390/biom11070918

36. Maumus M, Rozier P, Boulestreau J, Jorgensen C, Noël D. Mesenchymal stem cell-derived extracellular vesicles: opportunities and challenges for clinical translation. Front Bioeng Biotechnol. 2020;8:997. doi:10.3389/fbioe.2020.00997

37. Kim HY, Kwon S, Um W, et al. Functional extracellular vesicles for regenerative medicine. Small. 2022;18(36):e2106569. doi:10.1002/smll.202106569

38. Wu T, Wang L, Gao C, et al. Treg-derived extracellular vesicles: roles in diseases and theranostics. Mol Pharm. 2024;21(6):2659–2672. doi:10.1021/acs.molpharmaceut.4c00233

39. Wang Y, Xiao T, Zhao C, Li G. The regulation of exosome generation and function in physiological and pathological processes. Int J Mol Sci. 2023;25(1):255. doi:10.3390/ijms25010255

40. De Sousa KP, Rossi I, Abdullahi M, Ramirez MI, Stratton D, Inal JM. Isolation and characterization of extracellular vesicles and future directions in diagnosis and therapy. Wiley Interdiscip Rev Nanomed Nanobiotechnol. 2023;15(1):e1835. doi:10.1002/wnan.1835

41. Zhang Y, Pan Y, Liu Y, et al. Exosomes derived from human umbilical cord blood mesenchymal stem cells stimulate regenerative wound healing via transforming growth factor-β receptor inhibition. Stem Cell Res Ther. 2021;12(1):434. doi:10.1186/s13287-021-02517-0

42. Lv Q, Deng J, Chen Y, Wang Y, Liu B, Liu J. Engineered human adipose stem-cell-derived exosomes loaded with mir-21-5p to promote diabetic cutaneous wound healing. Mol Pharm. 2020;17(5):1723–1733. doi:10.1021/acs.molpharmaceut.0c00177

43. Shen C, Tao C, Zhang A, et al. Exosomal microRNA⁃93⁃3p secreted by bone marrow mesenchymal stem cells downregulates apoptotic peptidase activating factor 1 to promote wound healing. Bioengineered. 2022;13(1):27–37. doi:10.1080/21655979.2021.1997077

44. Yang C, Luo L, Bai X, et al. Highly-expressed micoRNA-21 in adipose derived stem cell exosomes can enhance the migration and proliferation of the HaCaT cells by increasing the MMP-9 expression through the PI3K/AKT pathway. Arch Biochem Biophys. 2020;681:108259. doi:10.1016/j.abb.2020.108259

45. Nataliya B, Mikhail A, Vladimir P, et al. Mesenchymal stromal cells facilitate resolution of pulmonary fibrosis by miR-29c and miR-129 intercellular transfer. Exp Mol Med. 2023;55(7):1399–1412. doi:10.1038/s12276-023-01017-w

46. Su J, Wei Q, Ma K, et al. P-MSC-derived extracellular vesicles facilitate diabetic wound healing via miR-145-5p/ CDKN1A-mediated functional improvements of high glucose-induced senescent fibroblasts. Burns Trauma. 2023;11:tkad010. doi:10.1093/burnst/tkad010

47. Hou Z, Yang F, Chen K, Wang Y, Qin J, Liang F. hUC-MSC-EV-miR-24 enhances the protective effect of dexmedetomidine preconditioning against myocardial ischemia-reperfusion injury through the KEAP1/Nrf2/HO-1 signaling. Drug Deliv Transl Res. 2024;14(1):143–157. doi:10.1007/s13346-023-01388-7

48. Ning Y, Huang P, Chen G, et al. Atorvastatin-pretreated mesenchymal stem cell-derived extracellular vesicles promote cardiac repair after myocardial infarction via shifting macrophage polarization by targeting microRNA-139-3p/Stat1 pathway. BMC Med. 2023;21(1):96. doi:10.1186/s12916-023-02778-x

49. Gong ZT, Xiong YY, Ning Y, et al. Nicorandil-pretreated mesenchymal stem cell-derived exosomes facilitate cardiac repair after myocardial infarction via promoting macrophage m2 polarization by targeting miR-125a-5p/TRAF6/IRF5 signaling pathway. Int J Nanomed. 2024;19:2005–2024. doi:10.2147/IJN.S441307

50. Pi L, Yang L, Fang BR, Meng XX, Qian L. Exosomal microRNA-125a-3p from human adipose-derived mesenchymal stem cells promotes angiogenesis of wound healing through inhibiting PTEN. Mol Cell Biochem. 2022;477(1):115–127. doi:10.1007/s11010-021-04251-w

51. Yu M, Liu W, Li J, et al. Exosomes derived from atorvastatin-pretreated MSC accelerate diabetic wound repair by enhancing angiogenesis via AKT/eNOS pathway. Stem Cell Res Ther. 2020;11(1):350. doi:10.1186/s13287-020-01824-2

52. Zhang S, Li D, Liu L, Shi Q, Ju X. Extracellular vesicles derived from HuMSCs alleviate daunorubicin-induced cardiac microvascular injury via miR-186-5p/PARP9/STAT1 signal pathway. Regen Ther. 2024;25:320–330. doi:10.1016/j.reth.2024.01.011

53. Zhu Q, Mao X, Zhu X, et al. Hypoxia-induced and glucuronic acid-modified extracellular vesicles from mesenchymal stromal cells treat pulmonary arterial hypertension by improving vascular remodeling. Nano Lett. 2024;24(51):16342–16350. doi:10.1021/acs.nanolett.4c04638

54. Xiao X, Xu M, Yu H, et al. Correction to: mesenchymal stem cell-derived small extracellular vesicles mitigate oxidative stress-induced senescence in endothelial cells via regulation of miR-146a/Src. Signal Transduct Target Ther. 2022;7(1):234. doi:10.1038/s41392-022-01075-y

55. Ye J, Kang Y, Sun X, Ni P, Wu M, Lu S. MicroRNA-155 inhibition promoted wound healing in diabetic rats. Int J Low Extrem Wounds. 2017;16(2):74–84. doi:10.1177/1534734617706636

56. Gondaliya P, Sayyed AA, Bhat P, et al. Mesenchymal stem cell-derived exosomes loaded with mir-155 inhibitor ameliorate diabetic wound healing. Mol Pharm. 2022;19(5):1294–1308. doi:10.1021/acs.molpharmaceut.1c00669

57. Wgealla MMAMA, Liang H, Chen R, et al. Amniotic fluid derived stem cells promote skin regeneration and alleviate scar formation through exosomal miRNA-146a-5p via targeting CXCR4. J Cosmet Dermatol. 2022;21(10):5026–5036. doi:10.1111/jocd.14956

58. Liu J, Xu Z, Yu J, et al. MiR-146a-5p engineered hucMSC-derived extracellular vesicles attenuate Dermatophagoides farinae-induced allergic airway epithelial cell inflammation. Front Immunol. 2024;15:1443166. doi:10.3389/fimmu.2024.1443166

59. Mellado S, Morillo-Bargues MJ, Perpiñá-Clérigues C, et al. The emerging role of mesenchymal stem cell-derived extracellular vesicles to ameliorate hippocampal NLRP3 inflammation induced by binge-like ethanol treatment in adolescence. Neural Regen Res. 2025;20(4):1153–1163. doi:10.4103/NRR.NRR-D-23-01397

60. Morishima Y, Kawabori M, Yamazaki K, et al. Intravenous administration of mesenchymal stem cell-derived exosome alleviates spinal cord injury by regulating neutrophil extracellular trap formation through exosomal mir-125a-3p. Int J Mol Sci. 2024;25(4):2406. doi:10.3390/ijms25042406

61. Feng Y, Bao X, Zhao J, Kang L, Sun X, Xu B. Msc-derived exosomes mitigate myocardial ischemia/reperfusion injury by reducing neutrophil infiltration and the formation of neutrophil extracellular traps. Int J Nanomed. 2024;19:2071–2090. doi:10.2147/IJN.S436925

62. Ji Z, Wang C. Mesenchymal stem cell-derived exosomal mir-21-5p inhibits YAP1 expression and improves outcomes in myocardial infarction. BMC Cardiovasc Disord. 2024;24(1):547. doi:10.1186/s12872-024-04197-z

63. Yu Z, Wen Y, Jiang N, et al. TNF-α stimulation enhances the neuroprotective effects of gingival MSCs derived exosomes in retinal ischemia-reperfusion injury via the MEG3/miR-21a-5p axis. Biomaterials. 2022;284:121484. doi:10.1016/j.biomaterials.2022.121484

64. Qian Y, Chen B, Sun E, et al. Mesenchymal stem cell-derived extracellular vesicles alleviate brain damage following subarachnoid hemorrhage via the interaction of mir-140-5p and HDAC7. Mol Neurobiol. 2024;61(11):9136–9154. doi:10.1007/s12035-024-04118-3

65. Yang Y, Gao L, Xi J, et al. Mesenchymal stem cell-derived extracellular vesicles mitigate neuronal damage from intracerebral hemorrhage by modulating ferroptosis. Stem Cell Res Ther. 2024;15(1):255. doi:10.1186/s13287-024-03879-x

66. Yu B, Wang K, Hao H, et al. Small extracellular vesicles derived from microRNA-22-3p-overexpressing mesenchymal stem cells protect retinal ganglion cells by regulating MAPK pathway. Commun Biol. 2024;7(1):807. doi:10.1038/s42003-024-06511-z

67. Nojima T, Proudfoot NJ. Mechanisms of lncRNA biogenesis as revealed by nascent transcriptomics. Nat Rev Mol Cell Biol. 2022;23(6):389–406. doi:10.1038/s41580-021-00447-6

68. Zhang N, Luo Y, Zhang H, Zhang F, Gao X, Shao J. Exosomes derived from mesenchymal stem cells ameliorate the progression of atherosclerosis in ApoE mice via FENDRR. Cardiovasc Toxicol. 2022;22(6):528–544. doi:10.1007/s12012-022-09736-8-/-

69. Li K, Liu Z, Wu P, et al. Micro electrical fields induced MSC-sEVs attenuate neuronal cell apoptosis by activating autophagy via lncRNA MALAT1/miR-22-3p/SIRT1/AMPK axis in spinal cord injury. J Nanobiotechnology. 2023;21(1):451. doi:10.1186/s12951-023-02217-2

70. Sui X, Liu W, Liu Z. Exosomal lncRNA-p21 derived from mesenchymal stem cells protects epithelial cells during LPS-induced acute lung injury by sponging miR-181. Acta Biochim Biophys Sin. 2021;53(6):748–757. doi:10.1093/abbs/gmab043

71. Yu H, Pan Y, Dai M, Wang X, Chen H. Mesenchymal stem cell-originated exosomal Lnc A2M-AS1 alleviates hypoxia/reperfusion-induced apoptosis and oxidative stress in cardiomyocytes. Cardiovasc Drugs Ther. 2023;37(5):891–904. doi:10.1007/s10557-022-07339-7

72. Li B, Luan S, Chen J, et al. The MSC-derived exosomal lncRNA H19 promotes wound healing in diabetic foot ulcers by upregulating PTEN via microRNA-152-3p. Mol Ther Nucleic Acids. 2020;19:814–826. doi:10.1016/j.omtn.2019.11.034

73. Qian L, Pi L, Fang BR, Meng XX. Adipose mesenchymal stem cell-derived exosomes accelerate skin wound healing via the lncRNA H19/miR-19b/SOX9 axis. Lab Invest. 2021;101(9):1254–1266. doi:10.1038/s41374-021-00611-8

74. Han ZF, Cao JH, Liu ZY, Yang Z, Qi RX, Xu HL. Exosomal lncRNA KLF3-AS1 derived from bone marrow mesenchymal stem cells stimulates angiogenesis to promote diabetic cutaneous wound healing. Diabet Res Clin Pract. 2022;183:109126. doi:10.1016/j.diabres.2021.109126

75. Qiu J, Shu C, Li X, Ye C, Zhang WC. Exosomes from linc00511-overexpressing ADSCs accelerates angiogenesis in diabetic foot ulcers healing by suppressing PAQR3-induced twist1 degradation. Diabet Res Clin Pract. 2021;180:109032. doi:10.1016/j.diabres.2021.109032

76. Cao Y, Wang D, Zhou D. MSC promotes the secretion of exosomal lncRNA KLF3-AS1 to regulate sphk1 through YY1-musashi-1 axis and improve cerebral ischemia-reperfusion injury. Mol Neurobiol. 2024;61(12):10462–10480. doi:10.1007/s12035-024-04150-3

77. Wang Y, Mo Y, Peng M, et al. The influence of circular RNAs on autophagy and disease progression. Autophagy. 2022;18(2):240–253. doi:10.1080/15548627.2021.1917131

78. Tang W, Du X, Wu Z, Nie Z, Yu C, Gao Y. circ-Erbb2ip from adipose-derived mesenchymal stem cell-derived exosomes promotes wound healing in diabetic mice by inducing the miR-670-5p/Nrf1 axis. Cell Signal. 2024;121:111245. doi:10.1016/j.cellsig.2024.111245

79. Wang Z, Feng C, Liu H, et al. Exosomes from circ-Astn1-modified adipose-derived mesenchymal stem cells enhance wound healing through miR-138-5p/SIRT1/FOXO1 axis regulation. World J Stem Cells. 2023;15(5):476–489. doi:10.4252/wjsc.v15.i5.476

80. Tang T, Chen L, Zhang M, et al. Exosomes derived from BMSCs enhance diabetic wound healing through circ-Snhg11 delivery. Diabetol Metab Syndr. 2024;16(1):37. doi:10.1186/s13098-023-01210-x

81. Meng F, Shen F, Ling H, Jin P, Zhou D, Li Q. CircARHGAP12 triggers mesenchymal stromal cell autophagy to facilitate its effect on repairing diabetic wounds by sponging mir-301b-3p/atg16l1 and mir-301b-3p/ulk2. J Invest Dermatol. 2022;142(7):1976–1989.e4. doi:10.1016/j.jid.2021.11.039

82. Shao M, Ye S, Chen Y, Yu C, Zhu W. Exosomes from hypoxic ADSCs ameliorate neuronal damage post spinal cord injury through circ-Wdfy3 delivery and inhibition of ferroptosis. Neurochem Int. 2024;177:105759. doi:10.1016/j.neuint.2024.105759

83. Liu J, Wang Z, Lin A, Zhang N. Exosomes from hypoxic pretreatment ADSCs Ameliorate cardiac damage post-MI via activated circ-Stt3b/miR-15a-5p/GPX4 signaling and decreased ferroptosis. Cardiovasc Toxicol. 2024;24(11):1215–1225. doi:10.1007/s12012-024-09915-9

84. McBride JD, Rodriguez-Menocal L, Guzman W, et al. Proteomic analysis of bone marrow-derived mesenchymal stem cell extracellular vesicles from healthy donors: implications for proliferation, angiogenesis, Wnt signaling, and the basement membrane. Stem Cell Res Ther. 2021;12(1):328. doi:10.1186/s13287-021-02405-7

85. Izquierdo-Altarejos P, Martínez-García M, Atienza-Pérez I, et al. Extracellular vesicles from mesenchymal stem cells reverse neuroinflammation and restore motor coordination in hyperammonemic rats. J Neuroimmune Pharmacol. 2024;19(1):52. doi:10.1007/s11481-024-10153-7

86. Deng M, Hou Y, Liu J, He J, Lan Z, Xiao H. Mesenchymal stem cell-derived exosomes overexpressing SRC-3 protect mice from cerebral ischemia by inhibiting ferroptosis. Brain Res Bull. 2024;211:110948. doi:10.1016/j.brainresbull.2024.110948

87. Chen Y, Hong S, Wang Z, et al. Overexpression of HIF2α enhances the angiogenesis-promoting effect of hUC-MSC-derived extracellular vesicles by stimulating miR-146a. Protein Pept Lett. 2025;32(1):62–74. doi:10.2174/0109298665347753241028072130

88. Łabędź-masłowska A, Vergori L, Kędracka-Krok S, et al. Mesenchymal stem cell-derived extracellular vesicles exert pro-angiogenic and pro-lymphangiogenic effects in ischemic tissues by transferring various microRNAs and proteins including ITGa5 and NRP1. J Nanobiotechnology. 2024;22(1):60. doi:10.1186/s12951-024-02304-y

89. Chen Z, Zhou T, Luo H, et al. HWJMSC-EVs promote cartilage regeneration and repair via the ITGB1/TGF-β/Smad2/3 axis mediated by microfractures. J Nanobiotechnology. 2024;22(1):177. doi:10.1186/s12951-024-02451-2

90. Pizzinat N, Ong-Meang V, Bourgailh-Tortosa F, et al. Extracellular vesicles of MSCs and cardiomyoblasts are vehicles for lipid mediators. Biochimie. 2020;178:69–80. doi:10.1016/j.biochi.2020.07.013

91. Clos-Sansalvador M, Garcia SG, Morón-Font M, et al. N-Glycans in immortalized mesenchymal stromal cell-derived extracellular vesicles are critical for EV-cell interaction and functional activation of endothelial cells. Int J Mol Sci. 2022;23(17):9539. doi:10.3390/ijms23179539

92. Liang X, Miao Y, Tong X, et al. Dental pulp mesenchymal stem cell-derived exosomes inhibit neuroinflammation and microglial pyroptosis in subarachnoid hemorrhage via the miRNA-197-3p/FOXO3 axis. J Nanobiotechnology. 2024;22(1):426. doi:10.1186/s12951-024-02708-w

93. Lou C, Jiang H, Lin Z, et al. MiR-146b-5p enriched bioinspired exosomes derived from fucoidan-directed induction mesenchymal stem cells protect chondrocytes in osteoarthritis by targeting TRAF6. J Nanobiotechnology. 2023;21(1):486. doi:10.1186/s12951-023-02264-9

94. Li N, Zhao L, Geng X, et al. Stimulation by exosomes from hypoxia-preconditioned hair follicle mesenchymal stem cells facilitates mitophagy by inhibiting the PI3K/AKT/mTOR signaling pathway to alleviate ulcerative colitis. Theranostics. 2024;14(11):4278–4296. doi:10.7150/thno.96038

95. Liu Y, Lin S, Xu Z, et al. High-performance hydrogel-encapsulated engineered exosomes for supporting endoplasmic reticulum homeostasis and boosting diabetic bone regeneration. Adv Sci. 2024;11(17):e2309491. doi:10.1002/advs.202309491

96. Wang Q, Guo W, Niu L, et al. 3D-hUMSCs exosomes ameliorate vitiligo by simultaneously potentiating treg cells-mediated immunosuppression and suppressing oxidative stress-induced melanocyte damage. Adv Sci. 2024;11(31):e2404064. doi:10.1002/advs.202404064

97. Xu Y, Wei C, Ma L, et al. 3D mesenchymal stem cell exosome-functionalized hydrogels for corneal wound healing. J Control Release. 2025;380:630–646. doi:10.1016/j.jconrel.2025.02.030

98. Jin Y, Wu O, Chen Q, et al. Hypoxia-preconditioned BMSC-derived exosomes induce mitophagy via the BNIP3-ANAX2 axis to alleviate intervertebral disc degeneration. Adv Sci. 2024;11(34):e2404275. doi:10.1002/advs.202404275

99. Wei S, Cheng RJ, Li S, et al. Microvesicles derived from human umbilical cord mesenchymal stem cells ameliorate renal ischemia-reperfusion injury via delivery of miR-21. J Nanobiotechnology. 2024;22(1):673. doi:10.1186/s12951-024-02922-6

100. Du T, Zhou J, Chen WX, et al. Microvesicles derived from human umbilical cord mesenchymal stem cells ameliorate renal ischemia-reperfusion injury via delivery of miR-21. Cell Cycle. 2020;19(11):1285–1297. doi:10.1080/15384101.2020.1748940

101. Aboulhoda BE, Rashed LA, Ahmed H, et al. Hydrogen sulfide and mesenchymal stem cells-extracted microvesicles attenuate LPS-induced Alzheimer’s disease. J Cell Physiol. 2021;236(8):5994–6010. doi:10.1002/jcp.30283

102. Xin L, Wei C, Tong X, et al. In situ delivery of apoptotic bodies derived from mesenchymal stem cells via a hyaluronic acid hydrogel: a therapy for intrauterine adhesions. Bioact Mater. 2021;12:107–119. doi:10.1016/j.bioactmat.2021.10.025

103. Liu H, Liu S, Qiu X, et al. Donor MSCs release apoptotic bodies to improve myocardial infarction via autophagy regulation in recipient cells. Autophagy. 2020;16(12):2140–2155. doi:10.1080/15548627.2020.1717128

104. Huang J, Zhang J, Xiong J, et al. Stem cell-derived nanovesicles: a novel cell-free therapy for wound healing. Stem Cells Int. 2021;2021:1285087. doi:10.1155/2021/1285087

105. Xing H, Liang C, Xu X, Sun H, Ma X, Jiang Z. Mesenchymal stroma/stem-like cells of GARP knockdown inhibits cell proliferation and invasion of mouse colon cancer cells (MC38) through exosomes. J Cell Mol Med. 2020;24(23):13984–13990. doi:10.1111/jcmm.16008

106. Xu HJ, Liao W, Liu XZ, et al. Down-regulation of exosomal microRNA-224-3p derived from bone marrow-derived mesenchymal stem cells potentiates angiogenesis in traumatic osteonecrosis of the femoral head. FASEB J. 2019;33(7):8055–8068. doi:10.1096/fj.201801618RRR

107. Liu H, Zhang X, Zhang M, et al. Mesenchymal stem cell derived exosomes repair uterine injury by targeting transforming growth factor-β signaling. ACS Nano. 2024;18(4):3509–3519. doi:10.1021/acsnano.3c10884

108. Zhou R, Guo J, Jin Z. Advancing osteoarthritis therapy with GMOCS hydrogel-loaded BMSCs-exos. J Nanobiotechnology. 2024;22(1):493. doi:10.1186/s12951-024-02713-z

109. Lian M, Qiao Z, Qiao S, et al. Nerve growth factor-preconditioned mesenchymal stem cell-derived exosome-functionalized 3d-printed hierarchical porous scaffolds with neuro-promotive properties for enhancing innervated bone regeneration. ACS Nano. 2024;18(10):7504–7520. doi:10.1021/acsnano.3c11890

110. Liu X, Jiang S, Jiang T, et al. Bioenergetic-active exosomes for cartilage regeneration and homeostasis maintenance. Sci Adv. 2024;10(42):eadp7872. doi:10.1126/sciadv.adp7872

111. Li X, Fang S, Wang S, et al. Hypoxia preconditioning of adipose stem cell-derived exosomes loaded in gelatin methacryloyl (GelMA) promote type H angiogenesis and osteoporotic fracture repair. J Nanobiotechnology. 2024;22(1):112. doi:10.1186/s12951-024-02342-6

112. Cui GH, Guo HD, Li H, et al. RVG-modified exosomes derived from mesenchymal stem cells rescue memory deficits by regulating inflammatory responses in a mouse model of Alzheimer’s disease. Immun Ageing. 2019;16:10. doi:10.1186/s12979-019-0150-2

113. Bai X, Qi Z, Zhu M, et al. The effect of lncRNA MIR155HG-modified MSCs and exosome delivery to synergistically attenuate vein graft intimal hyperplasia. Stem Cell Res Ther. 2022;13(1):512. doi:10.1186/s13287-022-03197-0

114. Malekpour K, Hazrati A, Soudi S, Roshangar L, Pourfathollah AA, Ahmadi M. Combinational administration of mesenchymal stem cell-derived exosomes and metformin reduces inflammatory responses in an in vitro model of insulin resistance in HepG2 cells. Heliyon. 2023;9(5):e15489. doi:10.1016/j.heliyon.2023.e15489

115. Lu X, Dai S, Huang B, et al. Exosomes loaded a smart bilayer-hydrogel scaffold with ROS-scavenging and macrophage-reprogramming properties for repairing cartilage defect. Bioact Mater. 2024;38:137–153. doi:10.1016/j.bioactmat.2024.04.017

116. Yan C, Wang X, Wang Q, et al. A novel conductive polypyrrole-chitosan hydrogel containing human endometrial mesenchymal stem cell-derived exosomes facilitated sustained release for cardiac repair. Adv Healthc Mater. 2024;13(10):e2304207. doi:10.1002/adhm.202304207

117. Cao JY, Wang B, Tang TT, et al. Exosomal miR-125b-5p deriving from mesenchymal stem cells promotes tubular repair by suppression of p53 in ischemic acute kidney injury. Theranostics. 2021;11(11):5248–5266. doi:10.7150/thno.54550

118. Guo J, Yang X, Chen J, et al. Exosomal miR-125b-5p derived from adipose-derived mesenchymal stem cells enhance diabetic hindlimb ischemia repair via targeting alkaline ceramidase 2. J Nanobiotechnology. 2023;21(1):189. doi:10.1186/s12951-023-01954-8

119. Chen W, Lin F, Feng X, et al. MSC-derived exosomes attenuate hepatic fibrosis in primary sclerosing cholangitis through inhibition of Th17 differentiation. Asian J Pharm Sci. 2024;19(1):100889. doi:10.1016/j.ajps.2024.100889

120. Chi PL, Cheng CC, Wang MT, et al. Induced pluripotent stem cell-derived exosomes attenuate vascular remodelling in pulmonary arterial hypertension by targeting HIF-1α and Runx2. Cardiovasc Res. 2024;120(2):203–214. doi:10.1093/cvr/cvad185

121. Kaur G, Bae EH, Zhang Y, et al. Biopotency and surrogate assays to validate the immunomodulatory potency of extracellular vesicles derived from mesenchymal stem/stromal cells for the treatment of experimental autoimmune uveitis. J Extracell Vesicles. 2024;13(8):e12497. doi:10.1002/jev2.12497

122. Liu X, Li X, Zhu W, et al. Exosomes from mesenchymal stem cells overexpressing MIF enhance myocardial repair. J Cell Physiol. 2020;235(11):8010–8022. doi:10.1002/jcp.29456

123. Mohammadi MR, Riazifar M, Pone EJ, et al. Isolation and characterization of microvesicles from mesenchymal stem cells. Methods. 2020;177:50–57. doi:10.1016/j.ymeth.2019.10.010

124. Xie L, Mao M, Zhou L, Jiang B. Spheroid mesenchymal stem cells and mesenchymal stem cell-derived microvesicles: two potential therapeutic strategies. Stem Cells Dev. 2016;25(3):203–213. doi:10.1089/scd.2015.0278

125. Bakhshandeh B, Jahanafrooz Z, Allahdadi S, et al. Transcriptomic and in vivo approaches introduced human iPSC-derived microvesicles for skin rejuvenation. Sci Rep. 2023;13(1):9963. doi:10.1038/s41598-023-36162-9

126. Liang Z, Luo Y, Lv Y. Mesenchymal stem cell-derived microvesicles mediate BMP2 gene delivery and enhance bone regeneration. J Mater Chem B. 2020;8(30):6378–6389. doi:10.1039/d0tb00422g

127. Shi P, Gao H, Cheng Z, et al. Static magnetic field-modulated mesenchymal stem cell-derived mitochondria-containing microvesicles for enhanced intervertebral disc degeneration therapy. J Nanobiotechnology. 2024;22(1):457. doi:10.1186/s12951-024-02728-6

128. Du T, Ju G, Zhou J, et al. Microvesicles derived from human umbilical cord mesenchyme promote M2 macrophage polarization and ameliorate renal fibrosis following partial nephrectomy via hepatocyte growth factor. Hum Cell. 2021;34(4):1103–1113. doi:10.1007/s13577-021-00525-z

129. Tang H, Luo H, Zhang Z, Yang D. Mesenchymal stem cell-derived apoptotic bodies: biological functions and therapeutic potential. Cells. 2022;11(23):3879. doi:10.3390/cells11233879

130. Zhu Y, Chen X, Liao Y. Mesenchymal stem cells-derived apoptotic extracellular vesicles (apoevs): mechanism and application in tissue regeneration. Stem Cells. 2023;41(9):837–849. doi:10.1093/stmcls/sxad046

131. Liu J, Qiu X, Lv Y, et al. Apoptotic bodies derived from mesenchymal stem cells promote cutaneous wound healing via regulating the functions of macrophages. Stem Cell Res Ther. 2020;11(1):507. doi:10.1186/s13287-020-02014-w

132. Li X, Jiang Y, Liu X, et al. Mesenchymal stem cell-derived apoptotic bodies alleviate alveolar bone destruction by regulating osteoclast differentiation and function. Int J Oral Sci. 2023;15(1):51. doi:10.1038/s41368-023-00255-y

133. Cheng Y, Zhu Y, Liu Y, et al. Tailored apoptotic vesicles promote bone regeneration by releasing the osteoinductive brake. Int J Oral Sci. 2024;16(1):31. doi:10.1038/s41368-024-00293-0

134. Shi MM, Yang QY, Monsel A, et al. Preclinical efficacy and clinical safety of clinical-grade nebulized allogenic adipose mesenchymal stromal cells-derived extracellular vesicles. J Extracell Vesicles. 2021;10(10):e12134. doi:10.1002/jev2.12134

135. Akhlaghpasand M, Tavanaei R, Hosseinpoor M, et al. Safety and potential effects of intrathecal injection of allogeneic human umbilical cord mesenchymal stem cell-derived exosomes in complete subacute spinal cord injury: a first-in-human, single-arm, open-label, phase I clinical trial. Stem Cell Res Ther. 2024;15(1):264. doi:10.1186/s13287-024-03868-0

136. Chandran NS, Bhupendrabhai MN, Tan TT, et al. A phase 1, open-label study to determine safety and tolerability of the topical application of mesenchymal stem/stromal cell (MSC) exosome ointment to treat psoriasis in healthy volunteers. Cytotherapy. 2025. doi:10.1016/j.jcyt.2025.01.007

137. Park GH, Kwon HH, Seok J, et al. Efficacy of combined treatment with human adipose tissue stem cell-derived exosome-containing solution and microneedling for facial skin aging: a 12-week prospective, randomized, split-face study. J Cosmet Dermatol. 2023;22(12):3418–3426. doi:10.1111/jocd.15872

138. Zamanian MH, Norooznezhad AH, Hosseinkhani Z, et al. Human placental mesenchymal stromal cell-derived small extracellular vesicles as a treatment for severe COVID-19: a double-blind randomized controlled clinical trial. J Extracell Vesicles. 2024;13(7):e12492. doi:10.1002/jev2.12492

139. Lightner AL. Bone marrow mesenchymal stem cell-derived extracellular vesicle infusion for the treatment of respiratory failure from covid-19: a randomized, placebo-controlled dosing clinical trial. Chest Vol. 2023;164(6):1444–1453. doi:10.1016/j.chest.2023.06.024

140. Zhu Y-G, Shi MM, Monsel A, et al. Nebulized exosomes derived from allogenic adipose tissue mesenchymal stromal cells in patients with severe COVID-19: a pilot study. Stem Cell Res Ther. 2022;vol. 13(1):220. doi:10.1186/s13287-022-02900-5

Creative Commons License © 2025 The Author(s). This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, 4.0) License. By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.