Back to Journals » International Journal of Nanomedicine » Volume 20
Diverse-Origin Exosomes Therapeutic Strategies for Diabetic Wound Healing
Authors Wang F, Yao J, Zuo H, Jiao Y, Wu J, Meng Z
Received 26 January 2025
Accepted for publication 20 May 2025
Published 12 June 2025 Volume 2025:20 Pages 7375—7402
DOI https://doi.org/10.2147/IJN.S519379
Checked for plagiarism Yes
Review by Single anonymous peer review
Peer reviewer comments 7
Editor who approved publication: Prof. Dr. RDK Misra
Fengyu Wang,1,2,* Jinling Yao,3,* Huiling Zuo,1,2 Yuhang Jiao,1,2 Junzi Wu,1,2 Zhuoran Meng1,2
1The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China; 2Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China; 3Yunnan Province Dai and Yi Medicine Primary Laboratory, Yunnan University of Chinese Medicine, Kunming, Yunnan, 650500, People’s Republic of China
*These authors contributed equally to this work
Correspondence: Junzi Wu, The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China, Email [email protected] Zhuoran Meng, The Key Laboratory of Microcosmic Syndrome Differentiation, Yunnan University of Chinese Medicine, Kunming, People’s Republic of China, Email [email protected]
Abstract: Diabetic wounds represent a pressing clinical challenge in the medical field. Compared to healthy individuals, patients with diabetes present with various complications, including abnormal blood sugar levels, microcirculation disorders, and impaired cellular function. Moreover, they are at a higher risk for skin damage and have a more difficult healing process. In recent years, exosome-based regenerative medicine has provided new strategies for diabetic wound treatment. The bioactive molecules contained in the exosomes, including functional proteins, bioactive lipids, and regulatory RNAs, allow them to suppress inflammation, enhance cell migration, and promote angiogenesis. As exosomes from different sources have different composition and function, the characteristics of their source must be considered when using them. Unlike traditional single source research, this review describes the mechanism of action of exosomes from different sources in diabetic wound-healing process, including mammalian cell-derived exosomes and plant-derived exosome-like nanoparticles. These findings not only provide a theoretical basis for the selection of exosome sources but also lay a foundation for the development of personalized, multimodal treatment plans.
Keywords: exosomes, diabetic wounds healing, mammalian-derived exosomes, plant-derived exosome-like nanoparticles, drug delivery, nanomedicine
Graphical Abstract:
Introduction
Diabetic wounds are a common complication of diabetes. Poor blood sugar control, microcirculation disorders, oxidative stress, and inflammation often result in delayed wound healing in diabetes. This can progress to severe ulcers or even require amputation, severely affecting the patient’s quality of life.1,2 Statistically, patients with diabetic foot ulcers (DFUs) have a 50% mortality rate over 5 years.3,4 Regardless of the most advanced clinical interventions, approximately 10% of the patients with DFUs undergo amputation, and the mortality rate following such procedures can be as high as 80%.5 Normal wound healing involves numerous factors, including cell proliferation and migration, angiogenesis, and extracellular matrix (ECM) deposition and remodeling.6 However, in patients with diabetes, sustained high blood sugar levels lead to substantial challenges in the typical wound-healing process, interfering with wound repair.7 Diabetic wound fails to enter the normal healing phase and instead falls into a chronic inflammatory state. This phase is characterized by excessive accumulation of M1 macrophages, leading to persistent inflammation. Moreover, there is a significant reduction in fibroblast proliferation, function, and differentiation into myofibroblasts, which further affects collagen synthesis and tissue remodeling.8–10 Multiple methods including wound dressings, hyperbaric oxygen therapy (HBOT), growth factor therapy, and stem cell therapy are currently used in clinical practice.11 For example, hydrogels can load active substances and its composition and structure can be adjusted to provide different functions such as antibacterial, antioxidant, or inflammatory factor expression regulation, effectively alleviating wound infection and inflammation.12–14 However, these methods have substantial clinical limitations. First, traditional dressings have insufficient penetration in severely infected wounds and cannot effectively control deep tissue infection; in addition, frequent dressing changes can exacerbate patient pain.15–17 Second, hyperbaric oxygen therapy requires specialized hyperbaric chambers and professional operation teams, with treatment cycles lasting 20–30 sessions, not only increasing the patient’s financial burden but also potentially causing complications such as middle ear barotrauma and oxygen toxicity.18 Therefore, developing new therapeutic systems with high biocompatibility, low immunogenicity, and targeted delivery capabilities is needed to improve diabetic wound treatment, providing a clear research direction for exosome-based regenerative medicine strategies.
To address the abovementioned issues, exosome-based regenerative medicine can be useful. Exosomes demonstrate unique therapeutic potential, providing multilevel repair mechanisms for diabetic wounds.19 They are extracellular vesicles approximately 30–150 nm in diameter that play a key role in intercellular communication by carrying bioactive molecules such as miRNAs, long non-coding RNAs, and functional proteins.20 Compared to traditional therapies, structural characteristics of exosomes address key barriers in drug delivery. Although their nanoscale size and lipid bilayer structure provide excellent tissue penetration capability, their vesicular nature makes them ideal carriers for biological molecules, capable of simultaneously delivering multiple therapeutic factors that work synergistically, such as vascular endothelial growth factor (VEGF) and transforming growth factor β (TGF-β).21 Furthermore, in terms of inflammation regulation, exosomes can promote the transformation of macrophages from proinflammatory M1 type to anti-inflammatory M2 type, substantially reducing local inflammation.22 Additionally, a single exosome can continuously release active components over an extended period, circumventing the issue of repeated interventions required by methods such as hyperbaric oxygen therapy.23 Finally, due to the absence of nuclear structures, exosomes circumvent the potential tumorigenesis risks associated with stem cell therapy.24 In the complex pathological environment of diabetic wounds, exosomes simultaneously regulate local inflammation, angiogenesis, and extracellular matrix remodeling to address multiple barriers in diabetic wound repair, providing an alternative for shortening existing treatment cycles and reducing recurrence rates.25
Considering the abovementioned properties, researchers aim to optimize the therapeutic efficacy of exosomes in wound healing through precise regulation strategies. The cross-integration of biomaterials science and nano-delivery systems has resulted in the development of exosome-hydrogel composite systems. These intelligent carriers not only extend the retention time of exosomes in the wound microenvironment but also achieve spatiotemporally controlled release of bioactive factors through microenvironment-responsive mechanisms.26–28 Surface chemical modification of exosomes by reshaping their membrane structural characteristics can significantly enhance tissue targeting and molecular loading, providing a structural foundation for precision treatment.29–31 Moreover, the composition and function of exosomes vary depending on their source. Based on their origin, exosomes can be classified into two major categories: mammalian-derived exosomes (MDEs) and plant-derived exosome-like nanoparticles (PELNs)32 (Figure 1). The former has a lipid bilayer constructed with cholesterol and glycosphingolipids as core scaffold, providing it with unique structural rigidity and biological stability,33,34 whereas the latter presents a differentiated lipid distribution pattern, characterized by enhanced membrane fluidity and biological solubility.35 MDEs can be isolated from various cell types, from mesenchymal stem cells to immune cells as well as from biological fluids such as blood and amniotic fluid, each having distinctive immunomodulatory capabilities and tissue repair functions.36 However, PELNs, extracted from various plant matrices, have garnered attention due to their rich natural components and relatively low immunogenicity and are abundant in natural active ingredients such as polyphenols and flavonoids, demonstrating unique advantages in oxidative stress regulation and inflammatory balance.37
![]() |
Figure 1 Derived from cells, body fluids, and plant exosomes. |
Unlike previous studies focusing on a single exosome source, this review comprehensively compares the mechanisms of action between mammalian and plant-derived exosomes in diabetic wound healing. Through a detailed discussion of the functional characteristics of exosomes from different sources, this review reveals the critical impact of exosome origin on their therapeutic effects. This innovative research on multisource exosomes provides a theoretical basis for developing individualized treatment plans, while exploring challenges and opportunities in exosome scale-up production, quality control, and clinical translation, establishing a foundation for innovative therapeutic strategies for diabetic wound healing.
Mammalian-Derived Exosomes
MDEs have become the focus of research in the field of diabetic wound healing due to their excellent biocompatibility, precise tissue targeting, and ability to carry various bioactives. MDEs can be classified as cell-derived (eg, MSCs, neural stem cells) and body fluid-derived (eg, plasma, amniotic fluid), based on their source. MDEs play a vital role in modulating inflammatory responses, fostering angiogenesis, augmenting cell proliferation and migration, and orchestrating ECM remodeling. Thus, a novel therapeutic strategy using exosomes can offer hope for treating diabetic wounds.
Cell-Derived Exosomes
MSC-Exos
MSCs are a subset of adult nonhematopoietic stem cells of mesodermal origin, which can be isolated from various tissues, such as bone marrow, umbilical cord blood and tissues, placental tissues, and adipose tissue. MSCs have the ability to self-renew and differentiate into various specialized cell types.38 MSC-Exos exhibit effects similar to MSCs in inducing cell proliferation and differentiation as well as angiogenesis in vitro, but their therapeutic effect on the injury site is much better than that of MSCs.39 MSCs-Exos coordinate various stages of wound healing by regulating inflammatory responses, promoting cell proliferation, enhancing cell migration ability, and stimulating angiogenesis.40 This section describes the mechanisms of action of the three main MSC-derived exosomes in diabetic wound healing.
BMSC-Exos
Bone marrow MSCs (BMSCs) are the earliest isolated MSCs and have been extensively researched and applied in stem cell therapy.41 BMSC-Exos have the advantages of a low infection rate by pathogenic microorganisms, stable biological performance, low immune rejection rate after transplantation, and high number of generations.42 BMSC-Exos play an anti-inflammatory, angiogenesis-promoting, and cell proliferation-promoting role in wound healing. Moreover, in the context of wound healing, BMSC-Exos exhibit anti-inflammatory properties, facilitate angiogenesis, and promote cellular proliferation.
First, during the inflammatory phase of wound healing, BMSC-Exos accelerate the polarization of M2 macrophages, thereby shortening the duration of inflammation.43 Luo et al observed that intramuscular injection of BMSC-Exos after muscle contusion reduced inflammation. This was attributed to the promotion of M2 macrophage polarization and anti-inflammatory factor expression as well as a reduction in inflammatory cytokine production in the inflammatory microenvironment.44 Additionally, BMSC-Exos significantly decreased M1 macrophage polarization and increased M2 macrophage polarization in a diabetic mouse air pocket model and a diabetic rat model of whole skin trauma. In contrast, melatonin-stimulated BMSC-Exos (MT-Exos) had a stronger effect.45 In a study, multifunctional BMSC-Exos-loaded carboxyethyl chitosan (CEC)-dialdehyde carboxymethylcellulose (DCMC) hydrogel (MSC-Exos@CEC-DCMC HG) promoted chronic diabetic wounds by modulating wound inflammation through the promotion of macrophage conversion from proinflammatory M1-type to reparative M2-type and significantly inhibiting bacterial growth to enhance antimicrobial effect repair.46
Moreover, stimulation of angiogenesis, particularly through the modulation of vascular endothelial growth factor A (VEGFA), is one of the key strategies for promoting neovascularization and accelerating wound repair in diabetic wound therapy.47 Han et al found that BMSC-Exos carry Kruppel-like factor 3 antisense RNA1, which promotes the proliferation of chondrocytes and cardiomyocytes and facilitates VEGFA signaling and cutaneous wound healing in patients with diabetes by downregulating miR-383.48 Similarly, Zhang et al found that endothelial-specific miRNA-126 (miR-126) derived from BMSC-Exos activated the PI3K/AKT signaling pathway by targeting phosphatidylinositol 3-kinase regulatory subunit 2 through in vitro experiments, upregulated the expression of angiogenesis-associated VEGF and Ang-1 genes, and promoted angiogenesis in HUVECs. In vivo experiments confirmed that the application of Exo-miR-126 considerably enhanced angiogenesis at the wound site and promoted wound healing.49 Tang et al found that circ-Snhg11 of BMSC-Exos promoted SLC7A11/GPX4-mediated anti-iron apoptotic signaling through sponge miR-144-3p and promoted angiogenesis.50 In another study, atorvastatin (ATV)-pretreated BMSC-Exos (ATV-Exos) upregulated the AKT/eNOS signaling pathway and enhanced the angiogenesis-promoting function of BMSC-Exos, which accelerated diabetic wound repair and regeneration.51 Further studies revealed that IFN-γ pretreated BMSC-Exos miR-126-3p promoted angiogenesis through the SPRED1/Ras/ERK axis, showing higher therapeutic efficacy than NExos in diabetic wound healing.52
The pathogenesis of diabetic wounds involves abnormalities in various cellular biological processes, such as epidermal-specific macrophage/autophagy damage, cell proliferation, and apoptosis.53 Notably, culturing BMSCs in a hypoxic environment enhanced the regenerative and cytoprotective effects of BMSCs.54 Shi et al conducted an innovative study revealing that exosomes from hypoxic bone marrow mesenchymal stem cells (hyBMSC-Exos) significantly enhance diabetic wound healing through a specific epidermal autophagy mechanism. Their research demonstrated that hyBMSC-Exos can deliver miR-4645-5p, which targets and inhibits MAPKAPK2 expression, to keratinocytes. This inhibition prevents MAPKAPK2-mediated activation of the AKT-mTORC1 signaling pathway, a known suppressor of cellular autophagy. The resulting restoration of keratinocyte autophagy promotes cell proliferation and migration, accelerating the re-epithelialization process essential for wound closure (Figure 2).55 By contrast, Shen et al found that miR-93-3p from BMSC-Exos restored cell function and inhibited apoptosis in epithelial HaCaT cells by inactivating apoptotic peptidase-activating factor 1—a finding that may help establish a new therapeutic strategy for skin wound healing.56 Furthermore, BMSC-Exos reduced the expression of transforming growth factor beta 1 (TGF-β1) and upregulated the expression of TGF-β3 and Smad7 in the TGF-β/Smad signaling pathway, effectively promoting the proliferation of human keratinocytes (HaCaT) and human dermal fibroblasts and facilitating skin wound healing.57
![]() |
Figure 2 Schematic of the therapeutic effect of hyBMSC-Exos on diabetic wounds. The complete treatment process is as follows. (1) Recruitment of hyBMSC-Exos at the wound site following injection. (2) Molecular mechanism showing hyBMSC-Exos delivering miR-4645-5p to epithelial cells, where it inhibits MAPKAPK2, preventing phosphorylation of AKT and subsequent activation of mTORC1, ultimately promoting autophagy. (3) The resulting increased keratinocyte proliferation and migration leads to enhanced re-epithelialization and wound closure. Reproduced with permission from Shi Y, Wang S, Liu D et al. Exosomal miR-4645-5p from hypoxic bone marrow mesenchymal stem cells facilitates diabetic wound healing by restoring keratinocyte autophagy. Burns Trauma. 2024;12. Copyright (2024) Oxford University Press.55 |
These findings provide a theoretical basis for the practical application of BMSC-Exos in the clinic. However, the speed of cultivating and amplifying germinated stem cells is slow, making it impossible to obtain a large number of BMSCs in a short time period. Second, red blood cell contamination is a risk during separation. Additionally, eliminating the invasion of MSCs from the bone marrow is a challenge.58 These issues may limit the clinical application of BMSCs-Exos. Therefore, further research and innovation is required to overcome these challenges.
ADSC-Exos
Adipose MSC-derived exosomes (ADSC-Exos) are highly stable and easy to store. Compared to MSCs derived from the bone marrow, ADSCs have many advantages.59 First, the amount of derived from the adipose tissue is approximately 500 times more than that of those derived from the bone marrow, making the sources for ADSCs abundant. ADSCs can also be obtained with high purification efficiency.60 Next, the extraction of ADSCs is relatively painless and less traumatic, and these can be obtained through liposuction surgery.42 Studies have shown that ADSC-Exos has a better effect than BMSC-Exos, ADSC-Exos are a more ideal source of cells. In wound repair, ADSCS-Exos regulates immune response and inflammation, promotes blood vessel production, accelerates the proliferation of skin cells and re-epithelization, and regulates collagen to reshape, thereby suppressing scar hyperplasia.
Studies have found that ADSC-Exos regulate macrophages to inhibit inflammation.61 Specifically, ADSC-Exos can directly interact with macrophages, resulting in the suppression of the macrophages’ nuclear factor κB (NF-κB) activity and specific inflammatory genes, which eventually leads to the reduction of inflammation in the mediators of macrophages. Studies have found that ADSC-Exos reduced the inflammatory response in mice RAW264.7 cells, reduced the level of impression of macrophages and inflammatory cytokines, and increased the secretion of anti-inflammatory cytokines.62 In macrophages, MiR-155 generated by ADSC-Exos combined its target SOCS1 and adjusted the JAK/STAT signal to promote M1 macrophage polarization and reduce chronic inflammation.63 Li et al found that ADSC-Exos overexpressing Nrf2 effectively reduced the levels of reactive oxygen species and inflammatory factors, promoted foot wound healing, and reduced the ulcer area in diabetic rats.64 By contrast, refractory diabetic wounds create a persistent inflammatory and hypoxic environment. Xiao et al found that under hypoxic conditions, HypADSCs-Exos exhibited higher survival and proliferation than that under normal oxygen conditions. The expression profiles of miRNAs in these exosomes change, and they may promote wound healing by regulating cell metabolism, differentiation, and TGF-β functions. Additionally, under hypoxic conditions, the exosomes derived from fattened cells promoted high-quality healing of diabetic wounds by activating the PI3K/AKT pathway.65
ADSC-Exos have proven that miRNA-125A can be transferred to endothelial cells and promote vascular production by inhibiting the expression of Delta-like protein 4 (DLL4).66 Zhang et al developed a HaCaT cell model and a mouse wound healing model to study the effect of ADSC-Exos on wound healing. Results showed that ADSC-Exos promoted the proliferation and migration of HaCaT cells by regulating the activation of the AKT/HIF-1α signaling pathway, thereby promoting wound healing.67 Wang et al showed that ADSC-Exos regulated the ratio of type III collagen: type I collagen, TGF-β3:TGF-β1, and MMP3:TIMP1 as well as regulated the differentiation of fibroblasts to reduce the formation of scars and promote ECM reconstruction in skin wound repair.68 Another study found that miRNA-146a-modified ADSC-Exos, by upregulating serine protease inhibitor family H member 1 and phosphorylating ERK, promoted fibroblast migration and proliferation as well as neovascularization to promote wound healing.69
ADSC-Exos help reduce scar formation. In specific cases, miR-192-5p present in ADSC-Exos regulates the Smad pathway in proliferative scar fibrosis by targeting IL-17RA and inhibits fibroblast proliferation, excessive collagen synthesis, and ECM deposition, thereby impeding scar formation.70 ADSC-Exos impeded the differentiation of fibroblasts to myofibroblasts by activating the ERK/MAPK pathway. This process led to an elevated COL-3/COL-1 ratio, TGF-β3/TGF-β1 ratio, and MMP3/TIMP1 ratio, promoting the reconstruction of the ECM during skin wound healing and reducing scar formation.71 Although ADSC-Exos accelerate wound healing, they may affect melanoma migration and invasion through the fatty acid oxidation pathway.42 Therefore, future studies must focus on the in-depth understanding of their mechanism of action as well as the safety assessment for clinical applications.
Umbilical Cord MSCs-Exos
Umbilical cord MSCs (UCMSCs) are present in neonatal umbilical cord tissue and are a versatile and ideal source of stem cells. They synthesize and secrete various trophic factors and cytokines, promoting the proliferation and function of other cell types.72 Compared to different types of stem cells, UCMSC-Exos have low immunogenicity and high stability. Moreover, as a drug carrier, it is easy to control their route of administration and dosage. UCMSC-Exos are safe and reliable for long-term application without liver or kidney toxicity. In addition, they have a high differentiation rate when applied in vivo and are not potentially tumorigenic. UCMSC-Exos are now widely used in regenerative medicine and for treating various diseases.73
UCMSC-Exos promote angiogenesis during wound healing through multiple mechanisms. Similar to BMSC-derived exosomes, UCMSC-Exos upregulated VEGF expression and induced the upregulation of VEGF and HIF-1a expression, thereby promoting angiogenesis in a rat model. The efficacy of HIF-1a in enhancing UCMSC-Exos-induced VEGF expression and promoting angiogenesis has been demonstrated by specific RNA inhibitors or siRNA.74 Zhang et al identified miR-21 in UCMSC-Exos as a potential intercellular messenger, activating the NOTCH1/DLL4 pathway and promoting the proliferation, migration, and angiogenesis of endothelial progenitor cells (EPCs).
UCMSC-Exos stimulates cell proliferation and collagen synthesis through multiple mechanisms. Kim et al demonstrated the ability of UCMSC-Exos to promote human dermal fibroblast proliferation and collagen synthesis.75 Application of UCMSC-Exos to the treatment of human skin trauma led to increased expression of COL-I and elastin. Furthermore, Teng et al showed that HucMSCs-exo grafts increased diabetic wound healing. In vitro, HucMSCs-exo promoted the proliferation of human umbilical vein endothelial cells (HUVECs) and NIH-3T3 cells. In vivo, HucMSCs-exo reduced wound area and inflammatory infiltration and increased collagen fibers. Furthermore, wound tissues in the HucMSCs-exo group had higher CD206, CD31, and VEGF expression than the control group at 14 days and lower TNF-α levels. The results suggest that HucMSCs-exo promotes diabetic wound repair by inducing anti-inflammatory macrophages and promoting angiogenesis and collagen deposition.43 Cytokine profiling showed that HucMSCs-Exos contained high doses of IL-6, IL-8, and other cytokines. HucMSCs-exos could stimulate cell proliferation and resist hydrogen peroxide-induced apoptosis, promote cell proliferation, and protect against oxidative stress-induced apoptosis by activating ERK1/2 and p38, thereby promoting wound healing.76
The role of UCMSC-Exos in inhibiting scar formation was also confirmed. Myofibroblast aggregation is a key factor in scar formation. Based on this, Fang et al used high-throughput RNA sequencing and functional to verify that the presence of certain miRNAs (eg, miR-21, −23a, −125b, and −145) in UCMSC-Exos that blocked the TGF-β2/SMAD2 pathway. In vitro and in vivo, these effectively impeded myofibroblast aggregation and reduced scar formation. During the organizational restructuring phase, the HucMSC-Exos-derived Wnt4 factor promotes nuclear translocation and activity of p-catenin. This enhances skin cell proliferation and migration.77 Additional studies have found that HucMSC-Exos increased the in vivo expression of CK19, PCNA, and collagen I. During the in vitro heat stress response, HucMSC-Exos promoted skin cell proliferation and inhibited apoptosis, thereby accelerating wound healing.78 In the treatment of type 2 diabetes, HucMSC-Exos showed remarkable potential. They attenuated the damage of insulin β-cells in rats, reversed insulin resistance in peripheral tissues, and improved glucose uptake and utilization by liver and muscle. This reduced blood glucose levels, providing a new strategy for treating diabetes.79 Studies have reported that HUCMSC-derived exosomes bind to gelatin methyl acrylate. Inhibition of vein graft restenosis was done by enhancing endothelial function.80 This provides new ideas for vascular repair and diabetic wound therapy.
Macrophage-Derived Exosomes
Macrophages are natural immune cells. They create an optimal microenvironment for reducing inflammation through paracrine mechanisms.81 Macrophage-derived Exos act as important messengers. They pay an important role in communication with neighboring cells by regulating the levels of cytokines and miRNA, thereby alleviating the inflammatory response of the recipient cells.82 In recent years, macrophage-derived exosomes (M-Exos) have exhibited a beneficial role in immunomodulation, cancer therapy, infection defense, and tissue repair.83 Dysfunction of macrophages constitutes a major obstacle in diabetic wound healing. Macrophages tend to favor M1 polarization, leading to difficulties in wound closure, poor neovascularization, and reduced collagen deposition. In addition, it exacerbates the inflammatory response and inhibits wound healing.84
Studies have shown that M-Exos play important immunomodulatory roles in diabetic wound repair. Exosomes can effectively stimulate the polarization of macrophages, especially promoting the transition from the M1 phenotype to M2 phenotype, which is crucial for wound healing.85 First, M-Exos reduce inflammation by decreasing the secretion of proinflammatory factors, such as TNF-α and IL-6. M-Exos promote wound re-epithelialization and neovascularization, thus accelerating diabetic wound healing.86 M-Exos can promote the shift of macrophages from proinflammatory M1 type to anti-inflammatory M2 type in vivo, which effectively ameliorates the inflammatory response of receptor cells and stimulates angiogenesis.87 A related report suggested that macrophage-derived exosomes attenuated thermal hyperalgesia in a mouse model of inflammatory pain, suggesting that they play a role in inflammatory dysregulation.88 In addition, M-Exos adjusted the level of cytokines and miRNAs. Another study showed that M2-Exo inhibited the expression of phosphatase and tensin homolog in HUVECs by transferring miR-21 and activating the AKT/mTOR pathway. By injecting M2-Exo into mouse skin wounds, it was demonstrated that M2-Exo acted as a promoter of angiogenesis and regeneration in vivo and accelerated skin healing.89
Additional reports indicate that macrophage-derived exosomes loaded with curcumin (Exos-cur) have good stabilizing, anti-inflammatory, antioxidant, and other biological activities. In in vitro experiments, Exos-cur promoted the proliferation, migration, and angiogenesis of HUVECs. It reduced the reactive oxygen species produced by high glucose-induced HUVECs and inhibited oxidative stress and inflammation. In vivo, Exos-cur activated the Nrf2/ARE pathway, promoting angiogenesis and accelerating wound healing, inhibiting the inflammatory response in diabetic rats.90 Zeng et al investigated an MEs@PMN hydrogel with good biocompatibility and considerable photothermal effect. MEs released from MEs@PMN hydrogels significantly promoted the shift in macrophage phenotype from a proinflammatory M1 phenotype to an anti-inflammatory M2 phenotype, inhibited inflammation, and promoted angiogenesis to accelerate diabetic wound healing.91 These studies suggest, M-Exos not only participate in disease progression but also serve as a new target for therapy, providing new research directions for disease diagnosis and treatment.
Endothelial Cells-Derived Exosomes
Current research has demonstrated that endothelial cells secrete exosomes and capture exosomes from various cell types.92 In a high glucose environment, endothelial cells release increased amounts of exosomes. These exosomes promote the repair of endothelial cells and enhance their migration, thus accelerating wound healing.93 Additionally, EPCs promote endothelial cell regeneration through a paracrine mechanism rather than through differentiation toward mature endothelial cells.94 The high-glycemic environment and inflammatory response in diabetic wounds can lead to a decrease in the number of EPCs. Proliferation, adhesion, migration, and other abilities to assist in vascular synthesis are diminished, inhibiting the function of blood vessels. The vessel wall’s advanced glycosylation end products (AGEs) reduce EPC activity. This hinders wound angiogenesis and reduces the rate of wound healing.95 Exosomes are key components of paracrine secretion and represents an important intercellular communication through the delivery of functional RNAs and proteins.96
EPC-Exos significantly promoted neoangiogenesis and accelerated skin wound healing in diabetic rats. One study found that Erk1/2 signaling plays a key role in the proangiogenic effects of EPC-Exos on endothelial cells, and inhibition of this signaling significantly blocked the proangiogenic effects of exosomes. Subsequently, Li et al97 found that EPC-derived exosomes increased the expression of angiogenesis-related molecules, including FGF-1, VEGFA, VEGFR-2, ANG-1, and several other factors, by stimulating endothelial cells. These factors eventually promoted wound healing by regulating the proliferation, migration, and tube formation function of vascular endothelial cells. Xu et al98 found that miRNA-221-3p in EPCs-Exos increased the protein expression levels of angiogenesis-associated factors VEGF and CD31 and the cell proliferation marker Ki67. miRNA-221-3p in EPCs-Exos may be involved in diabetic complications, cell cycle, and p53-mediated AGE-RAGE signaling pathway to promote skin wound healing in diabetic mice.
HUVEC is a type of endothelial cell. It is widely used to study mechanisms of tumor angiogenesis and cardiovascular diseases.99 HUVEC-Exos have shown promising results in promoting wound healing by modulating the immune response, reducing inflammation, promoting angiogenesis, accelerating skin proliferation and epithelialization, and modulating collagen remodeling to inhibit scar proliferation.100 miRNA-containing exosomes secreted by human coronary artery endothelial cells and a human microvascular endothelial cell line (HMEC-1) can be internalized by the recipient EC, thereby regulating gene expression and stimulating EC proliferation and migration.101 When inflammatory cells are restricted, there is an inadequate supply of oxygen and nutrients within the wound. Inadequate angiogenesis and impaired wound healing can occur.64 A study found that the GelMA/PEGDA microneedle patch of HUVECs-Exos loaded with tazarotene accelerated blood circulation, providing the necessary nutrients and oxygen for the repair of damaged tissues.100 This treatment has promising clinical applications. It provides a new direction for the clinical treatment of diabetic wounds.
Fibroblasts-Derived Exosomes
Fibroblasts are often defined as structural cells that are exclusively responsible for the deposition and remodeling of the ECM; they are essential for maintaining tissue integrity. Moreover, fibroblasts are closer to the skin than MSCs, and they can be obtained from the skin using less invasive techniques.102 Fibroblasts contribute to tissue homeostasis, wound healing, defense against pathogens and injury, and metabolism; however, they may become dysregulated during disease, leading to fibrosis, chronic inflammation, poor wound healing, and cancer. Research has found that fibroblasts play key roles in various chronic inflammatory diseases, such as rheumatoid arthritis, Lyme arthritis, scleroderma, and atopic dermatitis.103,104
Although research on fibroblast-derived exosomes in diabetic wound healing is relatively limited, it has been shown that in contrast to cell-based therapies, exosomes exhibit easier endocytosis and higher loading efficiency due to their unique structure.105 These exosomes are enriched with various growth factors and cytokines, such as TGF-β and matrix metalloproteinase. They can promote cell proliferation and migration and regulate the expression of type I and type III collagen and fibronectin. This promotes angiogenesis and accelerates wound healing.106 Fibroblast exosomes promote wound healing and improve the quality of wound healing by regulating the remodeling of the ECM.107 Investigators performed histopathologic of skin wound biopsies from a rat model of total skin ulceration and found that fibroblast-Exos can be used as a viable cell-free treatment to effectively treat skin wounds with a significant boost in repair.108 Dermal fibroblasts (DFs) produce abundant collagen and cytokines that are essential for skin regeneration and damage repair.109 Han et al discovered that DF-Exo significantly improved the functional properties of type 2 diabetes rat skin cells, leading to accelerated healing of diabetic skin wounds through the activation of the Akt/β-catenin pathway. By studying the role of miR-125b in exogenous body delivery in aging fibroblast FMT and migration, the treatment potential of miR-125b in age-related defect wound healing was tested. The findings revealed that young fibroblasts stimulated exogenous old-aged fibroblasts to increase miR-125b in the aging fiber cells, thereby promoting migration and FMT, which eventually accelerated wound healing in elderly mice.110 Although high-quality evidence on the specific mechanism of fibroblast-Exo in wound healing is lacking, these results provide new perspectives and therapeutic strategies for the use of fibroblast-Exo in cutaneous wound repair.
Other Cell-Derived Exosomes
Numerous studies have shown during wound healing, stem cell-derived exosomes can modulate inflammation and promote trabecular angiogenesis, migration and proliferation, collagen formation, and ECM remodeling. They can be encapsulated in biocompatible scaffolds to exert biological effects.111,112 On this basis, periodontal MSC-derived exosomes show potential applications in periodontal disease treatment, bone regeneration, and wound healing.113,114 These exosomes inhibited the inflammatory response of M1-type macrophages and reduced lipid accumulation in a hyperlipidemic microenvironment. This provides a new therapeutic strategy for the treatment of periodontitis associated with hyperlipidemia.115 Kuang et al demonstrated that keratinocyte-derived exosomal MALAT1 plays a crucial role in diabetic wound healing through a specific molecular pathway (Figure 3). This mechanism is based on MALAT1’s ability to act as a competitive RNA for miR-1914-3p, thereby preventing MFGE8 inhibition in macrophages. The increased MFGE8 expression drives three key beneficial effects in macrophages-enhanced phagocytosis, polarization toward the M2 phenotype, and reduced apoptosis. By modulating the TGFB1/SMAD3 signaling axis, this pathway effectively promotes tissue regeneration and wound closure in diabetic conditions.116 Subsequently, Zhou et al reported that keratinocyte-derived exosomes carry miRNAs that directly determine the number and function of macrophages within the granulation tissue. This is critical for functional wound healing. These exosomes regulate intercellular signaling and promote proliferation and migration of keratinized cells, thereby accelerating wound healing.117 In summary, exosomes derived from different sources have shown significant therapeutic potential in wound healing and tissue regeneration (Table 1) and hold promise in regenerative medicine and precision therapy.
![]() |
Table 1 Targets and Functions of Different Exosome Sources in Diabetic Wound Healing |
![]() |
Figure 3 Schematic showing how human keratinocyte-derived exosomal MALAT1 promotes diabetic wound healing by suppressing miR-1914-3p to activate MFGE8, resulting in enhanced macrophage function and inhibition of the TGFB1/SMAD3 pathway. Reproduced with permission from Kuang L, Zhang C, Li B et al. Human keratinocyte-derived exosomal MALAT1 promotes diabetic wound healing by upregulating MFGE8 via microRNA-1914-3p. International Journal of Nanomedicine. 2023;18:949–970. Copyright (2023) Taylor & Francis Group.116 |
Body Fluid-Derived Exosomes
Low yield of exosomes in cell culture supernatants is a challenge, limiting the potential application of exosomes in the field of drug delivery.133 To address this challenge, researchers are actively exploring more efficient ways to produce exosomes in large quantities. Exosomes from bodily fluids, such as the serum, saliva, breast milk, and amniotic fluid, are not only abundant but they also carry mRNAs that can be analyzed. Moreover, genetic material can be isolated and analyzed by minimally invasive techniques, providing new ways to gain insight into the host transcriptome.115 This property gives exosomes derived from of bodily fluids the potential to be used as a diagnostic tool.
Blood-Derived Exosomes
Studies have shown that blood-derived exosomes are not only potential biomarkers but also provide new wound healing treatment avenues. Xiong et al also emphasized that miR-15a-3p-enriched in Dia-Exos could downregulate the NOX5/reactive oxygen species (ROS) signaling pathway, thus impairing angiogenesis and diabetic wound healing.134 Human umbilical cord blood (UCB) is an excellent source of transplantable stem cells for wound repair. It has several distinct advantages, such as no risk to the donor, easy accessibility, and low incidence of graft-versus-host disease.135 However, the direct use of stem cells for therapeutic purposes is limited by many risk factors, such as tumor formation, thrombosis, and unwanted immune reactions.136 Some studies have reported that local injection of exosomes secreted by human UCB-derived stem cells can promote the proliferation and migration of skin fibroblasts and angiogenesis in animal models of diabetes or burns and promote wound healing.137 Another study found that miR-21-3p enriched with UCB-Exos, a miRNA that is a key mediator of UCB-Exos-induced regulation of functional properties of fibroblasts and endothelial cells. Furthermore, inhibition of phosphatase and tensin homolog as well as germination homolog 1 promotes angiogenesis and fibroblast function, thus enhancing skin wound healing.138 Blood-derived exosomes act by carrying and releasing growth factors, such as platelet-derived growth factor and TGF-β, to promote cell migration and proliferation and accelerate wound healing.139 For example, serum-derived exosomes promote migration of NIH-3T3 cells and tube formation of HUVECs to enhance angiogenesis and ECM generation to accelerate diabetic wound healing.140
Additionally, platelet-rich plasma (PRP) has been widely used for tissue repair and regeneration. PRP promotes wound healing by releasing antimicrobial peptides, growth factors, and micro-RNAs.141,142 PRP has anti-inflammatory and proproliferative effects for the treatment of DFUs by regulating miR-21 and PDCD4, inhibiting NF-κB activity, and providing new targets for the treatment of refractory wounds.143 Sphingosine-1-phosphate (S1P) is a key regulator of vascular homeostasis and angiogenesis. Chen et al isolated exosomes from PRP by ultracentrifugation and used diabetic mouse models to evaluate the effects of PRP-Exos on wound healing. They confirmed that PRP-Exos significantly promote angiogenesis and accelerate healing of diabetic wounds by activating the S1PR1/protein kinase B/FN1 signaling.144 Additionally, Rui et al explored how the diabetic environment affects PRP-Exos and their potential impact on neutrophil extracellular traps. They found that the miRNA-26b-5p contained in PRP-Exos promotes wound healing by reducing neutrophil infiltration by targeting MMP-8.145 Notably, different PRP activation methods affect the quantity, quality, and growth factor content of exosomes, with thrombin and calcium gluconate mixed activation producing exosomes with optimal biological function in promoting HUVEC cell proliferation, migration, and angiogenesis.146
Human Urine-Derived Exosomes
As isolation of MSCs from adult tissues is invasive and often limited by the source, finding a new source of autologous stem cells that can be easily obtained through noninvasive methods is imperative. Urine can replace plasma as a potential source of disease biomarkers. Stem cell-like cells were extracted from adult urine, showing self-renew ability. They have multifaceted differentiation ability and superior proliferation capacity.147,148 After serial propagation, hUSCs retained a normal karyotype with advanced colony-forming ability.149 Urinary exosomes contain various biologically active molecules that reflect the physiological and pathological state of the body. They promoted cell proliferation and migration and enhanced wound healing.150
In recent years, several studies have applied USCs to repair bone, cartilage defects, and wounds with favorable therapeutic results.151 USCs differentiated into osteoblasts, adipocytes, and chondrocytes. Urine-derived stem cell exosomes (USC-Exos) exhibit a cup- or sphere-shaped morphology, with a mean diameter of 51.57 ± 2.93 nm and are positive for CD63 and TSG101. USC-Exos enhance the angiogenic activity of endothelial cells. Specifically, a proangiogenic protein known as malignant brain tumor delete 1 is highly expressed in USC-Exos. Chen et al showed that USCs-Exos promoted angiogenesis in diabetic mice by upregulating the expression of the malignant brain tumor delete 1 gene.152 Additional studies have found that compared to conventional USC-Exos, exosomes from CD133+ human urine-derived stem cells (CD133+ USC-Exos) promoted the chondrogenic differentiation of BMSCs more effectively. This in turn promoted bone–tendon interface healing in rotator cuff injury repair.153 In addition, urine output and urinary microalbumin excretion were reduced in rats by intravenous administration of USCs-Exo. It was found that USCs-Exo may prevent diabetic kidney injury by inhibiting apoptosis of podocytes, growth factors in USCs-Exo, angiopoietins, and bone morphogenetic protein-7, among other potential factors, promoting vascular regeneration and cell survival to prevent diabetic kidney injury.154 These findings could be a promising treatment option for diabetic wound healing.
Although the origin of USCs remains controversial, USCs have more homology with the urinary system. Combining the advantages of USCs and exosomes, USC-Exos have less immune rejection, better differentiation, and a more stable and adequate supply.155 Therefore, exosomes in urine as potential biomarkers would be a promising therapeutic approach in regenerative medicine.
Saliva-Derived Exos
Saliva contains large amounts of proteins and growth factors and is an important source of tissue regeneration factors,156 especially during the healing of oral trauma. Salivary exosomes (SEs) are rich in growth factors and antimicrobial peptides; they can accelerate local tissue repair and have anti-inflammatory effects, thus promoting wound healing.157 Mi et al showed that saliva-derived Exos induced the proliferation, migration, and angiogenesis of HUVECs in an in vitro experiment. In in vivo experiments, saliva-derived Exos showed a strong ability to promote skin wound healing.158 Ubiquitin-binding enzyme E2O is one of the major mRNAs in SEs. Researchers have suggested that ubiquitin-binding enzyme E2O mainly acts by decreasing the level of SMAD homolog 6, which activates bone morphogenetic protein 2. The activation of bone morphogenetic protein 2 further induces angiogenesis. Although saliva-derived Exos represents a novel strategy to promote wound repair by promoting angiogenesis, saliva as an effective medium that promotes local health has not been widely recognized in the field of medicine and biomaterials. In-depth studies are needed to achieve clinical application.159
Notably, although exosomes from the saliva have been less studied, saliva-derived exosomes are thought to have greater versatility in the diagnosis and treatment of disease compared to exosomes from other body fluids. By contrast, SEs are being studied as an alternative to whole saliva. This is because whole saliva contains contaminating elements and higher levels of amylase. In addition to organ-specific pathologies, SEs have applications in systemic diseases, including autoimmune diseases, neurodegenerative diseases, and malignant tumors.157 Saliva has been reported to carry DNA, RNA, and metabolites present in both blood and saliva. SEs collection is easy and noninvasive, which improves patient compliance. Thus SEs can be easily obtained from patients. Furthermore, saliva does not clot and SEs are stable in biological fluids, such as blood and gastric juices. Thus, drug delivery using SEs shows great therapeutic potential in numerous innovative applications.
Milk-Derived Exosomes
Breast milk exosomes are rich in miRNAs, proteins, lipids, and other biomolecules. These components can influence numerous biological processes. They are key regulators of gene expression networks in normal physiological and disease contexts. Moreover, these components show potential promise as disease biomarkers. Breast milk exosomes are not only involved in the growth and development of newborns but also enhance their immune function.160 By regulating the intestinal microbiota and promoting the repair of intestinal epithelial cells, they helps newborns resist infection and promote wound healing.161 However, access to breast milk can be limited, and the quantity of breast milk can vary. Moreover, there may be ethical and privacy issues involved in the extraction and study of exosomes derived from breast milk. These issues affect the availability of exosomes. Therefore, researchers sought alternative sources, turning their attention to milk exosomes as a potential alternative.
In 1973, Plantz et al discovered milk exosomes, which are now considered promising candidates for the development of new drug delivery systems.162 First, the relatively high yield of exosomes isolated from milk is the greatest advantage that cannot be matched by other exosome sources.163 Additionally, milk-derived exosomes are highly resistant to the harsh gastrointestinal environment and can be used as an oral drug delivery system.164 Yan et al developed milk-derived exosomes for miR-31-5p delivery to treat diabetic wounds. A pharmacokinetic of mEXO-31 revealed remarkable stability metrics, with approximately 60% of intact encapsulated miR-31-5p after a 5-day incubation at 37°C while free miRNA underwent near-complete degradation. Dosage optimization demonstrated that administration fo 1.0 μg/μL mEXO-31 at three timepoints (days 0, 5, and 10) produced optimal therapeutic efficacy. Quantitative biological responses established dose-dependent effects, with mEXO-31 inducing approximately 2-fold enhancement in endothelial cell proliferation and angiogenic tube formation, and an 1.5-fold increase in migration capacity (Figure 4).165 In streptozotocin-induced diabetic mice and in methylglyoxal-treated HUVECs, the Keap1/Nrf2 signaling pathway is activated. Lactogenic exosomes acted as novel, efficient, and nontoxic siRNA carriers. The injection of M-Exos-siKeap1 significantly accelerated wound healing in a diabetic mouse wound model, enhancing collagen deposition and neovascularization.166 These results demonstrate the feasibility of milk exosomes as an siRNA delivery system and milk-derived exosomes can promote diabetic wound healing by ameliorating oxidative stress.
![]() |
Figure 4 Schematic illustrating milk exosome-mediated miR-31-5p delivery for diabetic wound healing. (A) Isolation process of milk-derived exosomes (mEXO) and preparation of miR-31-5p-loaded exosomes (mEXO-31) via electroporation. (B) Molecular mechanism of mEXO-31 in endothelial cells, where internalized miR-31-5p targets HIF1AN mRNA, reducing HIF1AN protein expression and consequently enhancing endothelial cell proliferation, migration, and tube formation. (C) Therapeutic application of mEXO-31 in diabetic wound model, showing local injection promotes angiogenesis and accelerates wound closure. Reproduced with permission from Yan C, Chen J, Wang C et al. Milk exosomes-mediated miR-31-5p delivery accelerates diabetic wound healing through promoting angiogenesis. Drug Delivery. 2022;29(1):214–228. Copyright (2022) Taylor & Francis Group.165 |
Amniotic Fluid-Derived Exosomes
In the field of reproductive biology, exosomes are associated with embryogenesis, placentation, maintenance of pregnancy, and delivery based on exosome characteristics (number, cargo content, and function) in various biological samples during pregnancy.167 These samples consist mainly of maternal plasma, cervicovaginal fluid, UCB, and amniotic fluid. Due to the enrichment of fetal exosomes in amniotic fluid, amniotic fluid exosomes play an important role in fetal development. They can support fetal growth and development by regulating intercellular signaling and provide protection when the fetus is exposed to stress.168 Fetal exosomes in maternal circulation account for approximately 35% of the total circulating exosomes.169 Although the collection of amniotic fluid involves ultrasound-guided invasive methods, amniotic exosomes provide valuable information about the status of the pregnancy and can reflect the functional status of the fetus in utero.167
The successful application of adult stem cells is limited because they retain epigenetic alterations even after reprogramming.170,171 Fetal stem cells can overcome this limitation, especially those derived from amniotic fluid collected during cesarean section, late gestational amniotic reduction, or routine amniocentesis.172 Studies have shown that human amniotic fluid stem cells accelerated skin wound healing with less fibrotic scarring, similar to fetal wound healing.173 It was later demonstrated that human amniotic fluid stem cells (hAFSC-exo) improved the regeneration of hair follicles, nerves, and blood vessels and increased skin cell proliferation and the natural distribution of collagen during wound healing; thus, demonstrated significant antifibrotic scarring properties during wound healing.174 Overall, human amniotic fluid stem cells independently produce paracrine effectors and are secreted in exosomes, thereby modulating local immune cell activity. They possessed anti-inflammatory and immunomodulatory properties, modulated the physicochemical microenvironment of the wound, and promoted complete wound regeneration, especially in scarless wound healing.175 Although studies addressing the specific mechanisms of amniotic fluid exocytosis in diabetic wound healing are limited, the results of the studies that have been conducted suggest their potential application in this field.
Plant-Derived Exosome-Like Nanoparticles
In recent years, researchers have gradually discovered and isolated exosomes from plants with particle sizes in the range of 30–500 nm and characterized them.176 It was found that plant-derived exosomes can be isolated from a wide range of fruits and vegetables, and some even from traditional herbs and fungi177 (Table 2). PENs from a wide range of sources have a specialized subcellular structure. As drug carriers, they can enhance drug stability and cellular uptake.178 Reports indicate that PENs have great potential in regulating immune function, inflammation, the microbiome, and tissue regeneration.179 PENs promote cell proliferation, migration, and angiogenesis, among other mechanisms, and can significantly accelerate wound healing. Wheat-derived PENs were found to have dose-dependent proliferative and migratory effects on endothelial cells, epithelial cells, and DFs in vitro. They promoted formation of tubular structures in endothelial cells and increased the transcription levels of type I collagen. Additionally, it promotes endothelial cell angiogenesis through the ERK and AKT/mTOR pathways, repairing full-thickness diabetic skin ulcers.180–182
![]() |
Table 2 Fruit, Vegetable, and Herb-Derived PELNs for Wound Healing |
A ginseng-derived exosome (G-Exo) was successfully prepared and proved to promote diabetic wound healing.197 Applying G-Exos to mouse skin wounds showed that they can promote skin cell proliferation, accelerate injured skin recovery, and reduce inflammation.198 Additionally, in animal models, G-Exos assisted in nerve regeneration by upregulating PI3K signaling to promote nerve repair, and by intervening in the RAS/ERK pathway to promote the expression of neurotrophic factors and accelerate nerve regeneration.199 Xiong et al developed a whole-course-repair hydrogel system (HA-ADH/OSA@Mg@sEVs) based on engineered ginseng-derived small extracellular vesicles (G-sEVs). This system promoted diabetic wound healing through neurogenesis-angiogenesis crosstalk and macrophage reprogramming. Furthermore, G-sEVs carrying didymin (DM) facilitated M1 to M2 macrophage transition while the released Mg2+ synergistically enhanced angiogenesis with differentiated neural cells. In STZ-induced diabetic mice, the system significantly accelerated wound healing without systemic toxicity, demonstrating its therapeutic potential as it addresses multiple phases of the wound healing process (Figure 5).200 Additionally, exosomes derived from aloe vera cortex (rAEVs) enhance the migration ability of HaCaT and HDF cells by scavenging free radicals and ROS. Moreover, rAEVs can promote tube formation by endothelial cells, increase blood supply, and reduce the secretion of inflammatory factors. They improve skin texture and scar appearance through heat shock proteins, promoting skin regeneration.201–203
![]() |
Figure 5 Schematic of the beneficial role of HA-ADH /OSA @ Mg@sEVs hydrogel. The one-step preparation of HA-ADH/OSA@Mg hydrogel encapsulates G-sEVsDM without compromising their activity. With its Mg2+ release, the hydrogel recruits mesenchymal stem cells and induces neurogenic differentiation and macrophage reprogramming, promoting a prohealing environment. This leads to enhanced angiogenesis and a neurogenesis-angiogenesis cycle at the wound site. Reproduced with permission from Xiong Y, Lin Z, Bu P et al. A Whole-Course-Repair System Based on Neurogenesis-Angiogenesis Crosstalk and Macrophage Reprogramming Promotes Diabetic Wound Healing. Advanced Materials. 2023, 35, 2212300. Copyright (2023) Wiley Online Library.200 |
Plant-derived exosomes can also regulate multiple cell signaling pathways, such as the Wnt/β-catenin signaling pathway and the TGF-β signaling pathway, thereby promoting the function of fibroblasts and endothelial cells.204,205 PENs enhanced the formation of tubular structures in HUVECs, marking an important milestone in therapeutic approaches to wound healing. Grapefruit-derived exos upregulated the expression of proliferation- and migration-related genes in a dose-dependent manner, increased the tube-forming capacity of HUVECs, in the HaCaT cell model,206 and played a dominant role in the anti-inflammatory regulation of Wnt.207 Another study found that ginger-derived exosomes can promote Nrf2 nuclear translocation and preferentially induce the expression levels of antioxidant HO-1 and anti-inflammatory cytokine IL-10, stimulating the production of anti-inflammatory cytokines, thereby effectively exerting their anti-inflammatory efficacy.178 Interestingly, human BMSCs internalized strawberry-derived exos, and incubation for 120 h did not produce negative effects. BMSCs pretreated with strawberry-derived exos were resistant to hydrogen peroxide-induced oxidative stress and possessed potentially beneficial activities.208 These findings may yield valuable insights for exploring the regenerative capacity of PENs in various tissues, requiring extensive empirical evidence to validate their therapeutic potential.209
In addition to acting as therapeutic agents for disease intervention, PELNs can function as carriers to deliver various therapeutic molecules (such as proteins, siRNA, and therapeutic drugs) to relevant disease sites. Moreover, their endogenous bioactive components can exert synergistic therapeutic effects, significantly enhancing clinical efficacy.210 Compared to mammalian exosomes, PELNs have unique advantages such as being less easily detected by the immune system as well as higher bioavailability and low toxicity211 (Table 3).
![]() |
Table 3 Comparison of MDEs and PELNs Used for Diabetic Wound Healing |
These structural and compositional differences directly influence the selection of delivery strategies, MDEs are typically combined with albumin or hyaluronic acid-based hydrogel systems to form controlled-release complexes for local administration, and can enhance their in vivo circulation time and targeting through covalent modification methods such as PEGylation;219,220 whereas PELNs, due to their unique phospholipid composition and stable membrane structure, are more suitable for combination with polysaccharide or polymer matrices, such as chitosan and polylactic acid, through non-covalent modification methods like electrostatic interactions, significantly enhancing their stability in the gastrointestinal environment and oral bioavailability.221 With respect to the drug release mechanism, MDEs commonly use pH-responsive strategies to accommodate endosomal/lysosomal escape needs, whereas PELNs, due to their special membrane composition, are more compatible with photosensitive and redox-sensitive materials for constructing intelligent responsive release systems.215,222,223 Understanding the structure-function relationship of these two types of extracellular vesicles can guide the development of complementary delivery platforms with combined advantages, providing new approaches for precision treatment of challenging conditions such as diabetic wounds.
Challenges in Exosomes Application
Exosomes have promising potential for treating diabetic wounds; however, several challenges must be overcome before their broad clinical use. The first issue is the stability and targeting of the exosomes. Research indicates that half-life of exosomes in the body typically ranges from just a few minutes to several hours, primarily due to their rapid clearance by the immune system. This short retention period substantially restricts the effectiveness of exosomes within the body.224 To address this, altering the surface properties of exosomes through modifications, such as PEGylation and incorporation of nanoparticles, can enhance their circulatory stability and extend their in vivo retention time, thereby augmenting their therapeutic impact. The second issue is that exosome contents and functions are influenced by the type and state of the secreting cells, with involve intricate signaling pathways. The transfer of unknown bioactive molecular mixtures in exosomes may produce unpredictable biological effects and pose a safety hazard. Thus, further investigation is essential to gain a comprehensive understanding of the exosome composition as well any potential toxicity associated with the exosomes.225 The third issue is that the large-scale production of exosomes faces multiple challenges. First, exosome production primarily relies on cell culture, requiring serum and growth factors that are expensive and cumbersome to obtain. Second, existing bioreactors have limited capacity, and large-scale cultivation increases contamination risks. Meanwhile, difficulties in quality control, batch-to-batch consistency issues, and the high associated costs limit the clinical application of exosomes in diabetic wound healing. To address these issues, cultivation conditions can be optimized by developing serum free or synthetic media to reduce costs and introducing automated equipment to decrease manual operations and improve efficiency. Additionally, standardized isolation and characterization techniques, including developing automated isolation and purification equipment, as well as a combination multiple technologies such as ultrafiltration and affinity chromatography to reduce purification steps while preserving exosome activity need to be established226 (Figure 6). These improvements not only increase yield and quality but also lay the foundation for exosome characteristics and safety assessment, promoting their clinical translation. Lastly, exosomes are temperature-sensitive, with harsh storage and transportation conditions, and freezing and thawing may affect their biological activity. The development of new exosome stabilization technology to ensure the consistency of its quality in the process of transformation and application is also a key direction worthy of attention.
![]() |
Figure 6 Schematic of common exosomal separation techniques. (A) Ultracentrifugation, (B) density gradient centrifugation, (C) dead-end filtration (DEF), (D) tangential flow filtration (TFF), (E) size-exclusion chromatography, and (F) immunoaffinity. Reproduced with permission from Chen J et al. Review on Strategies and Technologies for Exosome Isolation and Purification. Frontiers in Bioengineering and Biotechnology. 2022;9:811971. Copyright (2022) Frontiers Media.226 |
Beyond the numerous challenges inherent to exosomes, their application in the treatment of diabetic wounds introduces particular complexities. Patients with diabetes commonly experience peripheral neuropathy and vascular disorders, which can cause minor injuries to evolve into refractory ulcers. This progression not only increases susceptibility to infections but also hinders wound healing. Thus, the delivery of exosomes needs more exacting standards, and a single injection method may be inadequate. Developing innovative exosome delivery systems is imperative to enhance targeting precision and therapeutic effectiveness in the treatment of diabetic wounds. Furthermore, the preponderance of exosome research in wound healing has been conducted in rodent models, leaving open the question of whether these findings can be extrapolated to patients with diabetes.227 To the best of our knowledge, there have been no documented cases of exosome usage in individuals with either diabetic or nondiabetic wounds to date, because most clinical study products have yet to receive approval for human application.228 This constraint hampers our capacity to fully grasp the true efficacy of exosome therapy in the context of diabetic wound treatment. As pathophysiological mechanisms of diabetic wounds differ from those of typical wounds, it is essential to develop a diabetic animal model that more accurately represents human physiology and assess the therapeutic promise of exosomes within such a model.
Summary and Outlook
In the field of diabetic wound repair, exosomes are rapidly advancing from basic research to clinical application. Clinical trials have demonstrated that exosome-hydrogel composites together with negative pressure wound therapy can reduce healing time by up to 40% without increasing infection risk.229,230 Moreover, the combination of photodynamic therapy with exosomes has significant synergistic effects in eliminating multi-drug resistant bacterial biofilms. To better understand their mechanisms of action, researchers have developed diabetic foot ulcer organoid models, which not only facilitate treatment evaluation but also promote the development of personalized medicine strategies. By analyzing patients’ genetic backgrounds and differences in wound microenvironments, researchers can screen for exosome subgroups with specific miRNA or protein profiles, thereby achieving precision treatment. More importantly, using gene editing technology to modify HLA molecules in ADSC-Exos effectively addresses the key clinical challenge of immune rejection.231 Accordingly, interdisciplinary innovation further expands their application prospects. The integration of biomaterials science and nanoengineering enables silk fibroin patches to extend exosome retention time to over 72 h, achieving precise release of growth factors through pH-responsive mechanisms. Meanwhile, the combination of biomimetic scaffolds and optogenetic technology can regulate NO release, directly improving wound angiogenesis.232 These technological advances can have synergistic effects with artificial intelligence systems, providing data support for clinical decision-making.233
This review describes the current status of application of exosomes from various sources in diabetic wound healing. Although exosome research is still in the developmental stage, there has been substantial progress in clinical translation. With growing research into tissue repair mechanisms, this therapy aims to offer more effective treatment options for patients with diabetic wounds, fulfilling the translational medicine objective of moving from laboratory to bedside.
Abbreviations
DFUs, diabetic foot ulcers; ECM, extracellular matrix; MSC-Exos, mesenchymal stem cell-derived exosomes; BMSC-Exos, bone marrow mesenchymal stem cell-derived exosomes; ADSC-Exos, adipose-derived mesenchymal stem cell-derived exosomes; UCMSCs, umbilical cord mesenchymal stem cells; HUVECs, human umbilical vein endothelial cells; M-Exos, macrophage-derived exosomes; EPC-Exos, endothelial progenitor cell-derived exosomes; DF-Exos, dermal fibroblast-derived exosomes; USC-Exos, urine-derived stem cell-derived exosomes; SEs, salivary exosomes; MT-Exos, melatonin-stimulated bone marrow stem cell-derived exosomes; ATV-Exos, atorvastatin-pretreated bone marrow stem cell-derived exosomes; hyBMSC-Exos, hypoxic bone marrow mesenchymal stem cell-derived exosomes; UCB, umbilical cord blood; AGEs, advanced glycation end products; HBOT, hyperbaric oxygen therapy; PI3K, phosphoinositide 3-kinase; mTOR, mammalian target of rapamycin; S1P, sphingosine-1-phosphate; ERK, extracellular signal-regulated kinase; MAPK, mitogen-activated protein kinase; DM, didymin; rAEVs, exosomes derived from aloe vera cortex.
Acknowledgments
This work was supported by the open Project of Yunnan Clinical Medical Research Center for Geriatric Diseases, Grant/Award Numbers: 2022YJZX-LN18, 2022YJZX-LN20, 2023YJZX-LN08, 2023YJZX-LN10.
Disclosure
The authors report no conflicts of interest in this work.
References
1. Chen J, Qin S, Liu S, et al. Targeting matrix metalloproteases in diabetic wound healing. Front Immunol. 2023;14:1089001. doi:10.3389/fimmu.2023.1089001
2. Cho H, Blatchley MR, Duh EJ, Gerecht S. Acellular and cellular approaches to improve diabetic wound healing. Adv Drug Deliv Rev. 2019;146:267–288. doi:10.1016/j.addr.2018.07.019
3. Cheun TJ, Jayakumar L, Sideman MJ, et al. Short-term contemporary outcomes for staged versus primary lower limb amputation in diabetic foot disease. J Vasc Surg. 2020;72(2). doi:10.1016/j.jvs.2019.10.083
4. Khan NU, Chengfeng X, Jiang M-Q, et al. Obstructed vein delivery of ceftriaxone via poly(vinyl-pyrrolidone)-iodine-chitosan nanofibers for the management of diabetic foot infections and burn wounds. Int J Biol Macromol. 2024;277(Pt 2):134166. doi:10.1016/j.ijbiomac.2024.134166
5. Dixon D, Edmonds M. Managing diabetic foot ulcers: pharmacotherapy for wound healing. Drugs. 2021;81(1):29–56. doi:10.1007/s40265-020-01415-8
6. Zhou C, Zhang B, Yang Y, et al. Stem cell-derived exosomes: emerging therapeutic opportunities for wound healing. Stem Cell Res Ther. 2023;14(1):107. doi:10.1186/s13287-023-03345-0
7. Armstrong DG, Tan T-W, Boulton AJM, Bus SA. Diabetic foot ulcers: a review. JAMA. 2023;330(1):62–75. doi:10.1001/jama.2023.10578
8. Feng J, Yao Y, Wang Q, et al. Exosomes: potential key players towards novel therapeutic options in diabetic wounds. Biomed Pharmacother. 2023;166:115297. doi:10.1016/j.biopha.2023.115297
9. Khan ZU, Razzaq A, Khan A, et al. Physicochemical characterizations and pharmacokinetic evaluation of pentazocine solid lipid nanoparticles against inflammatory pain model. Pharmaceutics. 2022;14(2). doi:10.3390/pharmaceutics14020409
10. Song J, Razzaq A, Khan NU, Iqbal H, Ni J. Chitosan/poly (3-hydroxy butyric acid-co-3-hydroxy valeric acid) electrospun nanofibers with cephradine for superficial incisional skin wound infection management. Int J Biol Macromol. 2023;250:126229. doi:10.1016/j.ijbiomac.2023.126229
11. Feng W, Wang X. Review of diabetic foot ulcers. JAMA. 2023;330(17):1695. doi:10.1001/jama.2023.17191
12. Cheng S, Wang H, Pan X, et al. Dendritic hydrogels with robust inherent antibacterial properties for promoting bacteria-infected wound healing. ACS Appl Mater Interfaces. 2022;14(9):11144–11155. doi:10.1021/acsami.1c25014
13. Qi X, Xiang Y, Li Y, et al. An ATP-activated spatiotemporally controlled hydrogel prodrug system for treating multidrug-resistant bacteria-infected pressure ulcers. Bioact Mater. 2025;45:301–321. doi:10.1016/j.bioactmat.2024.11.029
14. Qi X, Shi Y, Zhang C, et al. A hybrid hydrogel with intrinsic immunomodulatory functionality for treating multidrug-resistant Pseudomonas aeruginosa infected diabetic foot ulcers. ACS Mater Lett. 2024;6(7):2533–2547.
15. Holl J, Kowalewski C, Zimek Z, et al. Chronic diabetic wounds and their treatment with skin substitutes. Cells. 2021;10(3). doi:10.3390/cells10030655
16. Zafar N, Uzair B, Niazi MBK, et al. Green synthesis of ciprofloxacin-loaded cerium oxide/chitosan nanocarrier and its activity against MRSA-induced mastitis. J Pharm Sci. 2021;110(10):3471–3483. doi:10.1016/j.xphs.2021.06.017
17. Abbas S, Uzair B, Sajjad S, et al. Dual-functional green facile CuO/MgO nanosheets composite as an efficient antimicrobial agent and photocatalyst. Arab J Sci Eng. 2022;47(5):5895–5909.
18. Capó X, Monserrat-Mesquida M, Quetglas-Llabrés M, et al. Hyperbaric oxygen therapy reduces oxidative stress and inflammation, and increases growth factors favouring the healing process of diabetic wounds. Int J Mol Sci. 2023;24(8):7040.
19. Li Y, Zhu Z, Li S, et al. Exosomes: compositions, biogenesis, and mechanisms in diabetic wound healing. J Nanobiotechnol. 2024;22(1):398. doi:10.1186/s12951-024-02684-1
20. Z L, W Q, G M, et al. Mesenchymal stem cell-derived exosomes in various chronic liver diseases: hype or hope? J Inflam Res. 2024;17:171–189. doi:10.2147/jir.S439974
21. Tienda-Vázquez MA, Hanel JM, Márquez-Arteaga EM, et al. Exosomes: a promising strategy for repair, regeneration and treatment of skin disorders. Cells. 2023;12(12). doi:10.3390/cells12121625
22. Gunassekaran GR, Poongkavithai Vadevoo SM, Baek M-C, Lee B. M1 macrophage exosomes engineered to foster M1 polarization and target the IL-4 receptor inhibit tumor growth by reprogramming tumor-associated macrophages into M1-like macrophages. Biomaterials. 2021;278:121137. doi:10.1016/j.biomaterials.2021.121137
23. Li C, Liu S-Y, Zhang M, et al. Sustained release of exosomes loaded into polydopamine-modified chitin conduits promotes peripheral nerve regeneration in rats. Neural Regen Res. 2022;17(9):2050–2057. doi:10.4103/1673-5374.335167
24. Ha DH, Kim S-D, Lee J, et al. Toxicological evaluation of exosomes derived from human adipose tissue-derived mesenchymal stem/stromal cells. Regul Toxicol Pharmacol. 2020;115:104686. doi:10.1016/j.yrtph.2020.104686
25. Saleem M, Shahzad KA, Marryum M, et al. Exosome-based therapies for inflammatory disorders: a review of recent advances. Stem Cell Res Ther. 2024;15(1):477. doi:10.1186/s13287-024-04107-2
26. Jin W, Li Y, Yu M, Ren D, Han C, Guo S. Advances of exosomes in diabetic wound healing. Burns Trauma. 2025;
27. Yu D-G, He W, He C, Liu H, Yang H. Versatility of electrospun Janus wound dressings. Nanomedicine. 2025;20(3):271–278. doi:10.1080/17435889.2024.2446139
28. Chaerani MAD, Baraja HA, Putranti IND, Reswari KE, Ainurofiq A. Alginate/collagen hydrogel containing calcium from eggshell waste as a potential diabetic foot ulcer wound dressing. Mater Today Commun. 2025;42:111417.
29. Wang Y, Jiang M, Zheng X, et al. Application of exosome engineering modification in targeted delivery of therapeutic drugs. Biochem Pharmacol. 2023;215:115691. doi:10.1016/j.bcp.2023.115691
30. Huang L, Wu E, Liao J, Wei Z, Wang J, Chen Z. Research advances of engineered exosomes as drug delivery carrier. ACS Omega. 2023;8(46):43374–43387. doi:10.1021/acsomega.3c04479
31. Ahmed W, Mushtaq A, Ali S, Khan N, Liang Y, Duan L. Engineering approaches for exosome cargo loading and targeted delivery: biological versus chemical perspectives. ACS Biomater Sci Eng. 2024;10(10):5960–5976. doi:10.1021/acsbiomaterials.4c00856
32. Jiao Y-R, Chen K-X, Tang X, et al. Exosomes derived from mesenchymal stem cells in diabetes and diabetic complications. Cell Death Dis. 2024;15(4):271. doi:10.1038/s41419-024-06659-w
33. Nishio M, Teranishi Y, Morioka K, Yanagida A, Shoji A. Real-time assay for exosome membrane fusion with an artificial lipid membrane based on enhancement of gramicidin A channel conductance. Biosens Bioelectron. 2020;150:111918. doi:10.1016/j.bios.2019.111918
34. Mashouri L, Yousefi H, Aref AR, Ahadi AM, Molaei F, Alahari SK. Exosomes: composition, biogenesis, and mechanisms in cancer metastasis and drug resistance. Mol Cancer. 2019;18(1):75. doi:10.1186/s12943-019-0991-5
35. An Y, Sun J-X, Ma S-Y, et al. From plant based therapy to plant-derived vesicle-like nanoparticles for cancer treatment: past, present and future. Int J Nanomed. 2025;20:3471–3491. doi:10.2147/IJN.S499893
36. Shao J, Zaro J, Shen Y. Advances in exosome-based drug delivery and tumor targeting: from tissue distribution to intracellular fate. Int J Nanomed. 2020;15:9355–9371. doi:10.2147/IJN.S281890
37. Bai C, Liu J, Zhang X, et al. Research status and challenges of plant-derived exosome-like nanoparticles. Biomed Pharmacother. 2024;174:116543. doi:10.1016/j.biopha.2024.116543
38. Jing S, Li H, Xu H. Mesenchymal stem cell derived exosomes therapy in diabetic wound repair. Int J Nanomed. 2023;18:2707–2720. doi:10.2147/IJN.S411562
39. Zhao W, Zhang R, Zang C, et al. Exosome derived from mesenchymal stem cells alleviates pathological scars by inhibiting the proliferation, migration and protein expression of fibroblasts via delivering miR-138-5p to target SIRT1. Int J Nanomed. 2022;17:4023–4038. doi:10.2147/IJN.S377317
40. Ha DH, Kim H-K, Lee J, et al. Mesenchymal stem/stromal cell-derived exosomes for immunomodulatory therapeutics and skin regeneration. Cells. 2020;9(5). doi:10.3390/cells9051157
41. Capilla-González V, Herranz-Pérez V, Sarabia-Estrada R, Kadri N, Moll G. Editorial: mesenchymal stromal cell therapy for regenerative medicine. Front Cell Neurosci. 2022;16:932281. doi:10.3389/fncel.2022.932281
42. Tang Y, Zhou Y, Li H-J. Advances in mesenchymal stem cell exosomes: a review. Stem Cell Res Ther. 2021;12(1):71. doi:10.1186/s13287-021-02138-7
43. Teng L, Maqsood M, Zhu M, et al. Exosomes derived from human umbilical cord mesenchymal stem cells accelerate diabetic wound healing via promoting M2 macrophage polarization, angiogenesis, and collagen deposition. Int J Mol Sci. 2022;23(18). doi:10.3390/ijms231810421
44. Luo Z, Lin J, Sun Y, Wang C, Chen J. Bone marrow stromal cell-derived exosomes promote muscle healing following contusion through macrophage polarization. Stem Cells Dev. 2021;30(3):135–148. doi:10.1089/scd.2020.0167
45. Liu W, Yu M, Xie D, et al. 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(1):259. doi:10.1186/s13287-020-01756-x
46. Geng X, Qi Y, Liu X, Shi Y, Li H, Zhao L. A multifunctional antibacterial and self-healing hydrogel laden with bone marrow mesenchymal stem cell-derived exosomes for accelerating diabetic wound healing. Biomater Adv. 2022;133:112613. doi:10.1016/j.msec.2021.112613
47. Huang K, Mi B, Xiong Y, et al. Angiogenesis during diabetic wound repair: from mechanism to therapy opportunity. Burns Trauma. 2025;13:tkae052. doi:10.1093/burnst/tkae052
48. Han Z-F, Cao J-H, Liu Z-Y, Yang Z, Qi R-X, Xu H-L. Exosomal lncRNA KLF3-AS1 derived from bone marrow mesenchymal stem cells stimulates angiogenesis to promote diabetic cutaneous wound healing. Diabetes Res Clin Pract. 2022;183:109126. doi:10.1016/j.diabres.2021.109126
49. Zhang L, Ouyang P, He G, et al. Exosomes from microRNA-126 overexpressing mesenchymal stem cells promote angiogenesis by targeting the PIK3R2-mediated PI3K/Akt signalling pathway. J Cell Mol Med. 2021;25(4):2148–2162. doi:10.1111/jcmm.16192
50. 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
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. Lu W, Du X, Zou S, et al. IFN-γ enhances the therapeutic efficacy of MSCs-derived exosome via miR-126-3p in diabetic wound healing by targeting SPRED1. J Diabetes. 2024;16(1):e13465. doi:10.1111/1753-0407.13465
53. dD A, D FM, K SL, G KA. Targeting epigenetic mechanisms in diabetic wound healing. Transl Res. 2019;204:39–50. doi:10.1016/j.trsl.2018.10.001
54. Ning K, Yang B, Chen M, et al. Functional heterogeneity of bone marrow mesenchymal stem cell subpopulations in physiology and pathology. Int J Mol Sci. 2022;23(19). doi:10.3390/ijms231911928
55. Shi Y, Wang S, Liu D, et al. Exosomal miR-4645-5p from hypoxic bone marrow mesenchymal stem cells facilitates diabetic wound healing by restoring keratinocyte autophagy. Burns Trauma. 2024;12:tkad058. doi:10.1093/burnst/tkad058
56. 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
57. 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(1):198. doi:10.1186/s13287-020-01723-6
58. M M, R P, B J, J C, N 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
59. Yu H, Wu Y, Zhang B, et al. Exosomes derived from E2F1-/- adipose-derived stem cells promote skin wound healing via miR-130b-5p/TGFBR3 axis. Int J Nanomed. 2023;18:6275–6292. doi:10.2147/IJN.S431725
60. Song Y, You Y, Xu X, et al. Adipose-derived mesenchymal stem cell-derived exosomes biopotentiated extracellular matrix hydrogels accelerate diabetic wound healing and skin regeneration. Adv Sci. 2023;10(30):e2304023. doi:10.1002/advs.202304023
61. L C, W J, G W, Q X, Y R, C X. Therapeutic potential of exosomes from adipose-derived stem cells in chronic wound healing. Front Surg. 2022;9:1030288. doi:10.3389/fsurg.2022.1030288
62. Shen K, Wang XJ, Liu KT, et al. Effects of exosomes from human adipose-derived mesenchymal stem cells on inflammatory response of mouse RAW264.7 cells and wound healing of full-thickness skin defects in mice. Zhonghua Shao Shang Yu Chuang Mian Xiu Fu Za Zhi. 2022;38(3):215–226. doi:10.3760/cma.j.cn501120-20201116-00477
63. Zhang Y, Mei H, Chang X, Chen F, Zhu Y, Han X. Adipocyte-derived microvesicles from obese mice induce M1 macrophage phenotype through secreted miR-155. J Mol Cell Biol. 2016;8(6):505–517.
64. Li X, Xie X, Lian W, et al. Exosomes from adipose-derived stem cells overexpressing Nrf2 accelerate cutaneous wound healing by promoting vascularization in a diabetic foot ulcer rat model. Exp Mol Med. 2018;50(4). doi:10.1038/s12276-018-0058-5
65. Wang J, Wu H, Peng Y, et al. Hypoxia adipose stem cell-derived exosomes promote high-quality healing of diabetic wound involves activation of PI3K/Akt pathways. J Nanobiotechnol. 2021;19(1):202. doi:10.1186/s12951-021-00942-0
66. 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(11):2182–2189. doi:10.1242/jcs.170373
67. Zhang Y, Han F, Gu L, et al. 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(4):375–383. doi:10.1007/s10735-020-09887-4
68. Wang L, Hu L, Zhou X, et al. Exosomes secreted by human adipose mesenchymal stem cells promote scarless cutaneous repair by regulating extracellular matrix remodelling. Sci Rep. 2017;7(1):13321. doi:10.1038/s41598-017-12919-x
69. Chen Md G, Wu Md Y, Zou Md L, Zeng Md Y. Effect of microRNA-146a modified adipose-derived stem cell exosomes on rat back wound healing. Int J Low Extrem Wounds. 2023;22(4):704–712. doi:10.1177/15347346211038092
70. An Y, Lin S, Tan X, et al. Exosomes from adipose-derived stem cells and application to skin wound healing. Cell Prolif. 2021;54(3):e12993. doi:10.1111/cpr.12993
71. Li Y, Zhang J, Shi J, et al. Exosomes derived from human adipose mesenchymal stem cells attenuate hypertrophic scar fibrosis by miR-192-5p/IL-17RA/Smad axis. Stem Cell Res Ther. 2021;12(1):221. doi:10.1186/s13287-021-02290-0
72. Shaikh MS, Shahzad Z, Tash EA, Janjua OS, Khan MI, Zafar MS. Human umbilical cord mesenchymal stem cells: current literature and role in periodontal regeneration. Cells. 2022;11(7). doi:10.3390/cells11071168
73. Liu C, Lu C, Yixi L, et al. Exosomal Linc00969 induces trastuzumab resistance in breast cancer by increasing HER-2 protein expression and mRNA stability by binding to HUR. Breast Cancer Res. 2023;25(1):124. doi:10.1186/s13058-023-01720-6
74. Zhang Y, Hao Z, Wang P, et al. Exosomes from human umbilical cord mesenchymal stem cells enhance fracture healing through HIF-1α-mediated promotion of angiogenesis in a rat model of stabilized fracture. Cell Prolif. 2019;52(2):e12570. doi:10.1111/cpr.12570
75. Kim Y-J, Yoo SM, Park HH, et al. Exosomes derived from human umbilical cord blood mesenchymal stem cells stimulates rejuvenation of human skin. Biochem Biophys Res Commun. 2017;493(2):1102–1108. doi:10.1016/j.bbrc.2017.09.056
76. Dehghani L, Owliaee I, Sadeghian F, Shojaeian A. The therapeutic potential of human umbilical cord mesenchymal stromal cells derived exosomes for wound healing: harnessing exosomes as a cell-free therapy. J Stem Cells Regen Med. 2024;20(1):14–23. doi:10.46582/jsrm.2003003
77. 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
78. Yan C, Xv Y, Lin Z, et al. Human umbilical cord mesenchymal stem cell-derived exosomes accelerate diabetic wound healing via ameliorating oxidative stress and promoting angiogenesis. Front Bioeng Biotechnol. 2022;10:829868. doi:10.3389/fbioe.2022.829868
79. Sun Y, Shi H, Yin S, et al. Human mesenchymal stem cell derived exosomes alleviate type 2 diabetes mellitus by reversing peripheral insulin resistance and relieving β-cell destruction. ACS Nano. 2018;12(8):7613–7628. doi:10.1021/acsnano.7b07643
80. Deng Y, Li Y, Chu Z, Dai C, Ge J. Exosomes from umbilical cord-derived mesenchymal stem cells combined with gelatin methacryloyl inhibit vein graft restenosis by enhancing endothelial functions. J Nanobiotechnol. 2023;21(1):380. doi:10.1186/s12951-023-02145-1
81. Liu N, Dong J, Li L, Liu F. Osteoimmune interactions and therapeutic potential of macrophage-derived small extracellular vesicles in bone-related diseases. Int J Nanomed. 2023;18:2163–2180. doi:10.2147/IJN.S403192
82. Cheng X, Zhou H, Zhou Y, Song C. M2 macrophage-derived exosomes inhibit apoptosis of HUVEC cell through regulating miR-221-3p expression. Biomed Res Int. 2022;2022:1609244. doi:10.1155/2022/1609244
83. Wang Y, Zhao M, Liu S, et al. Macrophage-derived extracellular vesicles: diverse mediators of pathology and therapeutics in multiple diseases. Cell Death Dis. 2020;11(10):924. doi:10.1038/s41419-020-03127-z
84. Clayton SM, Shafikhani SH, Soulika AM. Macrophage and neutrophil dysfunction in diabetic wounds. Adv Wound Care. 2024;13(9):463–484. doi:10.1089/wound.2023.0149
85. Zhao H, Huang J, Li Y, et al. ROS-scavenging hydrogel to promote healing of bacteria infected diabetic wounds. Biomaterials. 2020;258:120286. doi:10.1016/j.biomaterials.2020.120286
86. Wang Z, Xu H, Xue B, et al. MSC-derived exosomal circMYO9B accelerates diabetic wound healing by promoting angiogenesis through the hnRNPU/CBL/KDM1A/VEGFA axis. Commun Biol. 2024;7(1):1700. doi:10.1038/s42003-024-07367-z
87. Luo Z, Qi B, Sun Y, et al. Engineering bioactive M2 macrophage-polarized, anti-inflammatory, miRNA-based liposomes for functional muscle repair: from exosomal mechanisms to biomaterials. Small. 2022;18(34):e2201957. doi:10.1002/smll.202201957
88. Jean-Toussaint R, Lin Z, Tian Y, et al. Therapeutic and prophylactic effects of macrophage-derived small extracellular vesicles in the attenuation of inflammatory pain. Brain Behav Immun. 2021;94:210–224. doi:10.1016/j.bbi.2021.02.005
89. Lyu L, Cai Y, Zhang G, et al. Exosomes derived from M2 macrophages induce angiogenesis to promote wound healing. Front Mol Biosci. 2022;9:1008802. doi:10.3389/fmolb.2022.1008802
90. Li D, Zhang C, Gao Z, et al. Curcumin-loaded macrophage-derived exosomes effectively improve wound healing. Mol Pharm. 2023;20(9):4453–4467. doi:10.1021/acs.molpharmaceut.3c00062
91. Zeng J, Sun Z, Zeng F, Gu C, Chen X. M2 macrophage-derived exosome-encapsulated microneedles with mild photothermal therapy for accelerated diabetic wound healing. Mater Today Bio. 2023;20:100649. doi:10.1016/j.mtbio.2023.100649
92. Petersen JD, Mekhedov E, Kaur S, Roberts DD, Zimmerberg J. Endothelial cells release microvesicles that harbour multivesicular bodies and secrete exosomes. J Extracell Biol. 2023;2(4):e79. doi:10.1002/jex2.79
93. Fang X, Zhang Y, Zhang Y, et al. Endothelial extracellular vesicles: their possible function and clinical significance in diabetic vascular complications. J Transl Med. 2024;22(1):944. doi:10.1186/s12967-024-05760-0
94. Yan F, Liu X, Ding H, Zhang W. Paracrine mechanisms of endothelial progenitor cells in vascular repair. Acta Histochem. 2022;124(1):151833. doi:10.1016/j.acthis.2021.151833
95. Wan G, Chen Y, Chen J, et al. Regulation of endothelial progenitor cell functions during hyperglycemia: new therapeutic targets in diabetic wound healing. J Mol Med. 2022;100(4):485–498. doi:10.1007/s00109-021-02172-1
96. Xu M, Su X, Xiao X, et al. Hydrogen peroxide-induced senescence reduces the wound healing-promoting effects of mesenchymal stem cell-derived exosomes partially via miR-146a. Aging Dis. 2021;12(1):102–115. doi:10.14336/AD.2020.0624
97. Li P, Hong G, Zhan W, et al. Endothelial progenitor cell derived exosomes mediated miR-182-5p delivery accelerate diabetic wound healing via down-regulating PPARG. Int J Med Sci. 2023;20(4):468–481. doi:10.7150/ijms.78790
98. Xu J, Bai S, Cao Y, et al. miRNA-221-3p in endothelial progenitor cell-derived exosomes accelerates skin wound healing in diabetic mice. Diabetes Metab Syndr Obes. 2020;13:1259–1270. doi:10.2147/DMSO.S243549
99. Abhinand CS, Galipon J, Mori M, et al. Temporal phosphoproteomic analysis of VEGF-A signaling in HUVECs: an insight into early signaling events associated with angiogenesis. J Cell Commun Signal. 2023;17(3):1067–1079. doi:10.1007/s12079-023-00736-z
100. Yuan M, Liu K, Jiang T, et al. GelMA/PEGDA microneedles patch loaded with HUVECs-derived exosomes and Tazarotene promote diabetic wound healing. J Nanobiotechnol. 2022;20(1):147. doi:10.1186/s12951-022-01354-4
101. Hu H, Jiang C, Li R, Zhao J. Comparison of endothelial cell- and endothelial progenitor cell-derived exosomes in promoting vascular endothelial cell repair. Int J Clin Exp Pathol. 2019;12(7):2793–2800.
102. Hsieh C-H, Li L, Vanhauwaert R, et al. Miro1 Marks Parkinson’s disease subset and Miro1 reducer rescues neuron loss in Parkinson’s Models. Cell Metab. 2019;30(6):1131–1140.e7. doi:10.1016/j.cmet.2019.08.023
103. Croft AP, Campos J, Jansen K, et al. Distinct fibroblast subsets drive inflammation and damage in arthritis. Nature. 2019;570(7760):246–251. doi:10.1038/s41586-019-1263-7
104. Kirk T, Ahmed A, Rognoni E. Fibroblast memory in development, homeostasis and disease. Cells. 2021;10(11):2840. doi:10.3390/cells10112840
105. Qin W, Wu Y, Liu J, Yuan X, Gao J. A comprehensive review of the application of nanoparticles in diabetic wound healing: therapeutic potential and future perspectives. Int J Nanomed. 2022;17:6007–6029. doi:10.2147/IJN.S386585
106. Xu Z, Ni T, Zhang Q, et al. Exosomes derived from fibroblasts in DFUs delay wound healing by delivering miR-93-5p to target macrophage ATG16L1. Biochim Biophys Acta Mol Basis Dis. 2025;1871(3):167640. doi:10.1016/j.bbadis.2024.167640
107. Wang L, Chen J, Song J, et al. Activation of the Wnt/β-catenin signalling pathway enhances exosome production by hucMSCs and improves their capability to promote diabetic wound healing. J Nanobiotechnol. 2024;22(1):373. doi:10.1186/s12951-024-02650-x
108. Ahmadpour F, Rasouli HR, Talebi S, Golchin D, Esmailinejad MR, Razie A. Effects of exosomes derived from fibroblast cells on skin wound healing in Wistar rats. Burns. 2023;49(6):1372–1381. doi:10.1016/j.burns.2023.02.003
109. Thulabandu V, Chen D, Atit RP. Dermal fibroblast in cutaneous development and healing. Wiley Interdiscip Rev Dev Biol. 2018;7(2). doi:10.1002/wdev.307
110. Xia W, Li M, Jiang X, et al. Young fibroblast-derived exosomal microRNA-125b transfers beneficial effects on aged cutaneous wound healing. J Nanobiotechnol. 2022;20(1):144. doi:10.1186/s12951-022-01348-2
111. Han M, Yang H, Lu X, et al. Three-dimensional-cultured MSC-derived exosome-hydrogel hybrid microneedle array patch for spinal cord repair. Nano Lett. 2022;22(15):6391–6401. doi:10.1021/acs.nanolett.2c02259
112. Brennan MÁ, Layrolle P, Mooney DJ. Biomaterials functionalized with MSC secreted extracellular vesicles and soluble factors for tissue regeneration. Adv Funct Mater. 2020;30(37). doi:10.1002/adfm.201909125
113. Lei F, Li M, Lin T, Zhou H, Wang F, Su X. Treatment of inflammatory bone loss in periodontitis by stem cell-derived exosomes. Acta Biomater. 2022;141:333–343. doi:10.1016/j.actbio.2021.12.035
114. Hu Y, Wang Z, Fan C, et al. Human gingival mesenchymal stem cell-derived exosomes cross-regulate the Wnt/β-catenin and NF-κB signalling pathways in the periodontal inflammation microenvironment. J Clin Periodontol. 2023;50(6):796–806. doi:10.1111/jcpe.13798
115. Zhang Y, Wang Z, Shi B, et al. Effect of gingival mesenchymal stem cell-derived exosomes on inflammatory macrophages in a high-lipid microenvironment. Int Immunopharmacol. 2021;94:107455. doi:10.1016/j.intimp.2021.107455
116. Kuang L, Zhang C, Li B, Deng H, Chen R, Li G. Human keratinocyte-derived exosomal MALAT1 promotes diabetic wound healing by upregulating MFGE8 via microRNA-1914-3p. Int J Nanomed. 2023;18:949–970. doi:10.2147/IJN.S399785
117. Zhou X, Brown BA, Siegel AP, et al. Exosome-mediated crosstalk between keratinocytes and macrophages in cutaneous wound healing. ACS Nano. 2020;14(10):12732–12748. doi:10.1021/acsnano.0c03064
118. Zhou Z, Zheng J, Lin D, Xu R, Chen Y, Hu X. Exosomes derived from dental pulp stem cells accelerate cutaneous wound healing by enhancing angiogenesis via the Cdc42/p38 MAPK pathway. Int J Mol Med. 2022;50(6). doi:10.3892/ijmm.2022.5199
119. Dong M, Ma X, Li F. Dedifferentiated fat cells-derived exosomes (DFATs-Exos) loaded in GelMA accelerated diabetic wound healing through Wnt/β-catenin pathway. Stem Cell Res Ther. 2025;16(1):103. doi:10.1186/s13287-025-04205-9
120. Wei P, Zhong C, Yang X, et al. Exosomes derived from human amniotic epithelial cells accelerate diabetic wound healing via PI3K-AKT-mTOR-mediated promotion in angiogenesis and fibroblast function. Burns Trauma. 2020;8:tkaa020. doi:10.1093/burnst/tkaa020
121. Liu M, Liu Z, Chen Y, et al. Dendritic epidermal T cells secreting exosomes promote the proliferation of epidermal stem cells to enhance wound re-epithelialization. Stem Cell Res Ther. 2022;13(1):121. doi:10.1186/s13287-022-02783-6
122. Yu Y, Jin H, Li L, et al. An injectable, activated neutrophil-derived exosome mimetics/extracellular matrix hybrid hydrogel with antibacterial activity and wound healing promotion effect for diabetic wound therapy. J Nanobiotechnol. 2023;21(1):308. doi:10.1186/s12951-023-02073-0
123. Zhang M, Guo J, Xiang K, et al. Exosomes derived from oral squamous cell carcinoma tissue accelerate diabetic wound healing. Am J Physiol Cell Physiol. 2023;324(6):C1307–C1319. doi:10.1152/ajpcell.00541.2022
124. Wang H, Wang L, Zhou X, et al. OSCC exosomes regulate miR-210-3p targeting EFNA3 to promote oral cancer angiogenesis through the PI3K/AKT pathway. Biomed Res Int. 2020;2020:2125656. doi:10.1155/2020/2125656
125. Wang X, Jiao Y, Pan Y, et al. Fetal dermal mesenchymal stem cell-derived exosomes accelerate cutaneous wound healing by activating notch signaling. Stem Cells Int. 2019;2019:2402916. doi:10.1155/2019/2402916
126. Duan M, Zhang Y, Zhang H, Meng Y, Qian M, Zhang G. Epidermal stem cell-derived exosomes promote skin regeneration by downregulating transforming growth factor-β1 in wound healing. Stem Cell Res Ther. 2020;11(1):452. doi:10.1186/s13287-020-01971-6
127. Cui HS, Kim DH, Joo SY, Cho YS, Kim J-B, Seo CH. Exosomes derived from human hypertrophic scar fibroblasts induces smad and TAK1 signaling in normal dermal fibroblasts. Arch Biochem Biophys. 2022;722:109215. doi:10.1016/j.abb.2022.109215
128. Zhang G, Wang D, Ren J, et al. Antler stem cell-derived exosomes promote regenerative wound healing via fibroblast-to-myofibroblast transition inhibition. J Biol Eng. 2023;17(1):67. doi:10.1186/s13036-023-00386-0
129. 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(4):555–568. doi:10.1002/term.2799
130. Yang F, Cai D, Kong R, et al. Exosomes derived from cord blood Treg cells promote diabetic wound healing by targeting monocytes. Biochem Pharmacol. 2024;226:116413. doi:10.1016/j.bcp.2024.116413
131. Wang T, Li Y, Hao L, et al. Coriander-derived exosome-like nanovesicles laden hydrogel with antioxidant property accelerates wound healing. Macromol Biosci. 2025:e2400640. doi:10.1002/mabi.202400640
132. Tan M, Liu Y, Xu Y, et al. Plant-derived exosomes as novel nanotherapeutics contrive glycolysis reprogramming-mediated angiogenesis for diabetic ulcer healing. Biomater Res. 2024;28:0035. doi:10.34133/bmr.0035
133. Pirisinu M, Pham TC, Zhang DX, Hong TN, Nguyen LT, Le MT. Extracellular vesicles as natural therapeutic agents and innate drug delivery systems for cancer treatment: recent advances, current obstacles, and challenges for clinical translation. Semin Cancer Biol. 2022;80:340–355. doi:10.1016/j.semcancer.2020.08.007
134. Xiong Y, Chen L, Yu T, et al. Inhibition of circulating exosomal microRNA-15a-3p accelerates diabetic wound repair. Aging. 2020;12(10):8968–8986. doi:10.18632/aging.103143
135. Li R, Wang H, Wang X, et al. MSC-EVs and UCB-EVs promote skin wound healing and spatial transcriptome analysis. Sci Rep. 2025;15(1):4006. doi:10.1038/s41598-025-87592-6
136. Kanta J, Zavadakova A, Sticova E, Dubsky M. Fibronectin in hyperglycaemia and its potential use in the treatment of diabetic foot ulcers: a review. Int Wound J. 2023;20(5):1750–1761. doi:10.1111/iwj.13997
137. Liu K, Gong B, Li T, et al. Bioactive self-healing umbilical cord blood exosomes hydrogel for promoting chronic diabetic wound healing. Biochem Biophys Res Commun. 2024;690:149241. doi:10.1016/j.bbrc.2023.149241
138. Hu Y, Rao -S-S, Wang Z-X, et al. Exosomes from human umbilical cord blood accelerate cutaneous wound healing through miR-21-3p-mediated promotion of angiogenesis and fibroblast function. Theranostics. 2018;8(1):169–184. doi:10.7150/thno.21234
139. Tutanov O, Orlova E, Proskura K, et al. Proteomic analysis of blood exosomes from healthy females and breast cancer patients reveals an association between different exosomal bioactivity on non-tumorigenic epithelial cell and breast cancer cell migration in vitro. Biomolecules. 2020;10(4). doi:10.3390/biom10040495
140. Chen L, Qin L, Chen C, Hu Q, Wang J, Shen J. Serum exosomes accelerate diabetic wound healing by promoting angiogenesis and ECM formation. Cell Biol Int. 2021;45(9):1976–1985. doi:10.1002/cbin.11627
141. He M, Chen T, Lv Y, et al. The role of allogeneic platelet-rich plasma in patients with diabetic foot ulcer: current perspectives and future challenges. Front Bioeng Biotechnol. 2022;10:993436. doi:10.3389/fbioe.2022.993436
142. He M, Guo X, Li T, et al. Comparison of allogeneic platelet-rich plasma with autologous platelet-rich plasma for the treatment of diabetic lower extremity ulcers. Cell Transplant. 2020;29:963689720931428. doi:10.1177/0963689720931428
143. Li T, Ma Y, Wang M, et al. Platelet-rich plasma plays an antibacterial, anti-inflammatory and cell proliferation-promoting role in an in vitro model for diabetic infected wounds. Infect Drug Resist. 2019;12:297–309. doi:10.2147/IDR.S186651
144. Chen T, Song P, He M, et al. Sphingosine-1-phosphate derived from PRP-Exos promotes angiogenesis in diabetic wound healing via the S1PR1/AKT/FN1 signalling pathway. Burns Trauma. 2023;11:tkad003. doi:10.1093/burnst/tkad003
145. Rui S, Dai L, Zhang X, et al. Exosomal miRNA-26b-5p from PRP suppresses NETs by targeting MMP-8 to promote diabetic wound healing. J Control Release. 2024;372:221–233. doi:10.1016/j.jconrel.2024.06.050
146. Rui S, Yuan Y, Du C, et al. Comparison and investigation of exosomes derived from platelet-rich plasma activated by different agonists. Cell Transplant. 2021;30:9636897211017833. doi:10.1177/09636897211017833
147. Sun Y, Zhao H, Yang S, et al. Urine-derived stem cells: promising advancements and applications in regenerative medicine and beyond. Heliyon. 2024;10(6):e27306. doi:10.1016/j.heliyon.2024.e27306
148. Yu P, Bosholm CC, Zhu H, Duan Z, Atala A, Zhang Y. Beyond waste: understanding urine’s potential in precision medicine. Trends Biotechnol. 2024;42(8):953–969. doi:10.1016/j.tibtech.2024.01.009
149. Zhou Q, Cheng Y, Sun F, et al. A comprehensive review of the therapeutic value of urine-derived stem cells. Front Genet. 2021;12:781597. doi:10.3389/fgene.2021.781597
150. Wang Y, Zhang M. Urinary exosomes: a promising biomarker for disease diagnosis. Lab Med. 2023;54(2):115–125. doi:10.1093/labmed/lmac087
151. Chen L, Li L, Xing F, et al. Human urine-derived stem cells: potential for cell-based therapy of cartilage defects. Stem Cells Int. 2018;2018:4686259. doi:10.1155/2018/4686259
152. Chen C-Y, Rao -S-S, Ren L, et al. Exosomal DMBT1 from human urine-derived stem cells facilitates diabetic wound repair by promoting angiogenesis. Theranostics. 2018;8(6):1607–1623. doi:10.7150/thno.22958
153. Tong X, Xu Y, Zhang T, et al. Exosomes from CD133+ human urine-derived stem cells combined adhesive hydrogel facilitate rotator cuff healing by mediating bone marrow mesenchymal stem cells. J Orthop Transl. 2023;39:100–112. doi:10.1016/j.jot.2023.02.002
154. Qian G, Yu Y, Dong Y, Hong Y, Wang M. Exosomes derived from human urine-derived stem cells ameliorate IL-1β-induced intervertebral disk degeneration. BMC Musculoskelet Disord. 2024;25(1):537. doi:10.1186/s12891-024-07636-2
155. Hu C, Sun Y, Li W, Bi Y. Hypoxia improves self-renew and migration of urine-derived stem cells by upregulating autophagy and mitochondrial function through ERK signal pathway. Mitochondrion. 2023;73:1–9. doi:10.1016/j.mito.2023.09.001
156. Torres P, Díaz J, Arce M, et al. The salivary peptide histatin-1 promotes endothelial cell adhesion, migration, and angiogenesis. FASEB J. 2017;31(11):4946–4958. doi:10.1096/fj.201700085R
157. Liu A, Hefley B, Escandon P, Nicholas SE, Karamichos D. Salivary exosomes in health and disease: future prospects in the eye. Int J Mol Sci. 2023;24(7). doi:10.3390/ijms24076363
158. Mi B, Chen L, Xiong Y, et al. Saliva exosomes-derived UBE2O mRNA promotes angiogenesis in cutaneous wounds by targeting SMAD6. J Nanobiotechnol. 2020;18(1):68. doi:10.1186/s12951-020-00624-3
159. Ullah K, Zubia E, Narayan M, Yang J, Xu G. Diverse roles of the E2/E3 hybrid enzyme UBE2O in the regulation of protein ubiquitination, cellular functions, and disease onset. FEBS J. 2019;286(11):2018–2034. doi:10.1111/febs.14708
160. Freiría-Martínez L, Iglesias-Martínez-Almeida M, Rodríguez-Jamardo C, et al. Human breast milk microRNAs, potential players in the regulation of nervous system. Nutrients. 2023;15(14). doi:10.3390/nu15143284
161. Kandimalla R, Aqil F, Tyagi N, Gupta R. Milk exosomes: a biogenic nanocarrier for small molecules and macromolecules to combat cancer. Am J Reprod Immunol. 2021;85(2):e13349. doi:10.1111/aji.13349
162. Munagala R, Aqil F, Jeyabalan J, Gupta RC. Bovine milk-derived exosomes for drug delivery. Cancer Letters. 2016;371(1):48–61. doi:10.1016/j.canlet.2015.10.020
163. Sedykh S, Kuleshova A, Nevinsky G. Milk exosomes: perspective agents for anticancer drug delivery. Int J Mol Sci. 2020;21(18). doi:10.3390/ijms21186646
164. Betker JL, Angle BM, Graner MW, Anchordoquy TJ. The potential of exosomes from cow milk for oral delivery. J Pharm Sci. 2019;108(4):1496–1505. doi:10.1016/j.xphs.2018.11.022
165. Yan C, Chen J, Wang C, et al. Milk exosomes-mediated miR-31-5p delivery accelerates diabetic wound healing through promoting angiogenesis. Drug Delivery. 2022;29(1):214–228. doi:10.1080/10717544.2021.2023699
166. Xiang X, Chen J, Jiang T, et al. Milk-derived exosomes carrying siRNA-KEAP1 promote diabetic wound healing by improving oxidative stress. Drug Deliv Transl Res. 2023;13(9):2286–2296. doi:10.1007/s13346-023-01306-x
167. Sheller-Miller S, Menon R. Isolation and characterization of human amniotic fluid-derived exosomes. Methods Enzymol. 2020;645:181–194. doi:10.1016/bs.mie.2020.07.006
168. Ram Kumar RM, Logesh R, Joghee S. Breast cancer derived exosomes: theragnostic perspectives and implications. Clin Chim Acta. 2024;557:117875. doi:10.1016/j.cca.2024.117875
169. Menon R, Debnath C, Lai A, et al. Protein profile changes in circulating placental extracellular vesicles in term and preterm births: a longitudinal study. Endocrinology. 2020;161(4). doi:10.1210/endocr/bqaa009
170. Keshtkar S, Azarpira N, Ghahremani MH. Mesenchymal stem cell-derived extracellular vesicles: novel frontiers in regenerative medicine. Stem Cell Res Ther. 2018;9(1):63. doi:10.1186/s13287-018-0791-7
171. Castelli V, Antonucci I, d’Angelo M, et al. Neuroprotective effects of human amniotic fluid stem cells-derived secretome in an ischemia/reperfusion model. Stem Cells Transl Med. 2021;10(2):251–266. doi:10.1002/sctm.20-0268
172. Jiang Y, Li Y, Duan L, Jiang B. Amniotic fluid-derived stem cells: an overlooked source of stem cells for translational research. DNA Cell Biol. 2025;44(3):144–152. doi:10.1089/dna.2024.0254
173. Ochiai D, Abe Y, Fukutake M, et al. Cell sheets using human amniotic fluid stem cells reduce tissue fibrosis in murine full-thickness skin wounds. Tissue Cell. 2021;68:101472. doi:10.1016/j.tice.2020.101472
174. Zhang Y, Yan J, Liu Y, et al. Human amniotic fluid stem cell-derived exosomes as a novel cell-free therapy for cutaneous regeneration. Front Cell Develop Biol. 2021;9:685873. doi:10.3389/fcell.2021.685873
175. Luo H, Wang Z, Qi F, Wang D. Applications of human amniotic fluid stem cells in wound healing. Chinese Med J. 2022;135(19):2272–2281. doi:10.1097/CM9.0000000000002076
176. Maitra S, Sarkar S, Dhara B. Plant‐derived exosomes: a new frontier in nano‐medicine for cancer and microbial infection therapy. Clin Transl Discov. 2024;4(4):e342.
177. Liu B, Lu Y, Chen X, et al. Protective role of Shiitake mushroom-derived exosome-like nanoparticles in D-galactosamine and lipopolysaccharide-induced acute liver injury in mice. Nutrients. 2020;12(2). doi:10.3390/nu12020477
178. Wang Y, Wang J, Ma J, Zhou Y, Lu R. Focusing on future applications and current challenges of plant derived extracellular vesicles. Pharmaceuticals. 2022;15(6). doi:10.3390/ph15060708
179. Huang X, Wang H, Wang C, Cao Z. The applications and potentials of extracellular vesicles from different cell sources in periodontal regeneration. Int J Mol Sci. 2023;24(6). doi:10.3390/ijms24065790
180. Liao Q, Su L, Pang L, et al. Natural exosome-like nanoparticles derived from ancient medicinal insect Periplaneta americana L. as a novel diabetic wound healing accelerator. J Nanobiotechnol. 2023;21(1):169. doi:10.1186/s12951-023-01923-1
181. Feng H, Yue Y, Zhang Y, et al. Plant-derived exosome-like nanoparticles: emerging nanosystems for enhanced tissue engineering. Int J Nanomed. 2024;19:1189–1204. doi:10.2147/IJN.S448905
182. Şahin F, Koçak P, Güneş MY, Özkan İ, Yıldırım E, Kala EY. In vitro wound healing activity of wheat-derived nanovesicles. Appl Biochem Biotechnol. 2019;188(2):381–394. doi:10.1007/s12010-018-2913-1
183. Pomatto MAC, Gai C, Negro F, et al. Plant-derived extracellular vesicles as a delivery platform for RNA-based vaccine: feasibility study of an oral and intranasal SARS-CoV-2 vaccine. Pharmaceutics. 2023;15(3). doi:10.3390/pharmaceutics15030974
184. Urzì O, Cafora M, Ganji NR, et al. Lemon-derived nanovesicles achieve antioxidant and anti-inflammatory effects activating the AhR/Nrf2 signaling pathway. iScience. 2023;26(7):107041. doi:10.1016/j.isci.2023.107041
185. El-Refaiy AI, Salem ZA, Badawy AA, Dahran N, Desouky MA, El-Magd MA. Protective effects of lemon and Orange peels and olive oil on doxorubicin-induced myocardial damage via inhibition of oxidative stress and inflammation pathways. Front Pharmacol. 2025;16:1506673. doi:10.3389/fphar.2025.1506673
186. Bruno SP, Paolini A, D’Oria V, et al. Extracellular vesicles derived from citrus sinensis modulate inflammatory genes and tight junctions in a human model of intestinal epithelium. Front Nutr. 2021;8:778998. doi:10.3389/fnut.2021.778998
187. Xiao J, Feng S, Wang X, et al. Identification of exosome-like nanoparticle-derived microRNAs from 11 edible fruits and vegetables. PeerJ. 2018:
188. Trentini M, Zanotti F, Tiengo E, et al. An apple a day keeps the doctor away: potential role of miRNA 146 on macrophages treated with exosomes derived from apples. Biomedicines. 2022;10(2). doi:10.3390/biomedicines10020415
189. Deng Z, Rong Y, Teng Y, et al. Broccoli-derived nanoparticle inhibits mouse colitis by activating dendritic cell AMP-activated protein kinase. Mol Ther. 2017;25(7):1641–1654. doi:10.1016/j.ymthe.2017.01.025
190. Sundaram K, Mu J, Kumar A, et al. Garlic exosome-like nanoparticles reverse high-fat diet induced obesity via the gut/brain axis. Theranostics. 2022;12(3):1220–1246. doi:10.7150/thno.65427
191. Sundaram K, Teng Y, Mu J, et al. Outer membrane vesicles released from garlic exosome-like nanoparticles (GaELNs) train gut bacteria that reverses type 2 diabetes via the gut-brain axis. Small. 2024;20(20):e2308680. doi:10.1002/smll.202308680
192. Lee Y, Jeong D-Y, Jeun YC, Choe H, Yang S. Preventive and ameliorative effects of potato exosomes on UVB‑induced photodamage in keratinocyte HaCaT cells. Mol Med Rep. 2023;28(3). doi:10.3892/mmr.2023.13054
193. Wang Q, Liu K, Cao X, et al. Plant-derived exosomes extracted from Lycium barbarum L. loaded with isoliquiritigenin to promote spinal cord injury repair based on 3D printed bionic scaffold. Bioeng Transl Med. 2024;9(4):e10646. doi:10.1002/btm2.10646
194. Tu J, Jiang F, Fang J, et al. Anticipation and verification of dendrobium-derived nanovesicles for skin wound healing targets, predicated upon immune infiltration and senescence. Int J Nanomed. 2024;19:1629–1644. doi:10.2147/IJN.S438398
195. Wu J, Ma X, Lu Y, et al. Edible Pueraria lobata-derived exosomes promote M2 macrophage polarization. Molecules. 2022;27(23). doi:10.3390/molecules27238184
196. Tan S, Liu Z, Cong M, et al. Dandelion-derived vesicles-laden hydrogel dressings capable of neutralizing Staphylococcus aureus exotoxins for the care of invasive wounds. J Control Release. 2024;368:355–371. doi:10.1016/j.jconrel.2024.02.045
197. Yang S, Lu S, Ren L, et al. Ginseng-derived nanoparticles induce skin cell proliferation and promote wound healing. J Ginseng Res. 2023;47(1):133–143. doi:10.1016/j.jgr.2022.07.005
198. Lu S-Y, Yang S, Ren L-M, Wang J-W, Zhao D-Q. Panax ginseng exosomes promote HaCat cell proliferation and wound healing. Chin J Biochem Mol Biol. 2021;37(11):1510–1519.
199. Xu X-H, Yuan T-J, Dad HA, et al. Plant exosomes as novel nanoplatforms for microRNA transfer stimulate neural differentiation of stem cells in vitro and in vivo. Nano Lett. 2021;21(19):8151–8159. doi:10.1021/acs.nanolett.1c02530
200. Xiong Y, Lin Z, Bu P, et al. A whole-course-repair system based on neurogenesis-angiogenesis crosstalk and macrophage reprogramming promotes diabetic wound healing. Adv Mater. 2023;35(19):e2212300. doi:10.1002/adma.202212300
201. Kim MK, Choi YC, Cho SH, Choi JS, Cho YW. The antioxidant effect of small extracellular vesicles derived from aloe vera peels for wound healing. Tissue Eng Regen Med. 2021;18(4):561–571. doi:10.1007/s13770-021-00367-8
202. Kim M, Park JH. Isolation of aloe saponaria-derived extracellular vesicles and investigation of their potential for chronic wound healing. Pharmaceutics. 2022;14(9). doi:10.3390/pharmaceutics14091905
203. Ramírez O, Pomareda F, Olivares B, et al. Aloe vera peel-derived nanovesicles display anti-inflammatory properties and prevent myofibroblast differentiation. Phytomedicine. 2024;122:155108. doi:10.1016/j.phymed.2023.155108
204. Kantarcıoğlu M, Yıldırım G, Akpınar Oktar P, et al. Coffee-derived exosome-like nanoparticles: are they the secret heroes? Turk J Gastroenterol. 2023;34(2):161–169. doi:10.5152/tjg.2022.21895
205. Dutta S, Ghosh S, Rahaman M, Chowdhury SR. Plant-derived exosomes: pioneering breakthroughs in therapeutics, targeted drug delivery, and regenerative medicine. Pharm Nanotechnol. 2024. doi:10.2174/0122117385305245240424093014
206. Savcı Y, Kırbaş OK, Bozkurt BT, et al. Grapefruit-derived extracellular vesicles as a promising cell-free therapeutic tool for wound healing. Food Funct. 2021;12(11):5144–5156. doi:10.1039/d0fo02953j
207. Wu W, Zhang B, Wang W, et al. Plant-derived exosome-like nanovesicles in chronic wound healing. Int J Nanomed. 2024;19:11293–11303. doi:10.2147/IJN.S485441
208. Perut F, Roncuzzi L, Avnet S, et al. Strawberry-derived exosome-like nanoparticles prevent oxidative stress in human mesenchymal stromal cells. Biomolecules. 2021;11(1). doi:10.3390/biom11010087
209. Wei C, Zhang M, Cheng J, Tian J, Yang G, Jin Y. Plant-derived exosome-like nanoparticles - from Laboratory to factory, a landscape of application, challenges and prospects. Crit Rev Food Sci Nutr. 2024. doi:10.1080/10408398.2024.2388888
210. Chen Y-X, Cai Q. Plant exosome-like nanovesicles and their role in the innovative delivery of RNA therapeutics. Biomedicines. 2023;11(7). doi:10.3390/biomedicines11071806
211. Li Y, Wang Y, Zhao H, Pan Q, Chen G. Engineering strategies of plant-derived exosome-like nanovesicles: current knowledge and future perspectives. Int J Nanomed. 2024;19:12793–12815. doi:10.2147/IJN.S496664
212. Gurung S, Perocheau D, Touramanidou L, Baruteau J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal. 2021;19(1):47. doi:10.1186/s12964-021-00730-1
213. Lian MQ, Chng WH, Liang J, et al. Plant-derived extracellular vesicles: recent advancements and current challenges on their use for biomedical applications. J Extracell Vesicles. 2022;11(12):e12283. doi:10.1002/jev2.12283
214. Zhang L, Yu D. Exosomes in cancer development, metastasis, and immunity. Biochim Biophys Acta Rev Cancer. 2019;1871(2):455–468. doi:10.1016/j.bbcan.2019.04.004
215. Cong M, Tan S, Li S, et al. Technology insight: plant-derived vesicles-How far from the clinical biotherapeutics and therapeutic drug carriers? Adv Drug Deliv Rev. 2022;182:114108. doi:10.1016/j.addr.2021.114108
216. Zhao Y, Tan H, Zhang J, et al. Plant-derived vesicles: a new era for anti-cancer drug delivery and cancer treatment. Int J Nanomed. 2023;18:6847–6868. doi:10.2147/IJN.S432279
217. Feng J, Xiu Q, Huang Y, Troyer Z, Li B, Zheng L. Plant-derived vesicle-like nanoparticles as promising biotherapeutic tools: present and future. Adv Mater. 2023;35(24):e2207826. doi:10.1002/adma.202207826
218. Dad HA, Gu T-W, Zhu A-Q, Huang L-Q, Peng L-H. Plant exosome-like nanovesicles: emerging therapeutics and drug delivery nanoplatforms. Mol Ther. 2021;29(1):13–31. doi:10.1016/j.ymthe.2020.11.030
219. O’Brien K, Breyne K, Ughetto S, Laurent LC, Breakefield XO. RNA delivery by extracellular vesicles in mammalian cells and its applications. Nat Rev Mol Cell Biol. 2020;21(10):585–606. doi:10.1038/s41580-020-0251-y
220. Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol. 2021;16(7):748–759. doi:10.1038/s41565-021-00931-2
221. Mu N, Li J, Zeng L, et al. Plant-derived exosome-like nanovesicles: current progress and prospects. Int J Nanomed. 2023;18:4987–5009. doi:10.2147/IJN.S420748
222. Li Z, Wang H, Yin H, Bennett C, Zhang H-G, Guo P. Arrowtail RNA for ligand display on ginger exosome-like nanovesicles to systemic deliver siRNA for cancer suppression. Sci Rep. 2018;8(1):14644. doi:10.1038/s41598-018-32953-7
223. Langellotto MD, Rassu G, Serri C, Demartis S, Giunchedi P, Gavini E. Plant-derived extracellular vesicles: a synergetic combination of a drug delivery system and a source of natural bioactive compounds. Drug Deliv Transl Res. 2025;15(3):831–845. doi:10.1007/s13346-024-01698-4
224. Choi H, Choi Y, Yim HY, Mirzaaghasi A, Yoo J-K, Choi C. Biodistribution of exosomes and engineering strategies for targeted delivery of therapeutic exosomes. Tissue Eng Regen Med. 2021;18(4):499–511. doi:10.1007/s13770-021-00361-0
225. Kimiz-Gebologlu I, Oncel SS. Exosomes: large-scale production, isolation, drug loading efficiency, and biodistribution and uptake. J Control Release. 2022;347:533–543. doi:10.1016/j.jconrel.2022.05.027
226. Chen J, Li P, Zhang T, et al. Review on strategies and technologies for exosome isolation and purification. Front Bioeng Biotechnol. 2021;9:811971. doi:10.3389/fbioe.2021.811971
227. Qin X, He J, Wang X, Wang J, Yang R, Chen X. The functions and clinical application potential of exosomes derived from mesenchymal stem cells on wound repair: a review of recent research advances. Front Immunol. 2023;14:1256687. doi:10.3389/fimmu.2023.1256687
228. Bailey AJM, Li H, Kirkham AM, et al. MSC-derived extracellular vesicles to heal diabetic wounds: a systematic review and meta-analysis of preclinical animal studies. Stem Cell Rev Rep. 2022;18(3):968–979. doi:10.1007/s12015-021-10164-4
229. Xie H, Wang Z, Wang R, Chen Q, Yu A, Lu A. Self‐healing, injectable hydrogel dressing for monitoring and therapy of diabetic wound. Adv Funct Mater. 2024;34(36):2401209.
230. Duan W, Jin X, Zhao Y, et al. Engineering injectable hyaluronic acid-based adhesive hydrogels with anchored PRP to pattern the micro-environment to accelerate diabetic wound healing. Carbohydr Polym. 2024;337:122146. doi:10.1016/j.carbpol.2024.122146
231. Liu Y, Jia D, Li L, Wang M. Advances in nanomedicine and biomaterials for endometrial regeneration: a comprehensive review. Int J Nanomed. 2024;19:8285–8308. doi:10.2147/IJN.S473259
232. Zhao X, Fu L, Zou H, et al. Optogenetic engineered umbilical cord MSC-derived exosomes for remodeling of the immune microenvironment in diabetic wounds and the promotion of tissue repair. J Nanobiotechnol. 2023;21(1):176. doi:10.1186/s12951-023-01886-3
233. Gupta NS, Kumar P. Perspective of artificial intelligence in healthcare data management: a journey towards precision medicine. Comput Biol Med. 2023;162:107051. doi:10.1016/j.compbiomed.2023.107051
© 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.
Recommended articles

Long-Term Effects of Severe Burns on the Kidneys: Research Advances and Potential Therapeutic Approaches
Yang G, Tan L, Yao H, Xiong Z, Wu J, Huang X
Journal of Inflammation Research 2023, 16:1905-1921
Published Date: 1 May 2023
Accelerated Wound Healing in Diabetic Rat by miRNA-185-5p and Its Anti-Inflammatory Activity
Wang KX, Zhao LL, Zheng LT, Meng LB, Jin L, Zhang LJ, Kong FL, Liang F
Diabetes, Metabolic Syndrome and Obesity 2023, 16:1657-1667
Published Date: 7 June 2023
MicroRNA-221-3p Targets THBS1 to Promote Wound Healing in Diabetes
Hu K, Liu X, Chang H, Zhang Y, Zhou H, Liu L, Zhang X, Jiao Z, Shen B, Zhang Q
Diabetes, Metabolic Syndrome and Obesity 2023, 16:2765-2777
Published Date: 11 September 2023

Effect of Diabetes on Wound Healing: A Bibliometrics and Visual Analysis
Lang X, Li L, Li Y, Feng X
Journal of Multidisciplinary Healthcare 2024, 17:1275-1289
Published Date: 20 March 2024
Advances in Nanomedicine and Biomaterials for Endometrial Regeneration: A Comprehensive Review
Liu Y, Jia D, Li L, Wang M
International Journal of Nanomedicine 2024, 19:8285-8308
Published Date: 14 August 2024