Skip to main content
Heliyon logoLink to Heliyon
. 2024 Aug 29;10(17):e37031. doi: 10.1016/j.heliyon.2024.e37031

Advancements in diabetic foot ulcer research: Focus on mesenchymal stem cells and their exosomes

ShuHui Wu 1,1, ZhongSheng Zhou 1,1, Yang Li 1, Jinlan Jiang 1,
PMCID: PMC11403009  PMID: 39286219

Abstract

Diabetes represents a widely acknowledged global public health concern. Diabetic foot ulcer (DFU) stands as one of the most severe complications of diabetes, its occurrence imposing a substantial economic burden on patients, profoundly impacting their quality of life. Despite the deepening comprehension regarding the pathophysiology and cellular as well as molecular responses of DFU, the current therapeutic arsenal falls short of efficacy, failing to offer a comprehensive remedy for deep-seated chronic wounds and microvascular occlusions. Conventional treatments merely afford symptomatic alleviation or retard the disease's advancement, devoid of the capacity to effectuate further restitution of compromised vasculature and nerves. An escalating body of research underscores the prominence of mesenchymal stem cells (MSCs) owing to their paracrine attributes and anti-inflammatory prowess, rendering them a focal point in the realm of chronic wound healing. Presently, MSCs have been validated as a highly promising cellular therapeutic approach for DFU, capable of effectuating cellular repair, epithelialization, granulation tissue formation, and neovascularization by means of targeted differentiation, angiogenesis promotion, immunomodulation, and paracrine activities, thereby fostering wound healing. The secretome of MSCs comprises cytokines, growth factors, chemokines, alongside exosomes harboring mRNA, proteins, and microRNAs, possessing immunomodulatory and regenerative properties. The present study provides a systematic exposition on the etiology of DFU and elucidates the intricate molecular mechanisms and diverse functionalities of MSCs in the context of DFU treatment, thereby furnishing pioneering perspectives aimed at harnessing the therapeutic potential of MSCs for DFU management and advancing wound healing processes.

Keywords: Diabetic foot ulcers, Mesenchymal stem cells, Exosomes

Graphical abstract

Image 1

Highlights

  • Comprehensive Elucidation of Mesenchymal Stem Cells and Their Secreted Extracellular Vesicles.

  • Exploring the Intricate Molecular Mechanisms by Which Mesenchymal Stem Cells Facilitate Diabetic Foot Ulcer Healing.

  • Summarizing Key RNAs Secreted in Extracellular Vesicles by Mesenchymal Stem Cells that Promote Wound Healing。.

Funding

This work was supported by the National Natural Science Foundation of China (Grant No.8217081220), the Jilin Scientific and Technological Development Program (Grant No. 20240205001YY), the Changchun Science and Technology Development Plan Project (Grant No. 21ZGY29)

1. Introduction

Diabetes manifests as a metabolic disorder marked by diminished pancreatic function and insulin resistance, culminating in hyperglycemia, attributable to the intricate interplay of immune, genetic, psychological, and environmental factors. Moreover, individuals afflicted with diabetes present with the hallmark clinical manifestations coined as "polyuria, polydipsia, polyphagia, and weight loss. Diabetes is typically categorized into the following four types: type 1 diabetes (insulin-dependent diabetes), type 2 diabetes (non-insulin-dependent diabetes), other distinct variants of diabetes, and gestational diabetes [1]. Type 1 diabetes mellitus typically manifests in childhood or adolescence due to an autoimmune-mediated destruction of pancreatic β-cells, resulting in insufficient or completely absent insulin production. Consequently, patients must rely on exogenous insulin to regulate blood glucose levels. In contrast, Type 2 diabetes mellitus, the most prevalent form of diabetes, generally develops in adulthood. It is characterized by a dual pathophysiology of insulin resistance coupled with inadequate insulin secretion, leading to hyperglycemia. Other distinct forms of diabetes are attributable to specific etiologies, commonly encompassing genetic defects, drug or chemical-induced diabetes, and diabetes secondary to endocrine disorders. Gestational diabetes mellitus (GDM) is defined as glucose intolerance first identified during pregnancy. Although blood glucose levels usually normalize postpartum, these patients are at an elevated risk for progressing to Type 2 diabetes mellitus later in life. Presently, as a result of enhancements in quality of life and the exacerbation of the aging population conundrum, the global incidence of diabetes exhibits an annual upward trajectory. The prognostications from the International Diabetes Federation project a surge in the global diabetic populace to 643 million (constituting 11.3 % of the populace) by 2030, and a subsequent escalation to 783 million by 2045 (comprising 12.2 % of the populace) [2]. Over recent years, there has been a discernible trend towards a gradual reduction in the age of diabetes mellitus onset, concomitant with the protracted course of the condition and its myriad complications, which have exerted an augmented impact on both the health status and economic outlays of patients.

Owing to diabetes being characterized as a systemic metabolic disorder, as the disease advances, patients endure prolonged hyperglycemia, disrupting not only the internal milieu's equilibrium but also instigating chronic impairment and dysfunction across diverse bodily tissues, culminating in irreversible damage to assorted organs and tissues, consequently precipitating a myriad of complications. Common complications associated with diabetes encompass diabetic nephropathy, diabetic retinopathy, diabetic foot ulcers, cardiovascular disease, neuropathy, and cerebrovascular complications. As the disease progresses, sustained hyperglycemia exacerbates non-enzymatic glycation, leading to the accumulation of advanced glycation end-products (AGEs) that are detrimental to vascular endothelial cells and adjacent tissues. Simultaneously, elevated blood glucose levels contribute to increased oxidative stress, resulting in the generation of excessive free radicals that inflict damage on endothelial cells. Moreover, chronic inflammation is frequently observed in diabetic patients, which further compromises endothelial function and accelerates the onset of microvascular pathology, thereby contributing to the development of chronic wounds [3,4]. Among these, diabetic foot ulcers(DFU) represent one of the most prevalent complications. DFU primarily ensues from ischemic, neuropathic, or a confluence of ischemic-neuropathic aberrations [5]. Failure to promptly mitigate the condition post-incidence can instigate limb necrosis, culminating in a heightened disability quotient [6]. Studies elucidate that roughly 25 % of individuals afflicted with diabetes will progress to diabetic foot ulcers, with an additional 30 % of diabetic foot ulcer patients destined for amputation following disease advancement [7]. The malady is distinguished by its protracted trajectory, intricacy, dismal prognosis, exerting a profound toll on the quality of life of afflicted individuals, and exacting a substantial economic burden on both societal and individual levels [8]. Presently, conventional therapeutic modalities in clinical settings encompass blood glucose regulation, wound dressings alterations, hyperbaric oxygen therapy, negative pressure wound therapy, total contact casting, wound debridement, tissue grafting, among others [9]. However, Numerous constraints beset these therapeutic approaches, meriting them capable solely of mitigating patient symptoms or retarding disease advancement, devoid of any reparative effect on impaired blood vessels and nerves, thus perpetuating the recalcitrance of wound healing.

MSCs represent a cadre of cells distinguished by their remarkable secretion, immunomodulatory, and homing attributes. These cells can be derived from a plethora of tissues, encompassing bone marrow, umbilical cord, adipose tissue, amniotic fluid, placenta, menstrual blood, and beyond. Owing to their capacity to generate a plethora of biologically active substances including growth factors, cytokines, chemokines, and beyond, coupled with their immunomodulatory prowess and aptitude for differentiation into specialized tissue cells, these cells wield significant influence in stimulating vascular regeneration, mitigating inflammation, engaging in extracellular matrix remodeling, fostering wound healing, and beyond [10]. Through paracrine action, MSCs have the capacity to liberate exosomes laden with proteins, miRNA, long non-coding RNA(lncRNA), and other bioactive molecules. The secretome originating from MSCs is presently deemed the principal mechanism through which they mitigate DFU. Consequently, MSCs and their exosomes have garnered frequent utilization in investigations pertaining to diabetic wound healing. The present paper systematically expounds upon the role and mechanism of action of MSCs and their exosomes in DFU healing.

2. Pathophysiology of DFU

Upon the onset of diabetes, the organism experiences a state of metabolic disruption. The presence of a hyperglycemic and inflammatory microenvironment results in endothelial injury, microvascular hemorrhage, leakage, and microthrombus formation. Furthermore, it induces the accumulation of Advanced Glycation End-products (AGEs) within the body [11]. AGEs constitute a category of highly active end products resulting from the non-enzymatic glycation of proteins, fatty acids, and nucleic acids within the organism, involving their amino and reducing sugar residues, with a profound correlation observed between their generation and the myriad complications of diabetes [12]. The accrual of AGEs within the organism manifests a potent deleterious impact on neural tissues. The activation of specific transcription factors by AGEs escalates the organism's inflammatory cascade and oxidative stress levels, eliciting consequential functional derangements in tissue cells. This, in turn, suppresses monocyte-macrophage functionality and diminishes cytokine secretion levels, thereby compromising vascular homeostasis, culminating in the manifestation of skin thinning in diabetic patients [13]. The thinning of the skin at specific regions renders the skin's protective barrier increasingly vulnerable to breach, thereby exposing the underlying deep tissues. Various infectious bacteria and viruses are more likely to invade the deeper tissues and further multiply and grow, eventually causing infection and suppuration of the wound. Moreover, as the condition advances, the wound exhibits gradual enlargement and ulceration [14]. Moreover, the inflammatory microenvironment in diabetic wounds obstructs the progression of wound healing during the proliferative phase and disrupts crucial processes such as angiogenesis, extracellular matrix (ECM) remodeling, and re-epithelialization [15].

In addition to disruptions in glucose metabolism, metabolic dysregulation in the organism extends to abnormalities in lipid metabolism. The disorder in lipid metabolism facilitates the release of inflammatory mediators, consequently provoking infiltration by immune cells, predominantly macrophages [16]. Stimulated by elevated levels of blood glucose and blood lipids, the organism undergoes further release of inflammatory mediators, thereby perpetuating a state of heightened inflammation within the organism. In addition to the systemic increase in pro-inflammatory cytokines, diabetic skin also exhibits elevated concentrations of pro-inflammatory cytokines and their receptors [17]. Concurrently, diabetic patients experience a decrease in the phagocytic ability of immune cells, including white blood cells [18]. Within the region of the wound, there is a sustained elevation in local inflammation factor levels, concurrent with a reduction in white blood cell activity. This phenomenon imposes a significant inhibitory effect on the functionality of vascular endothelial cells and fibroblasts, thereby hampering the formation of granulation tissue. Owing to the persistent stimulation by high glucose levels, the organism experiences heightened levels of oxidative stress, culminating in an increase in oxygen species (ROS) expression [19]. This, in turn, diminishes the organism's antioxidant capacity, thereby exerting inhibitory effects on the release of cytokines and growth factors. Concurrently, the activity of fibroblasts, the synthesis of collagen fibers, and the formation of new blood vessels are subject to varying degrees of influence. Consequently, this culminates in capillary constriction, thereby exacerbating microcirculatory disturbances. In the protracted presence of high glucose stimuli, the nervous system is subject to significant impact [20], predominantly characterized by the degeneration of peripheral nerve axons and nerve membrane cells. Consequently, this suppresses the functionality of the organism's motor, sensory, and autonomic nervous systems, thereby impeding limb perfusion and culminating in muscle atrophy, as well as tendon and ligament sclerosis [21]. Studies have shown that in skin injuries of diabetic rabbits and mice induced by alloxan and streptozotocin, the expression of Substance P and other neuropeptides released by nociceptive C-fibers is reduced. This may result in decreased leukocyte recruitment following injury [22]. Additionally, studies have found that the skin of streptozotocin-induced diabetic mice exhibits mast cell degranulation, which correlates with a diminished ability to mount an inflammatory response to tissue injury [23]. With further progression of the disease, foot deformities may manifest. Owing to the buildup of metabolic byproducts within the organism, insufficient blood and oxygen delivery to the surrounding wound area, compounded by factors such as prolonged exposure to high glucose environments and inflammatory responses, diabetic wounds manifest features indicative of impaired healing. In the event of infection, it can instigate the exacerbation of the local wound, gradually evolving into necrosis [24].

The formation and progressive deterioration of DFU are intricately linked with numerous pathophysiological processes, which have the potential to interact, interconvert, and even synergistically compound each other, thereby underscoring the treatment of DFU as one of the pressing and pivotal issues that needs immediate attention [25].(Fig. 1).

Fig. 1.

Fig. 1

Schematic pathophysiology of DFUs.

The investigation into the mechanisms behind DFU formation motivates us to further explore the interplay between the local microenvironment of diabetes and the process of vascular formation. This exploration helps in offering more scientifically grounded and efficient avenues for clinical treatment, while also elucidating the root causes behind the delayed healing of diabetic wounds.

3. Mechanisms underlying wound healing

The process of wound healing encompasses four dynamic stages: hemostasis, inflammation, proliferation, and remodeling [26]. Upon injury, the body promptly initiates hemostasis to halt bleeding. This intricate process entails a myriad of biological responses [27](Fig. 2). Initially, there is reactive constriction of small blood vessels and capillaries surrounding the wound, leading to reduced local blood flow, thereby preventing excessive bleeding [28]. Moreover, exposure of endothelial cells prompts the aggregation of nearby platelets, drawn by collagen fibers and tissue factors, culminating in the formation of a blood clot to avert further blood loss. The blood clot comprises cross-linked fibrin, activated factor XIII, and fibrinogen, with fibrin acting as a provisional extracellular matrix (ECM) [29]. Recruited platelets release adhesive proteins such as serotonin, thromboxane A2, fibrinogen, and fibrinogen-binding protein. Under the influence of adhesive proteins and local clotting factors, additional platelet aggregation occurs, leading to platelet plug formation [30]. Through the action of thrombin, fibrinogen gradually converts into fibrin, thereby forming a fibrin clot. This process not only promotes hemostasis but also prevents pathogen invasion, serving as a scaffold for the attachment of various cells in the later stages. Upon entering the inflammation stage of the wound healing process, local vasoconstriction induces tissue ischemia, prompting the release of histamine and various vasoactive substances, consequently facilitating the dilation of local blood vessels [31]. Provoked by necrotic tissue and invading microorganisms, the body instigates an inflammatory response, characterized by the migration of white blood cells and other pertinent cells toward the wound site. The initial cells to infiltrate the wound are neutrophils, proficient in eradicating invading microorganisms while simultaneously engulfing necrotic debris and microbes. Neutrophils execute their bactericidal function by releasing proteases. Furthermore, neutrophils play a pivotal role in initiating the production of growth factors for macrophages, keratinocytes, and fibroblasts [32]. Following neutrophils, monocytes enter the wound and differentiate into macrophages, constituting indispensable cells in the wound healing cascade. Macrophages are primarily tasked with phagocytosing necrotic debris, microbes, and apoptotic neutrophils. Furthermore, during the inflammatory phase, macrophages secrete growth factors and other substances conducive to wound healing, which hold substantial importance in cell proliferation and migration [33]. Neutrophils and macrophages possess the capability to modify the provisional ECM, facilitating the progressive development of granulation tissue, which holds pivotal significance in the wound healing process [34]. The repair phase can be delineated into two stages: epithelial regeneration and granulation tissue formation. Wound repair commences from the wound edges, where residual basal cells proliferate and progressively migrate inward. Throughout this process, the proliferative activity of basal cells stimulates reactive proliferation of capillaries and connective tissue within the wound bed. Simultaneously, basal cell proliferation exerts a promotive effect on the growth of granulation tissue. Macrophages secrete a multitude of growth factors, such as platelet-derived growth factor (PDGF), transforming growth factor-beta (TGF-β), and transforming growth factor-alpha (TGF-α), among others, which play a pivotal role in fostering the conversion of the provisional ECM into granulation tissue [35]. The ECM acts as a scaffold for fibroblasts and endothelial cells, facilitating angiogenesis and fibroblast proliferation. Granulation tissue fills tissue defects while also safeguarding the wound site, thereby thwarting bacterial infection. The remodeling phase is typified by cellular apoptosis and collagen remodeling. Endothelial cells and myofibroblasts within granulation tissue undergo apoptosis, progressively transitioning into wound scars. Notably, the apoptosis of endothelial cells results in a decrease in blood vessel density, leading to a gradual lightning of scar color. In order to augment the tensile strength of the wound, collagen precursor fibers progressively align into bundles and undergo intermolecular cross-linking [27], culminating in scab formation at the wound site.

Fig. 2.

Fig. 2

The process of wound healing comprises four distinct stages: hemostasis, inflammation, proliferation, and remodeling.

Following the aforementioned dynamic processes, the wound gradually undergoes healing. Nevertheless, diabetic patients present with anomalous wound characteristics: owing to the sustained hyperglycemic and inflammatory conditions within the body, the formation of granulation tissue is compromised, consequently hindering tissue remodeling [36]. Consequently, wound healing in patients stagnates or regresses to a certain stage, resulting in delayed wound healing.

4. Factors affecting DFU healing

In normal circumstances, the healing of skin wounds represents a transient, time-bound process of self-repair aimed at reinstating the damaged skin's anatomical structure and physiological function [37]. When patients are afflicted with diabetes, factors including high blood glucose stimulation, aberrant local wound microenvironment, and microvascular occlusion, among intrinsic factors, as well as external factors like wound infection, inadequate debridement, and the patient's unhealthy lifestyle habits [38], impede DFU wound healing(Fig. 3). The mechanism behind delayed wound healing is intricate and remains incompletely understood to date. Presently, several acknowledged factors influencing DFU healing comprise the following.

Fig. 3.

Fig. 3

Comparative analysis of wound healing in diabetic wounds versus normal wounds.

4.1. Inflammation caused by bacteria

The high glucose microenvironment provides an optimal environment for the growth and proliferation of bacteria and various pathogens, rendering bacteria less susceptible to clearance, leading to their accumulation around the wound site. Common bacteria found in wound sites include Staphylococcus aureus, Pseudomonas aeruginosa, and β-hemolytic streptococcus. Apart from directly harming the host, these bacteria amplify inflammatory cytokines, proteases, and ROS to attract white blood cells, thus initiating and perpetuating inflammatory cascade reactions [39]. The proteases and ROS produced by bacteria degrade ECM and growth factors, thereby affecting cell migration and impeding the wound healing process.

4.2. Hypoxia at the site of localized injury

Elevated blood glucose levels in diabetic patients damage microvascular endothelial cells, resulting in microcirculatory abnormalities, erythrocyte membrane narrowing, local tissue ischemia, and circulatory disturbances. This ultimately leads to inadequate oxygen delivery to the local injured area, causing local tissue hypoxia, which is a primary contributing factor to diabetic wound damage. Furthermore, during the inflammatory phase, there is an increased oxygen demand by cells at the wound site, exacerbating wound hypoxia [40]. This ultimately results in inadequate energy supply for wound repair, impeding fibroblast proliferation, cell proliferation, and tissue self-repair mechanisms. Local tissue hypoxia in diabetic patients may be exacerbated by increasing ROS to amplify the inflammatory response.

4.3. Imbalance in matrix metalloproteinases (MMPs) expression

MMPs, as zinc-dependent endopeptidases, play a crucial role in the degradation of ECM components [41]. MMPs are involved in all stages of wound healing. During the inflammatory phase, MMPs remove damaged proteins and temporary ECM; in the proliferative phase, MMPs facilitate the degradation of capillary basement membrane, angiogenesis, and cell migration. In the remodeling phase, MMPs regulate tissue contraction and remodeling [42]. Based on the specificity of MMP substrates, they can be categorized into collagenases (MMP-1, MMP-8, MMP-13, MMP-18), gelatinases (MMP-2, MMP-9), stromelysins (MMP-3, MMP-10, MMP-11), membrane-type (MT) metalloproteinases (MMP-14, MMP-15, MMP-16, MMP-17, MMP-24, MMP-25), matrilysins (MMP-7, MMP-26), and others (MMP-12, MMP-19, MMP-20, MMP-21, MMP-23, MMP-27, MMP-28) [43,44]. Among these, gelatinases (MMP-2 and MMP-9) and collagenases (MMP-1 and MMP-8) play indispensable roles in the wound healing process. Tissue inhibitors of metalloproteinases (TIMPs) regulate MMPs by blocking their activity. Under normal physiological conditions, the interaction between MMPs, growth factors, and receptors ensures the equilibrium of the ECM. However, in the state of DFU, the expression of ECM proteins and their inhibitors becomes imbalanced, leading to matrix degradation [45]. This further results in decreased levels of growth factors and TIMPs, and increased expression of proteases and MMPs, with notable changes in MMP-1, MMP-2, MMP-8, and MMP-9. Concurrently, neutrophils and macrophages release pro-inflammatory cytokines such as interleukin-1 (IL-1) and tumor necrosis factor-alpha (TNF-α). Among the various upregulated MMPs, MMP-9 has been identified through research as a biochemical marker associated with the formation and persistence of chronic wounds [46]. In DFU, MMP-9 plays roles in regulating keratinocyte function and degrading collagen. Its overexpression impairs the migratory ability of keratinocytes, preventing their migration to the wound site, thereby hindering wound healing.

4.4. Abnormal macrophage phenotype

The dysregulation of macrophage polarization induced by high blood sugar and oxidative stress is one of the primary factors contributing to DFU. Excessive inflammation at the wound site prolongs the healing phase, causing it to remain in the inflammatory stage for an extended period, preventing a normal transition to the stages of cell proliferation and tissue remodeling, thus substantially delaying wound healing and significantly heightening the risk of wound infection. The excessive inflammatory response at the wound site is intimately linked to aberrant macrophage phenotypes. Macrophages are immune effector cells that play a crucial role in tissue damage response and the maintenance of homeostasis. Functionally, activated macrophages can be broadly classified into two types: classically activated macrophages (caMϕ or M1 type) and alternatively activated macrophages (aaMϕ or M2 type). M1 macrophages represent a pro-inflammatory activation state, whereas M2 macrophages are characterized by anti-inflammatory and tissue repair-promoting functions. M1 macrophages are activated by interferon-γ secreted by CD4+ T helper cells (Th1), lipopolysaccharides (LPS) from Gram-negative bacteria, macrophage colony-stimulating factor (M-CSF), and tumor necrosis factor (TNF) [47]. The release of antimicrobial and anti-tumor inflammatory factors by M1 macrophages leads to tissue damage induced by ROS, which in turn affects tissue regeneration and wound healing. In contrast, M2 macrophages are predominantly induced by cytokines such as IL-4 and IL-13 and exhibit high levels of CD206 expression. These macrophages demonstrate enhanced phagocytic abilities and secrete anti-inflammatory cytokines, including IL-10 and TGF-β. M2 macrophages play a pivotal role in promoting tissue repair and angiogenesis while mitigating inflammatory responses in inflammatory diseases. In diabetic wounds with impaired healing, there is an accumulation of M1 macrophages and a reduction in M2 macrophages, leading to the retention of wound healing in the inflammatory phase and impeding the transition to the phases of cell proliferation and migration [48]. The impaired transition of macrophage phenotypes from M1 to M2 in DFU is associated with compromised wound closure, inadequate vascularization, and diminished collagen deposition [49].

4.5. Excessive deposition of collagen fibers

The excessive deposition of collagen fibers results in the formation of dense fibrotic tissue, which diminishes tissue elasticity and flexibility, restricts cell migration, and obstructs the transport of nutrients and oxygen. Furthermore, collagen fibers entrap and sequester growth factors, reducing their availability for interaction with cell surface receptors, thus significantly impairing tissue repair and remodeling processes. This may be associated with reduced secretion of urinary plasminogen activator (uPA) and increased secretion of urinary plasminogen activator inhibitor (uPAR) at the site of diabetic wounds [50].

4.6. Impaired angiogenesis and neovascularization injury

Continuously elevated blood glucose levels inhibit the typical flow of blood to the extremities, resulting in localized tissue hypoxia and insufficient nutrient delivery, consequently limiting angiogenesis. Additionally, abnormalities in endothelial progenitor cell function induced by diabetes could further aggravate the impairment of neovascularization in DFU [51].

4.7. Reduced activity of functional factors

Elevated blood glucose levels impact the expression of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor 1α (HIF-1α), concurrently promoting the non-enzymatic glycation of numerous vital proteins. Non-enzymatic glycation of proteins refers to a chemical reaction in which sugar molecules covalently bond with amino groups of proteins without the participation of enzymatic catalysts. This process disrupts protein structure and functionality, thereby impeding multiple physiological processes crucial for wound healing [52]. In individuals with diabetes, non-enzymatic glycation is a major factor contributing to delayed wound repair. Therefore, effective management of blood glucose levels and regulation of non-enzymatic glycation reactions are critical for enhancing wound healing.

5. Treatment methods

Presently, standard therapeutic modalities for DFU predominantly involve wound debridement, dressing changes, hyperbaric oxygen therapy, negative pressure wound therapy, and wound offloading. These prevalent foundational therapies contribute to the management and amelioration of symptoms. Moreover, MSC-based therapies have garnered significant attention due to their novel approach in enhancing the healing of DFUs (Refer to Table 1 for a comparative analysis of the advantages and limitations of various treatment modalities.).

Table 1.

A comparative analysis of the advantages and limitations of various treatment modalities for DFU.

Treatment methods Advantages limitations
Regulate blood glucose levels By addressing the underlying causes of DFUs, wound healing can be significantly promoted. Indirect treatment methods for DFUs fail to directly target the wound itself.
Manage and control infections By managing infections to eliminate the substantial presence of pathogenic microorganisms around the wound, the inflammatory response induced by bacteria can be effectively suppressed. The sources of infection in DFU can be diverse and complex and conventional antibiotic therapy alone may not effectively address all infection-related issues. Furthermore, deep-seated infections commonly associated with DFU are often difficult to control with topical antibiotic therapy alone and may require more invasive treatment approaches.
Perform wound debridement Debridement facilitates the healing of healthy granulation tissue by removing necrotic tissue and foreign debris from the wound bed, making it a fundamental approach in the treatment of DFUs. Debridement is only one component of a comprehensive treatment strategy for DFUs. Relying solely on debridement may not achieve complete resolution of DFUs. To attain optimal outcomes, debridement must be integrated with other therapeutic modalities.
Apply wound dressings Wound dressings not only serve as a protective barrier for DFUs but also play a role in antimicrobial activity and the promotion of vascular and tissue regeneration. Wound types and conditions vary among patients, and universal dressings may not meet the needs of all individuals. Therefore, treatment plans must be tailored based on the specific wound characteristics and the overall health status of the patient.
Decompression of wounds Implementing offloading therapy to reduce plantar pressure is crucial for enhancing the healing of DFUs. Wound decompression therapy does not address the underlying causes of delayed wound healing. To achieve the best therapeutic outcomes, it is essential to combine offloading therapy with other treatment modalities.
Employ negative pressure wound therapy (NPWT) NPWT not only maintains a clean and dry wound environment but also reduces the frequency of dressing changes. The safety and efficacy of NPWT for DFU remain largely unclear. with ongoing debate and uncertainty regarding its application [95].
Administer hyperbaric oxygen therapy (HBOT) HBOT not only promotes collagen synthesis, growth factor production, and neovascularization but also exhibits significant antimicrobial effects against anaerobic bacteria. HBOT is currently limited to improving the short-term outcomes of DFUs, and there remains substantial debate regarding the overall efficacy of this treatment.
Employ skin substitutes Skin substitutes can mimic the natural ECM and serve as carriers for cells and growth factors, thereby effectively enhancing wound healing. As foreign materials, skin substitutes carry the risk of being rejected by the body.
Treatment utilizing MSCs Unlike traditional therapies that merely protect the wound and rely on the body's inherent healing capacity, MSC therapy promotes wound healing through systematic regulation of multiple aspects related to DFUs. Therefore, it presents a more effective alternative to conventional treatments. MSCs therapy is associated with limitations such as the risk of recipient cell rejection and potential for tumorigenesis.

5.1. Regulate blood glucose levels

The sustained hyperglycemic state in diabetic individuals can result in complications such as Diabetic Neuropathy (DN), Peripheral Arterial Disease (PAD), and immune suppression, all of which exert substantial influence on the healing process of DFU. Hence, blood sugar control can profoundly facilitate wound healing from its root. Research indicates that a notable enhancement in wound healing occurs with decreased blood sugar level [53]. Currently, clinical practices typically employ biguanides, sulfonylureas, and insulin to regulate patients' abnormally elevated blood glucose levels. Additionally, maintaining dietary control, regular exercise, and healthy lifestyle habits play an indispensable role in regulating blood glucose levels.

5.2. Manage and control infections

Owing to the extended duration required for wound healing in diabetic patients, wounds are exposed for prolonged periods, resulting in a significant presence of pathogenic microorganisms surrounding the wound. Research suggests that the infection rate at the wound site of DFU patients is approximately 40 % [54]. To initiate targeted therapy, it is crucial to accurately identify the causative agent. Microbiological analysis of soft tissue or bone samples from the patient is commonly performed to ascertain the pathogen's identity. Antibiotics are commonly employed for infection control, with frequently used medications including penicillins, cephalosporins, fluoroquinolones, carbapenems, and others. In cases where antibiotic therapy fails to yield satisfactory outcomes, surgical intervention may be warranted.

5.3. Perform wound debridement

Debridement encompasses the elimination of infected tissue, aging and necrotic tissue, bacterial biofilm, and foreign bodies from the wound bed to promote the healing of surrounding healthy granulation tissue [55]. This method stands as the cornerstone in the treatment of DFU. Multiple approaches, such as surgical debridement, enzymatic debridement, mechanical debridement, ultrasound debridement, and bio-surgical debridement, can be employed. The objective is to stimulate wound healing through the targeted stimulation of the wound bed. Debridement stands as the most fundamental method for treating DFU.

5.4. Apply wound dressings

Dressings function as a protective barrier for DFU. Moreover, certain advanced bandages possess antimicrobial properties while concurrently stimulating vascular and tissue regeneration [56]. The materials for wound dressings are varied, and the choice of suitable dressings should be meticulously aligned with the wound type, severity, and specific characteristics, aiming to minimize treatment duration and facilitate DFU healing. Presently, commonly used materials for medical dressings encompass hydrogels, alginate, foam, and silver-containing dressings [57]. Among these, hydrogel dressings are the most prevalent. Studies have shown that hydrogels exhibit better therapeutic efficacy in treating DFU compared to conventional dressings [58]. Hydrogels, characterized by their high water content and three-dimensional porous network, are capable of loading therapeutic agents or cytokines that promote the healing of diabetic wounds. Current research has extensively utilized hydrogels for the delivery of various glycemic control medications [[59], [60], [61], [62]] and antimicrobial agents [[63], [64], [65], [66]] to enhance wound healing. Moreover, studies indicate that hydrogels can also incorporate ROS scavengers [67,68] and angiogenic compounds [69,70] to synergistically promote the healing of DFUs. Moreover, hydrogels can serve as carriers for cells or growth factors [49,50], which have been demonstrated to play a crucial role in facilitating DFU healing. In comparison to hydrogels, alginate demonstrates more favorable swelling properties, mechanical characteristics, and maximal adhesion to biological tissues [71]. Studies indicate that alginate sodium hydrogel loaded with vicenin-2 can suppress pro-inflammatory cytokines and facilitate diabetic wound healing under elevated blood glucose levels [72].

5.5. Decompression of wounds

DFU arise due to the presence of localized high-pressure areas on the foot, either short-term and concentrated or long-term and diffuse low-pressure regions. Additionally, the presence of peripheral arterial disease and diabetic neuropathy complicates diabetic ulceration further. As DFU occurrence has been linked to elevated plantar pressure, offloading assumes a pivotal role in DFU treatment. Common offloading techniques encompass the utilization of crutches, surgical shoes, and bandages [73]. Total contact casts (TCCs), made of special materials, are regarded as the gold standard for offloading in DFU treatment. Employing TCCs can notably reduce the healing time of DFUs compared to conventional dressing therapy [74]. Moreover, alternative offloading strategies encompass removable knee-high devices, removable ankle-high devices, or felted foam, which can be inserted into suitable footwear for cushioning purposes [75].

5.6. Employ negative pressure wound therapy (NPWT)

To clear fluid and infection from the wound site, negative pressure wound therapy (NPWT) is commonly used to apply negative pressure to the covered wound. This approach effectively reduces the frequency of dressing changes while ensuring wound cleanliness and dryness [76]. Research findings suggest that NPWT can suppress the mRNA expression of pro-inflammatory cytokines IL1β and MMP-9, while concurrently enhancing the expression levels of VEGF and TGF-β1 [77].

5.7. Administer hyperbaric oxygen therapy (HBOT)

HBOT encompasses both local oxygenation of ulcers and systemic oxygenation. This approach stimulates collagen synthesis, promotes growth factor production, and facilitates neovascularization [78]. Furthermore, HBOT demonstrates notable inhibitory effects on anaerobic bacteria, leading to a reduction in antibiotic usage [79]. Nevertheless, HBOT is currently surrounded by considerable controversy. Current studies indicate that HBOT can only enhance short-term treatment outcomes for DFU [80].

5.8. Employ skin substitutes

With the successful therapeutic outcomes observed with skin substitutes, the application of skin substitutes in treating DFU has seen a steady rise. Skin substitutes mimic the natural ECM, boasting commendable biocompatibility and serving as carriers for cells and growth factors, thereby fostering effective wound healing. Nonetheless, the risk of rejection as an exogenous substance remains present. Presently, bioengineered membranes composed of materials such as collagen are frequently employed to mitigate this risk [81].

5.9. Utilizing the tissue regeneration-promoting properties of MSCs

In recent years, MSCs therapy has emerged as a promising avenue for enhancing tissue regeneration and wound healing [[82], [83], [84]], owing to the myriad characteristics and advantages of stem cells. Numerous studies have substantiated the role of MSCs in enhancing wound healing. Shi et al. employed intravenous transplantation of human umbilical cord mesenchymal stem cells (hUC-MSCs) in a diabetic rat model induced by streptozotocin. The findings indicate that intravenous infusion of hUC-MSCs can enhance epithelialization and granulation tissue formation in DFUs, thereby promoting tissue regeneration [85]. Additionally, hUC-MSCs play a significant role in angiogenesis, cellular proliferation, and apoptosis reduction [[85], [86], [87]] (Fig. 4). Moreover, the investigation by Han et al. underscored the regulatory influence of the Wnt signaling pathway on the proliferation and differentiation of hUC-MSCs. Activation of the Wnt signaling cascade facilitated the proliferation and differentiation of hUC-MSCs on the CCLDADM scaffold, thereby effectively fostering the resolution of diabetic cutaneous injuries [88]. Wei et al. discerned that hUC-MSCs exhibited the potential to enhance the healing process of cutaneous wounds in a murine model of DFU by inhibiting the expression of MMP-9 while concurrently fostering the expression of MMP-8 [89]. Gustavo et al.'s investigation demonstrates the efficacy of Allogeneic adipose-derived mesenchymal stromal cells (ASCs) in alleviating bleomycin-induced pulmonary and dermal fibrosis in elderly mice, concurrently accelerating wound healing [90]. Furthermore, Liu et al. elucidated that hUC-MSCs significantly augmented the expression levels of CD31 and VEGF, thereby fostering the formation of neovessels and consequently accelerating the regeneration of radiation-induced cutaneous ulcers [91]. Additionally, Wang et al. revealed that LPS-pretreated BM-MSCs facilitated the healing process of diabetic foot ulcers by enhancing angiogenesis and local epithelialization at the wound site [92]. Although MSCs exhibit significant advantages in promoting tissue regeneration and wound healing, as a novel cellular therapy, MSC treatment also has certain limitations. MSCs typically disappear within 24 h, preventing their long-term presence at injury sites and sustained action. Additionally, standardized protocols for MSC therapy in DFUs have yet to be established. For the clinical translation of MSC therapy for DFUs, numerous challenges must be addressed, including conditions for cell isolation and culture, cryopreservation methods, MSC infusion dosage, timing, frequency, and routes of administration, as well as donor MSC heterogeneity. On the other hand, MSCs exhibit a propensity for tumorigenesis. Studies have shown that under certain culture conditions, MSCs can bypass senescence and generate malignant cells [93]. Furthermore, despite their immune-privileged status, allogeneic MSCs can be recognized by the host immune system and induce donor-specific immune responses due to the presence of alloantigens on the cell surface [94]. The associated challenges and limitations of MSC therapy warrant further research and improvement.

Fig. 4.

Fig. 4

Samples of wound tissues were collected for analysis on the 16th day post-treatment. hUC-MSCs exert significant effects on enhancing epithelialization, promoting granulation tissue formation, facilitating tissue regeneration, stimulating angiogenesis, fostering cell proliferation, and mitigating apoptosis in DFUs.(a) Histological analysis using H&E staining revealed enhanced re-epithelialization effects on DFU wounds following hUC-MSCs treatment compared to the control group. (b) Remarkable thickening of granulation tissue was observed in the hUC-MSCs treatment group compared to the control group.(c) Masson's trichrome staining revealed that the collagen deposition effect was superior in the hUC-MSCs treatment group compared to the control group. (d) Immunohistochemical staining was employed to assess CD31 expression. In comparison to the control group, the hUC-MSCs group exhibited a greater abundance of blood vessel structures at the wound site. (e)Ki-67 expression was assessed through staining. The hUC-MSCs group displayed a substantially higher number of Ki-67-positive cells than the control group, indicating its superior capacity to promote cell proliferation. (f) The TUNEL assay was employed to evaluate cell apoptosis. Relative to the control group, the hUC-MSCs group demonstrated a marked reduction in apoptotic cells, underscoring its enhanced ability to suppress cell apoptosis.Data were presented as mean ± standard deviation for each group. ∗P < 0.05. Scale bar, 100 μm.Reproduced under the terms of the CC-BY license [85]. Copyright 2020, the authors.

6. The characteristics of MSCs

MSCs, originating from the mesoderm, are a multipotent non-hematopoietic adult stem cell population. They were initially identified in the 1960s during investigations into bone marrow, characterized by their fibroblast-like morphology [96]. MSCs are characterized by their adhesion and plasticity; they have the potential to differentiate into osteoblasts, adipocytes, and chondrocytes. Additionally, they express CD73, CD90, CD105, and lack the expression of CD14, CD34, CD45, CD11b, CD79a, CD19, HLA-DR. MSCs have a broad spectrum of sources [97]. Apart from bone marrow, they can be extracted from adipose tissue, placenta, umbilical cord, amniotic fluid, dental pulp, Wharton's jelly, and tissues including liver, spleen, kidney, lung, and brain [98]. Among these, bone marrow-derived mesenchymal stem cells (BM-MSCs), adipose tissue-derived mesenchymal stem cells (ADSCs), and umbilical cord-derived mesenchymal stem cells(UC-MSCs) are the most prevalent sources utilized for therapy. MSCs exhibit a plethora of characteristics including self-renewal, colony formation, and multi-lineage differentiation abilities. They have the capability to differentiate into diverse cell types including osteoblasts, chondrocytes, adipocytes, endothelial cells, cardiomyocytes, hepatocytes, and neurons [99]. The differentiation potential of MSCs is contingent upon factors such as cell source, cell culture environment, and expansion conditions. Apart from the properties mentioned above, MSCs secrete a plethora of growth factors, cytokines, and other bioactive substances. These bioactive molecules can recruit other cells to the site of tissue damage, promoting angiogenesis and facilitating tissue repair [100]. Additionally, MSCs play a role in immune modulation, thereby mitigating inflammatory responses [101]. Furthermore, MSCs can differentiate into keratinocytes [102,103], endothelial cells [[104], [105], [106]], and fibroblasts [[107], [108], [109]], which play crucial roles in wound healing by replacing damaged cells. This promotes wound healing, stimulates angiogenesis, reduces inflammation, regulates extracellular matrix remodeling, and minimizes scar formation.

Since most stem cells exhibit low expression levels of HLA class I and lack or have low expression of HLA class II molecules, along with the absence of co-stimulatory factors such as CD80, CD40, and CD86, as well as hematopoietic cell surface markers including CD14, CD34, and CD45 [110,111], they do not trigger immune rejection during allogeneic transplantation. Research indicates that the downregulation of HLA class I expression can mitigate the cytotoxic effects of natural killer (NK) cells on MSCs [112]. Moreover, MSCs can regulate the expression of interleukin-4 (IL-4) and interferon-gamma (IFN-γ) in effector T cells, facilitating the conversion of helper T cells (Th2) to Th1 cells and exerting inhibitory effects on disease responses. Furthermore, studies have reported that BM-MSCs, by expressing HLA-DR molecules, evade recognition by NK cells [113]. Consequently, transplanting allogeneic MSCs does not trigger the production of alloantibodies or the initiation of T cells [114]. As cytotoxic immune factors can trigger the dissolution of MSCs [115], IFN-γ serves as an antagonist to NK cells, safeguarding MSCs from NK cell attacks [116]. Additionally, IFN-γ enhances the differentiation potential of these cells through both nuclear factor kappa B (NF-κB)-dependent and -independent pathways [117]. MSCs, owing to these immunological attributes, emerge as a viable source for cell transplantation experiments.

7. Paracrine effects of MSCs

The paracrine actions of MSCs were first characterized by Haynesworth et al. [118]. Their research revealed that MSCs have the capability to produce and secrete diverse bioactive molecules including growth factors, cytokines, chemokines, proteins, and miRNAs. These bioactive factors exert regulatory effects on neighboring cells. Research has indicated that MSCs secrete a plethora of bioactive factors that facilitate angiogenesis, stimulate ECM remodeling, impede cellular fibrosis and apoptosis, dampen local inflammatory responses, and modulate immune reactions [[119], [120], [121]]. This evidence suggests that MSCs contribute to tissue regeneration by either direct intervention or paracrine signaling [96]. Presently, extensive research has investigated the therapeutic implications of bioactive factors secreted by MSCs across a spectrum of diseases, encompassing neurological disorders, cardiovascular ailments, bone and cartilage regeneration, immune-related conditions, liver injury, among others [122]. These investigations elucidate that the bioactive factors released by MSCs serve as mediators to activate target cells, while also stimulating neighboring cells to secrete pertinent bioactive factors [123]. Additionally, the paracrine effects of MSCs encompass the secretion of exosomes. Exosomes are nanoscale extracellular vesicles with a lipid bilayer membrane structure secreted by MSCs. By transmitting signaling molecules such as non-coding RNAs and proteins, they facilitate intercellular communication [124], promoting the survival, proliferation, migration, and gene expression of recipient cells while reprogramming the behavior of target cells, thereby regulating physiological and pathological processes. Multiple studies have confirmed that exosomes secreted by MSCs possess therapeutic effects similar to those of MSCs [[125], [126], [127], [128]].

8. Exosomes released by MSCs

Exosomes, non-replicative lipid bilayer spherical structures, constitute a diverse array of double-layered lipid membrane-enclosed extracellular vesicles secreted by eukaryotic cells or prokaryotes. Their dimensions typically span from 30 nm to 200 nm. Moreover, they encompass two subtypes: microvesicles, ranging from 100 to 1000 nm, and apoptotic bodies, ranging from 500 to 2000 nm [129]. Exosomes were initially identified in investigations into iron uptake in reticulocytes, presenting as small vesicles suspended in the reticulocyte's supernatant [130]. Exosomes originate from cellular endocytosis, whereby components of the extracellular milieu, such as proteins, lipids, and diverse small molecules, undergo internalization to form early endosomes. These early endosomes gradually mature into late endosomes and eventually transform into multivesicular bodies (MVBs). The fusion of MVBs with the plasma membrane occurs through interactions with microtubules and the cellular cytoskeleton, culminating in exosome formation. Notably, a subset of MVBs engages in fusion with lysosomes for degradation purposes. Upon their formation, exosomes employ various mechanisms, including endocytosis, uptake by macrophages, phagocytosis, and direct fusion with the plasma membrane, to enter recipient cells [131](Fig. 5). Exosomes manifest distinct physiological attributes and functionalities contingent upon divergent physiological or pathological milieus. They are disseminated by an array of dynamic cellular entities within the organism, orchestrating an information relay via the payload they harbor. Inter-cellular communication assumes paramount significance in upholding the homeostasis of the microenvironment in multicellular organisms. Exosomes partake in intercellular communication, a function that transcends interactions solely among cells within the same organism, extending its relevance to cells of disparate organisms, irrespective of species affiliation. Initially, exosome communication was posited to entail the transfer of extracellular proteins and certain soluble factors. Nonetheless, as investigations progressed, the consensus recognizing exosomes' ability to disseminate non-coding RNA and proteins, alongside other signaling molecules, to mediate intercellular communication has gained widespread acceptance [132]. Exosomes, serving as conduits for intercellular information transfer, harbor a plethora of unparalleled advantages: their nano-sized dimensions facilitate unhindered intercellular transport, while their distinctive phospholipid bilayer structure safeguards bioactive cargo from degradation enroute. Moreover, the abundance of specific proteins on the exosome surface endows them with superior targeting prowess. Exploiting these distinctive merits, contemporary research endeavors concentrate on harnessing exosomes as vectors for delivering drugs, RNA, proteins, and various other payloads [133]. The RNA cargo transported by exosomes encompasses mRNA, miRNA, ribosomal RNA, circular RNA, DNA, lncRNA, and various others, pivotal in orchestrating intercellular regulation [134]. Aside from harboring proteins linked to membrane transport and fusion, such as annexins, ESCRT complexes, Rab-GTPases, actin, and β-tubulin, exosomes are enriched with membrane surface markers, including CD9, CD63, CD81, and CD82, alongside HSP70, HSP90, Tsg101, and Alix [135]. Accordingly, these are conventionally acknowledged as ubiquitous exosomal markers. Exosomes demonstrate heterogeneity, whereby exosomes sourced from distinct tissues and cells, or even from identical tissues but disparate cells, manifest notable disparities. This diversity stems from variances in gene expression across different cells, discrepancies in intercellular culture conditions, divergent concentrations of membrane-bound proteins, and disparities in the exosomes' cargo-loading capabilities [136]. The contemporary techniques employed for exosome isolation and purification encompass ultracentrifugation, ultrafiltration, size-exclusion chromatography, immunoseparation, and microfluidics, among a plethora of methodologies [137]. Subsequent to exosome retrieval, elucidation is conducted employing an array of methodologies, including transmission electron microscopy, nanoparticle tracking analysis, protein imprinting, enzyme-linked immunosorbent assay (ELISA), flow cytometry, scanning electron microscopy, atomic force microscopy, dynamic light scattering, fluorescence-activated cell sorting, and resistive pulse sensing, to name a few [138].

Fig. 5.

Fig. 5

The process of exosome generation and secretion. Reproduced under the terms of the CC-BY license [151]. Copyright 2023, the authors.

Research has demonstrated MSCs as the predominant origin of exosomes [139]. The concept of utilizing exosomes derived from MSCs for the treatment of myocardial ischemia/reperfusion injury was introduced early on, thereby shifting the research focus towards elucidating the role and therapeutic applications of exosomes in diseases [140]. The morphological attributes and isolation techniques of exosomes originating from MSCs closely resemble those from alternative sources. In addition to their identification via ubiquitous exosomal markers like CD9 and CD81, discernment can also be facilitated through the utilization of MSC surface markers including CD29, CD44, CD73, and CD90. Analogously, exosomes originating from MSCs encapsulate a diverse array of bioactive substances. Profiling of exosomes released by BM-MSCs unveiled the presence of 730 functional proteins, pivotal in governing cellular proliferation, migration, and assorted physiological processes [141]. Furthermore, exosomes derived from MSCs harbor a plethora of cytokines conducive to angiogenesis and immune regulation, including VEGF, transforming growth factor-β1 (TGF-β1), interleukin-6 (IL-6), interleukin-10 (IL-10), among others [142]. Exosomes sourced from MSCs convey a plethora of signaling molecules capable of transmitting and exerting effects on endothelial cells, fibroblasts, immune cells, keratinocytes, among others, thereby fostering wound vascularization, enhancing wound circulation, modulating inflammatory responses, and thereby facilitating tissue repair [143]. Alongside functional proteins and cytokines, exosomes derived from MSCs encapsulate a myriad of RNA species. Presently, extensive research endeavors concentrate on elucidating the enrichment of miRNA within exosomes secreted by MSCs [144]. Contemporary investigations posit that the miRNA and mRNA encapsulated within exosomes wield pivotal roles in the intricate process of biological information transmission [145]. Owing to the advantageous attributes of exosomes secreted by MSCs, including low immunogenicity, small size, prolonged half-life, potent penetrative capacity, and elevated biocompatibility, they harbor immense therapeutic potential in disease management [146]. Reports indicate that exosomes sourced from MSCs exhibit analogous functionalities to stem cells in terms of anti-inflammatory responses, tissue regeneration, and immune modulation [147]. Moreover, unlike stem cells, exosomes have the added advantage of mitigating the adverse reactions associated with stem cell therapy [148]. Exosomes serve as a mirror of the functional capabilities of MSCs and constitute crucial mediators in the paracrine activity of MSCs. The existence of exosomes renders cell-free therapies for wound healing plausible [149]. Recent research endeavors have been focused on augmenting the reparative potential of exosomes for tissue injury by manipulating the expression profile of exosomal contents, preconditioning MSCs, and artificially modifying the receptors on the exosome surface [149].

In conclusion, exosomes secreted by MSCs manifest superior advantages in facilitating wound healing compared to MSCs themselves. Prevalent MSCs employed in contemporary wound healing investigations involving exosomes comprise BM-MSCs,UC-MSCs and ADSCs [150].

9. The role of MSCs and their derived exosomes in DFU

Due to their advantages in regenerative medicine, MSCs play a crucial role in facilitating the healing of DFU. MSCs exert their healing effects through a multifaceted and coordinated approach. As subcellular components of MSCs, exosomes inherit the regenerative and immunomodulatory properties of their parent MSCs. These exosomes possess a unique bilayer membrane structure that safeguards their contents from degradation or damage. Research indicates that exosomes can exert functions similar to those of MSCs, underscoring their crucial role in promoting tissue repair and regeneration [125](Table .2).

Table 2.

The role of MSCs and their derived exosomes in DFU.

Source Mechanisms Researchers References
Regulation of keratinocyte function BM-MSCs BM-MSCs enhance the levels of angiogenin-1, VEGF, and keratin, a keratinocyte-specific protein, at the wound site, thereby stimulating keratinocyte proliferation. W. Zhang et al. [152]
hUC-MSCs hUC-MSCs precisely target the site of injury, stimulating the growth of cytokeratin 19 and enhancing the regenerative potential of the epithelial tissue at the wound site. L. Chen et al. [153]
BM-MSCs The conditioned medium from BM-MSCs increases the levels of phosphorylated focal adhesion kinase, matrix metalloproteinase-2, epidermal growth factor, and insulin-like growth factor 1 in human keratinocytes. J. Kato et al. [154]
Promotes angiogenesis ADSCs Enhances the expression of ANG1, FILK1, and VEGF, while simultaneously inhibiting the expression of VASH1 and TSP1.Enhanced endothelial cell activity and vascular regenerative potential. Y. Zhang et al. [161]
ADSCs Overexpression of Nrf2-exosomes enhances endothelial progenitor cell proliferation and angiogenesis by promoting the phosphorylation levels of SMP30, VEGF, and VEGFR2. X. Li et al. [162]
hUC-MSCs Inhibition of high glucose-induced oxidative stress in HUVECs promotes their activity, proliferation, and angiogenesis. C. Yan et al. [163]
hUC-MSCs CircHIPK3 in exosomes inhibits miR-20b-5p, thereby promoting the expression of Nrf2 and VEGFA. Furthermore, circHIPK3 suppresses the expression of miR-124 in endothelial cells, protecting them from high glucose-induced damage. The protective role of circHIPK3 extends to mitigating cellular damage, significantly reducing apoptosis and inflammation, and thereby enhancing angiogenesis. Z. H. Liang et al. [165]
hiPSC-MSCs Exosomes are internalized by HUVECs, enhancing their proliferation and migratory capabilities. Furthermore, these exosomes activate the Erk-1/2 signaling pathway, thereby promoting angiogenesis. J. Zhang et al. [166]
Promotes collagen deposition ADSCs HSP90 within exosomes binds to the LRP1 receptor on the cell membrane, thereby activating the downstream AKT signaling pathway. This activation subsequently enhances the proliferation and migratory capabilities of keratinocytes and fibroblasts. S. Ren et al. [167]
ADSCs By stimulating monocytes or macrophages to secrete higher levels of TGF-β1, and subsequently activating the TGF-β/Smad3 signaling pathway, the proliferation of fibroblasts is enhanced, which in turn promotes the synthesis of type I collagen in diabetic wounds. H. H. Hsu et al. [169]
ADSCs hypADSCs-exo promote the proliferation, migration, and ECM secretion of fibroblasts by activating the PI3K/AKT signaling pathway. J. Wang et al. [170]
Promoting cell proliferation BM-MSCs Promotes re-epithelialization, epidermal junction formation, skin appendage regeneration, inflammatory infiltration, vascularization, granulation tissue formation, and the development of dense collagen fibers, while simultaneously concentrating regenerative factors and proteins. T. de Mayo et al. [171]
ADSCs Facilitates the proliferation and migration of fibroblasts while simultaneously enhancing collagen synthesis capacity. L. Hu et al. [172]
ADSCs Promotes the formation of neovascularization, protects ischemic/reperfusion flap grafts, and increases IL-6 levels. C. M. Pu et al. [173]
hiPSC-MSCs Enhances fibroblast proliferation and migration, increases the synthesis of collagen and elastin, and stimulates the formation of capillary networks in vitro. J. Zhang et al. [174]
Antiapoptotic effect BM-MSCs By activating APAF1, apoptosis in epithelial HaCaT cells is effectively inhibited. C. Shen et al. [170]
hP-MSCs Enhance proliferation and anti-inflammatory capabilities while suppressing apoptosis of corneal epithelial cells. H. Tao et al. [176]
ADSCs miR-10b within exosomes targets PEA15 to enhance the expression of CDK6, thereby promoting the proliferation and migration of H2O2damaged HaCaT cells and inhibiting their apoptosis. X. Liao et al. [179]
ADSCs Through Wnt/β-catenin signaling pathway, the proliferation and migration of HaCaT cells are promoted while simultaneously inhibiting their apoptosis T. Ma et al. [180]
Anti-inflammatory effect hUC-MSCs In a hyperglycemic environment, inhibition of inflammatory factors such as IL-1β, IL-6, and TNF-α, as well as the expression levels of NOX1 and NOX in HUVECs, effectively suppresses oxidative stress and promotes cellular proliferation. C. Yan et al. [163]
ADSCs Exosomes with high Nrf2 expression inhibit the levels of inflammatory and oxidative stress-related proteins in HaCaT cells, fibroblasts, and HUVECs. This inhibition facilitates the formation of granulation tissue, angiogenesis, and growth factor expression at the wound site. X. Li et al. [162]
ADSCs By modulating SIRT3/SOD2, oxidative stress levels induced by high glucose are suppressed, leading to reduced ROS accumulation, minimized mitochondrial dysfunction and inflammation, and activation of endothelial cells. Y. Zhang et al. [161]
BMMSCs, JMMSCs By transferring miR-223 to target human PKNOX1, macrophage polarization towards the M2 phenotype is induce. X. He et al. [184]
Regulation of macrophage polarization. hUC-MSCs Regulating macrophage polarization improves pancreatic dysfunction in type 2 diabetic mice. D. Philipp et al./ [190]
hUC-MSCs Regulating macrophage polarization promotes the restoration of endothelial cell function. Secreting PGE2 modulates the expression levels of IL-10 and VEGF in the local microenvironment of endothelial cells. J. B. Tefft et al./ [155]
G-MSCs Inducing macrophage polarization plays a crucial role in wound healing. X. Yu et al./ [24]
ADSCs By delivering miR-21 to target the PI3K/Akt signaling pathway, this process promotes the polarization of macrophages from the M1 to the M2 phenotype, thereby facilitating angiogenesis. D. Zhu et al. [193]
BMMSCs By delivering miR-223, the polarization of macrophages towards the M2 phenotype is facilitated, which accelerates wound healing. X. He et al. [184]
Immortalized E1-MYC 16.3 human embryonic stem cell-derived MSCs Inhibiting the expression levels of M1 macrophages and inflammation-related cytokines, while promoting the infiltration of M2 macrophages. S. Zhang et al. [194]
BM-MSCs Promoting macrophage polarization towards an M2-like phenotype, thereby facilitating tissue repair and modulating the inflammatory response. C. S. Chamberlain et al. [195]
Inhibition of ROS generation BM-MSCs Inhibition of myeloperoxidase and induction of inducible nitric oxide synthase lead to reduced expression of inflammatory cytokines IL-1β, IL-4, IL-6, IL-9, TNF-α, andIFN-γ. K. F. Al-Massri et al. [198]
ASCs Inhibition of ROS and myeloperoxidase expression in stimulated monocytes and macrophages results in a reduction of M1 macrophage expression. M. Ortiz-Virumbrales et al. [199]
BM-MSCs mitochondrial ROS production in macrophages, inhibits the activation of intracellular NLRP3 and caspase-1, thereby affecting the transcription of TNF-α and IL-6, and modulating the expression of IL-1β. Y. Zhou et al. [200]
BM-MSCs Hypoxia-preconditioned MSC-derived exosomes regulate the accumulation of ROS in epithelial cells through HIF-1α. F. Zhu et al. [201]
WJSCs Modulates the GP91/ROS/inflammasome signaling pathway while simultaneously enhancing the expression of antioxidant proteins. W. S. Hu et al. [202]
hUC-MSCs The released exosomes inhibit glucocorticoid-induced osteoblast apoptosis by modulating the MAPK pathway and regulating ROS levels. H. Lu et al. [203]
BM-MSCs Modulating the innate immune response capability through the ROS/NLRP3 signaling pathway. A. Qin et al. [204]
Regulation of T cells BM-MSCs Inhibiting the differentiation and function of pro-inflammatory T cells while inducing a regulatory T cell phenotype, thereby effectively modulating inflammation at the site of injury. A. Li et al. [211]
Regulation of NETs formation BM-MSCs When used in conjunction with antibiotics, it significantly reduces bacterial load at the wound site, thereby promoting wound healing. L. Chow et al. [216]
hAM-MSCs By mediating immune regulation through PGE2, it effectively inhibits the release of NETs. G. A. Estúa-Acosta et al. [217]
hAM-MSCs Regulate the formation of NETs through miR-125a-3p contained within the exosomes. Y. Morishima et al. [218]
Inhibition of ferroptosis hUC-MSCs The miRNA-17-92, which is highly expressed and enriched in exosomes, promotes the proliferation and migration of HUVECs, thereby accelerating cellular proliferation, migration, angiogenesis, and resistance to ferroptosis. W. Nie et al. [231]
hUC-MSCs Exosomes derived from MSCs inhibit the expression of NETs, which, in turn, regulate ferroptosis by suppressing the PI3K/AKT signaling pathway. W. Lu et al. [232]
BM-MSCs Exosomes derived from BM-MSCs carry miR-367-3p, which binds to EZH2 and inhibits its expression, resulting in the overexpression of SLC7A11. This overexpression of SLC7A11 subsequently leads to the activation of GPX4 and the suppression of ferroptosis. J. Fan et al. [233]
hUC-MSCs Exosomes derived from hUC-MSCs utilize miR-129-5p to target ACSL4, thereby inhibiting LPO and ferroptosis. This inhibition significantly alleviates inflammation and promotes tissue repair. Z. Wei et al. [234]
Regulation of signaling pathways BM-MSCs The secretion of various growth factors upregulates the expression levels of VEGF-1α, MMP-9, pAKT, and VEGF-r in HUVECs, thereby activating the VEGF/AKT signaling pathway. This activation enhances HUVEC proliferation, migration, and tube formation capabilities while inhibiting apoptosis. J. Liu et al. [235]
BM-MSCs MSC-VEGF-CM activates the PI3K/AKT/mTOR/eNOS and p38/MAPK signaling pathways, thereby enhancing the activity and migratory capacity of pancreatic microvascular endothelial cells. T. Xu et al. [236]
BM-MSCs Significantly enhances the migratory and proliferative capabilities of human umbilical vein endothelial cells through the activation of the AKT signaling pathway. C. Hou et al. [237]
BM-MSCs Mechanism by which the PI3K/Akt signaling pathway regulates cell activity, proliferation, migration capabilities, and vascularization functions. J. Chen et al. [238]
hUC-MSCs The hUC-MSCs-CCLDADM scaffold material promotes wound healing in diabetic rats through the Wnt signaling pathway. Y. Han et al. [88]
AF-MSCs Increasing endogenous vascular growth factor levels enhances the regenerative potential of epithelial cells. Under hypoxic conditions, AF-MSCs enhance the proliferation and migration abilities of dermal fibroblasts through the TGF-β/SMAD2 and PI3K/AKT signaling pathways, thereby promoting wound healing. E. K. Jun et al. [240]
ADSCs Combined with PRP therapy, modulation of the Notch signaling pathway promotes angiogenesis and the proliferation of epidermal stem cells, thereby significantly accelerating wound healing. N. Ebrahim et al. [241]
Recruitment of other cells BM-MSCs It reduces excessive ROS production mediated by HG and/or LPS. Additionally, it effectively regulates the phosphorylation of MEK1/2 and ERK1/2 induced by high glucose and/or LPS, thereby enhancing the proliferation and migration abilities of keratinocytes under hyperglycemic conditions. M. Li et al. [242]
BM-MSCs Activation of the β2-AR signaling pathway enhances the migration of keratinocytes around the wound site, thereby promoting ulcer healing. J. Huo et al. [243]
BM-MSCs Exosomes released by MSCs induce the expression of HGF, IGF1, NGF, and SDF1 while simultaneously activating the Akt, ERK, and STAT3 signaling pathways, thereby enhancing fibroblast proliferation and migration capabilities. A. Shabbir et al. [244]
Secretion of growth factors ADSCs Release of TGF-β1 and HGF promotes the differentiation of myofibroblasts and the synthesis of collagen types I and III. J. Ma et al. [245]
USCs The secretion of PGE2 inhibits the differentiation of CD4+ T cells into Th17 cells. C. Zhou et al. [247]
BM-MSCs, ADSCs The secretion of anti-inflammatory cytokines promotes the polarization of macrophages from M1 to M2 type, thereby accelerating the wound healing process. M. El-Sayed et al. [248]
Production of antimicrobial peptides BM-MSCs The interaction between MSCs and AMP influences the proliferation, migration, and regeneration of MSCs. A. Silva-Carvalho et al. [249]
Release of apoptotic vesicles BM-MSCs The release of ABs from mesenchymal stem cells promotes macrophage polarization towards the M2 phenotype, thereby facilitating skin wound healing. J. Liu et al. [254]

9.1. Regulation of keratinocyte function

Keratinocytes play a pivotal role in the wound healing process. MSCs possess the capability to regulate the functionality of keratinocytes, thereby augmenting their contribution to epidermal formation at the wound site. Research indicates that the administration of BM-MSCs significantly enhances the proliferative capacity of endothelial cells and keratinocytes in mouse wounds. Additionally, MSCs promote the migration of endothelial cells, keratinocytes, and macrophages to the site of injury, thereby fostering wound healing [152]. Administration of BM-MSCs to wounds in genetic diabetic db/db mice results in elevated levels of angiopoietin-1, VEGF, and keratinocyte-specific protein keratin at the wound site. BM-MSCs effectively stimulate the proliferation of keratinocytes at the wound site. Moreover, hUC-MSCs demonstrate robust targeting specificity; in diabetic foot ulcer animal models, hUC-MSCs exhibit precise targeting to the injured site, stimulating the growth of cellular keratin 19 and consequently augmenting the regenerative potential of the wound epithelium, thereby fostering wound healing [153]. Furthermore, studies have reported that culturing human keratinocytes in BM-MSCs conditioned medium containing high glucose levels results in elevated levels of phosphorylated focal adhesion kinase, matrix metalloproteinase-2, EGF, and insulin-like growth factor 1. These outcomes suggest that BM-MSCs can ameliorate keratinocyte functionality, thereby fostering wound healing in a diabetic foot rat model [154].

9.2. Promote angiogenesis

Through exploration of the difficulties in DFU healing, it becomes apparent that local vascular injury and pathology contribute to the development of diabetic foot. Interventions aimed at addressing this cause, thereby enhancing neovascularization at the site of the lesion, hold immense importance for ulcer healing and repair. Upon local tissue injury, MSCs exhibit migratory tendencies toward the injured site and undergo targeted differentiation into endothelial and smooth muscle cells. Upon differentiation, they integrate into the damaged vasculature, thereby facilitating neovascularization. Furthermore, MSCs promote vascular formation through their paracrine activity, secreting various cytokines including VEGF, basic fibroblast growth factor (bFGF), stromal cell-derived factor-1 (SDF-1), hypoxia-inducible factor-1 (HIF-1), keratinocyte growth factor-2 (KGF-2), epidermal growth factor (EGF), and insulin-like growth factor-1 (IGF-1). These factors facilitate the production of ECM while suppressing endothelial cell apoptosis, thereby fostering vascular formation and wound healing [24,28,155]. Among these cytokines, VEGF plays a pivotal role [156]. Research indicates that the localized administration of VEGF can mobilize bone marrow-derived cells while concurrently promoting growth factors conducive to tissue repair. By recruiting these cells to the site of local injury, wound healing at the local wound site can be facilitated. Consequently, VEGF can be utilized in the treatment of diabetic complications characterized by vascular impairment [157].

Extensive research has demonstrated the angiogenic potential of MSC-derived exosomes [[158], [159], [160]]. Zhang et al. developed a full-thickness skin injury model in diabetic mice to investigate the role of exosomes secreted by ADSCs in diabetic wound healing. The results confirmed that treatment with ADSC-derived exosomes led to a significant increase in the expression levels of Angiopoietin 1 (ANG1), Fetal Liver Kinase 1 (FILK1), and VEGF, while the expression levels of endogenous angiogenesis inhibitors Angiopoietin 1 (VASH1) and Thrombospondin 1 (TSP1) were decreased. This finding validates that ADSC-derived exosome treatment significantly enhances endothelial cell activity and angiogenic potential, thereby contributing to the accelerated healing of diabetic wounds in mice [161]. Xue et al. demonstrated that exosomes derived from ADSCs overexpressing Nrf2 enhance endothelial progenitor cell proliferation and angiogenesis by promoting the phosphorylation of SMP30, VEGF, and VEGFR2, thereby accelerating the healing of diabetic wounds [162]. Yan et al. confirmed that exosomes secreted by hUC-MSCs significantly inhibit oxidative stress induced by high glucose in HUVECs, thereby promoting HUVEC activity, proliferation, and angiogenesis. This finding offers a novel approach for the repair of chronic diabetic wounds [163]. Circular RNA (CircRNA) represents a novel class of non-coding RNA characterized by the covalent linkage between the 5′ and 3′ ends. This unique circular structure imparts greater stability compared to linear RNA, and CircRNA effectively regulates the pathogenesis of various diseases [164]. Liang et al. demonstrated that circHIPK3, present in exosomes secreted by hUC-MSCs, upregulates the expression levels of Nrf2 and VEGFA while inhibiting miR-20b-5p [165]. Furthermore, overexpression of circHIPK3 significantly suppresses the expression levels of miR-124 in endothelial cells, thereby protecting these cells from high glucose-induced damage. Additionally, overexpression of circHIPK3 confers protective effects against cellular damage, significantly reducing apoptosis and inflammation while promoting angiogenesis [165]. Moreover, studies have shown that exosomes can be internalized by HUVECs in vitro, significantly enhancing their proliferation and migration capabilities. Building upon this, exosomes promote angiogenesis by activating the extracellular signal-regulated kinase 1/2 (Erk-1/2) signaling pathway, thereby effectively facilitating the healing process of skin wounds in diabetic rats [166].

9.3. Promotes collagen deposition

MSCs and their derived exosomes exhibit significant capacity to promote re-epithelialization and collagen deposition. The heat shock protein (HSP) family comprises a group of highly conserved proteins that protect the organism from various stress-induced damages. Ren et al. demonstrated that HSP90 in exosomes derived from ADSCs binds to the low-density lipoprotein receptor-related protein 1 (LRP1) on the recipient cell membrane, thereby activating the downstream AKT signaling pathway. This interaction enhances the proliferation and migration abilities of keratinocytes and fibroblasts, thereby accelerating the healing of diabetic wounds [167]. The TGF-β/Smad3 pathway is a well-established signaling pathway closely associated with collagen production and tissue fibrosis. TGF-β activates the Smad3 complex by binding to receptors on fibroblasts, which in turn promotes nuclear transcription and facilitates the synthesis of collagen and α-smooth muscle actin (α-SMA) [168]. Hsu et al. demonstrated that exosomes derived from ADSCs stimulate monocytes or macrophages to secrete increased levels of TGF-β1, which subsequently activates the TGF-β/Smad3 pathway, thereby promoting fibroblast proliferation and enhancing the production of type I collagen in diabetic wounds. This application provides a novel cell-free therapeutic approach for accelerating the healing of diabetic wounds [169]. Wang et al. demonstrated that ADSCs secrete a higher amount of exosomes (hypADSCs-exo) under hypoxic conditions, which activate the PI3K/AKT signaling pathway to enhance fibroblast proliferation, migration, and ECM secretion [170].

9.4. Promoting cell proliferation

Studies have demonstrated that BM-MSC-derived products promote wound closure in diabetic mice by facilitating re-epithelialization, dermal-epidermal junction restoration, skin appendage regeneration, inflammatory infiltration, vascularization, granulation tissue formation, and dense collagen deposition. Simultaneously, these products concentrate regenerative factors and proteins that further enhance wound healing. This effect surpasses that achieved by BM-MSCs alone in terms of therapeutic efficacy [171]. Moreover, the proliferative effects of ADSCs have been extensively validated. Exosomes derived from ADSCs facilitate the proliferation and migration of fibroblasts while enhancing collagen synthesis, thereby significantly accelerating the healing process of skin wounds in mice [172]. Additionally, ADSCs can promote wound repair by facilitating the formation of new blood vessels, protecting ischemic/reperfusion flaps, and enhancing IL-6 levels at the injury site [173]. Furthermore, exosomes derived from MSCs obtained from induced pluripotent stem cells (iPSCs) promote wound healing in rats by enhancing fibroblast proliferation and migration, increasing the synthesis of collagen and elastin, and stimulating the formation of capillary networks in vitro [174].

9.5. Antiapoptotic effect

The anti-apoptotic effects of MSCs and their derived exosomes have been confirmed by numerous studies. Exosomes derived from BM-MSCs inhibit apoptosis in epithelial HaCaT cells by activating apoptotic protease-activating factor 1 (APAF1), which facilitates wound healing [175]. Exosomes secreted by placenta-derived MSCs (hP-MSCs) not only enhance proliferation and anti-inflammatory capabilities but also inhibit apoptosis of corneal epithelial cells, thereby improving corneal wound healing in mice [176]. Cyclin-dependent kinase 6 (CDK6) is an oncogenic kinase that functions as a cell cycle regulator. Research indicates that CDK6 may be closely associated with wound or injury healing [177,178]. Exosomes derived from ADSCs contain miR-10b, which targets PEA15 to enhance the expression of CDK6. This, in turn, promotes the proliferation and migration of H2O2damaged HaCaT cells and inhibits cell apoptosis, thereby facilitating skin wound healing [179]. Exosomes secreted by ADSCs modulate Wnt/β-catenin signaling, thereby enhancing HaCaT cell proliferation and migration while simultaneously inhibiting apoptosis. This mechanism plays a crucial role in skin wound healing [180].

9.6. Anti-inflammatory effect

Numerous studies have shown that MSCs and their secreted exosomes significantly suppress systemic inflammation, making them a focal point in wound repair research. The study conducted by Yan et al. revealed that exosomes derived from hUC-MSCs significantly suppress the expression levels of inflammatory cytokines such as IL-1β, IL-6, and TNF-α, as well as NOX1 and NOX in HUVECs under hyperglycemic conditions. This suppression, in turn, reduces oxidative stress and promotes cellular proliferation [163]. The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) plays a pivotal role in mitigating oxidative stress and additionally exerts its effects by modulating the activity of antioxidant enzymes mediated through the antioxidant response element (ARE) [181]. Diabetic wound sites frequently exhibit diminished Nrf2/ARE activity coupled with elevated oxidative stress levels [182]. Xue et al. have demonstrated that exosomes derived from ADSCs, which exhibit high expression of Nrf2, effectively suppress the expression levels of inflammation- and oxidative stress-related proteins in HaCaT cells, fibroblasts, and HUVECs. This suppression, in turn, promotes the formation of granulation tissue at the wound site, enhances angiogenesis, and increases the expression levels of growth factors, thereby significantly accelerating wound healing in diabetic rats [162]. Superoxide dismutase (SOD) is an essential antioxidant enzyme that scavenges harmful superoxide anion radicals, playing a pivotal role in maintaining cellular redox balance [183]. Zhang et al. have shown that exosomes secreted by ADSCs can modulate the SIRT3/SOD2 axis to inhibit hyperglycemia-induced oxidative stress, reduce ROS accumulation, mitigate mitochondrial dysfunction and inflammatory responses, and activate endothelial cells, thereby accelerating wound healing in diabetic models [161]. Furthermore, exosomes secreted by MSCs can exert anti-inflammatory effects by modulating macrophage polarization. Exosomes derived from BM-MSCs can accelerate wound healing by transferring miR-223, which targets human PBX/knotted 1 homeobox 1 (PKNOX1) to induce M2 macrophage polarization [184].

9.7. Regulation of macrophage polarization

As widely recognized, macrophages are pivotal in orchestrating the body's immune response. Induced by specific external cues, macrophages undergo polarization. This polarization aids in modulating the organism's inflammatory response, safeguarding against pathogen incursion, and orchestrating tissue repair mechanisms. Macrophage polarization encompasses classical M1 activation and alternative M2 activation. M1 macrophages are conventionally regarded as exerting pro-inflammatory effects during the early stages of the inflammatory response [185]. M1 macrophages are known to release pro-inflammatory cytokines like IL-1b, IL-12, iNOS, TNF, among others [186]. In contrast, M2 macrophages are associated with anti-inflammatory and wound healing properties. They secrete molecules such as IL-1, IL-10, TGF-β, arginase-1, CD206, chitinase 3-like protein 3 (also known as YM1), and resistin-like molecule-alpha (also known as Fizz1) [187]. These molecules play pivotal roles in combatting parasitic infections, facilitating tissue repair, promoting angiogenesis, and more. M1 and M2 macrophages do not exhibit complete antagonism; they can undergo mutual transformation under specific conditions. During the healing process of diabetic ulcers, macrophages secrete a plethora of pro-inflammatory cytokines under sustained high-level glucose stimulation, resulting in the sustained M1 phenotype of macrophages at the ulcer site [188]. Hence, promoting the polarization of M1 macrophages to M2 macrophages at the site of injury might potentially serve as an effective therapeutic approach for treating diabetic ulcers.

As reported, hUC-MSCs have been reported to exert a modulatory effect on macrophage polarization, thereby ameliorating pancreatic dysfunction in type 2 diabetic mice [189,190]. Numerous studies have indicated that intravenous infusion of BM-MSCs and jaw bone-derived mesenchymal stem cells (JM-MSCs) can selectively migrate to the injured site, while also promoting the polarization of M1 macrophages into M2 macrophages, thus facilitating wound healing. In vitro experiments have also demonstrated that co-culturing MSCs with macrophages can promote their polarization into M2 macrophages [191]. UC-MSCs possess the ability to modulate macrophage polarization, thereby facilitating the recovery of endothelial cell function in diabetic mice and expediting wound healing processes [155]. Mesenchymal stem cells derived from human gingiva have the capability to induce macrophage polarization, thus playing a crucial role in the wound healing process [24]. Prostaglandin E2 (PGE2) secreted by hUC-MSCs has the capacity to modulate the expression levels of IL-10 and VEGF in the local microenvironment of endothelial cells. This induction of M2 polarization in macrophages infiltrating local diabetic defects fosters wound healing processes [155].

At the site of injury, skin cells are exposed to various danger signals, including damage-associated molecular patterns (DAMPs) and pathogen-associated molecular patterns (PAMPs). These signals are recognized by the immune system, triggering the initiation of the inflammatory process [192]. Recent studies have demonstrated that exosomes derived from ADSCs promote macrophage polarization from the M1 to M2 phenotype by delivering miR-21, which targets the PI3K/Akt pathway, thereby directly enhancing the pro-angiogenic effects of exosomes [193]. Similarly, He et al. have also shown that exosomes secreted by BM-MSCs promote macrophage polarization towards the M2 phenotype through the delivery of miR-223, a mechanism that accelerates wound healing [184]. Zhang et al. demonstrated that MSC-derived exosomes significantly enhance the infiltration of M2 macrophages and sustain this effect within cartilage, thereby contributing to the overall therapeutic impact. Following MSC-derived treatment, the expression levels of M1 macrophages and inflammation-related cytokines were significantly reduced [194]. These studies collectively demonstrate that microRNAs in MSC-derived exosomes may serve as critical therapeutic targets in tissue repair processes. Furthermore, exosomes secreted by BM-MSCs promote the polarization of macrophages to an M2-like phenotype, effectively modulating tissue repair and inflammatory responses in a tendon injury model, thereby facilitating tendon healing [195].

9.8. Inhibition of ROS generation

In a state of low ROS content, the body facilitates cell proliferation and differentiation, whereas elevated ROS levels stimulate the immune response, resulting in cell damage and dysfunction [196]. When tissues are damaged by various external stimuli, phagocytic cells in the body ingest foreign pathogens, cellular debris, and apoptotic inflammatory cells to eliminate pathogens. During this process, long-lived neutrophils produce abundant ROS after phagocytosis, initiating a respiratory burst, ultimately leading to tissue damage. Recent studies have demonstrated that MSCs alleviate protein oxidation and lipid peroxidation through their paracrine secretion of exosomes [197]. Moreover, MSCs display notable mitigating effects on various inflammatory conditions and pathological oxidative stress states. MSCs suppress myeloperoxidase, induce inducible nitric oxide synthase, and concurrently decrease the expression of inflammatory cytokines, including IL-1b, IL-4, IL-6, IL-9, TNF-α, and IFN-g [198]. Moreover, MSCs suppress the expression of ROS and myeloperoxidase elicited by monocytes and macrophages upon stimulation, consequently mitigating the M1 macrophage expression [199]. Additionally, MSCs augment the expression of stanniocalcin (STC), which diminishes ROS generation in macrophage mitochondria and restrains the activation of nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) and caspase-1 in these cells, thereby influencing the transcription of TNF-α and IL-6, and the expression of IL-1β [200].

Studies demonstrate that exosomes released by MSCs post hypoxic preconditioning exert regulatory effects on the accumulation of ROS in epithelial cells via HIF-1α. This process concurrently prevents DNA damage, preserving immune homeostasis, and ultimately mitigates the damage induced by ulcerative colitis [201]. Furthermore, the transplantation of MSCs improves the GP91/ROS/inflammasome signaling in diabetic cardiomyopathy while enhancing the expression of antioxidant proteins [202]. Studies have documented that exosomes released by human MSCs exert a significant inhibitory effect on glucocorticoid-induced apoptosis of osteoblasts through the modulation of the MAPK pathway and regulation of ROS levels [203]. Additionally, it has been demonstrated that human MSCs possess the capability to modulate the innate immune response through the ROS/NLRP3 signaling pathway, thereby mitigating the severity of MHV-68 pneumonia [204]. Considering the antioxidative and immunomodulatory properties of MSCs, their application in clinical therapy for DFU offers a plethora of possibilities, thereby expanding the horizons for DFU treatment.

9.9. Suppression of pro-inflammatory T cells and regulatory Treg cells

Certain chemokines and their corresponding receptors within the organism serve to regulate wound healing processes, orchestrating functions such as epithelialization, angiogenesis induction, and tissue remodeling [205]. The chemokine receptor CCR4, accompanied by its ligands CCL17 (TARC/Thymus and Activation Regulated Chemokine) and CCL22 (MDC/Macrophage-Derived Chemokine), intricately modulates the recruitment process of T cells (Tregs) [206]. Tregs, constitutively expressing the transcription factor Foxp3-, within the CD4+ T cell subset, are deemed pivotal in immune surveillance [207]. Treg cells constitute a substantial fraction of T cells within human skin and play a suppressive role in the body's autoimmune responses [208]. In instances of local inflammatory reactions, Treg cells migrate to the inflamed site, where they enact suppression upon both innate and adaptive immune responses [209]. Research has indicated that the chemokine receptor CCR4 inhibits the wound healing process, potentially attributed to the recruitment of Tregs to the wound site [207]. Moreover, the Th1 and Th17 subsets of helper T cells orchestrate inflammation in the pathogenesis of inflammatory diseases [210]. MSCs possess the capacity to suppress the differentiation and functionality of pro-inflammatory T cells, including Th17 cells, while inducing a regulatory T cell phenotype [211], thus efficiently modulating inflammation at the injury site. This property of MSCs introduces a novel therapeutic approach for the healing of DFU.

9.10. Regulation of neutrophil extracellular trapping networks (NETs) formation

Neutrophils, the initial innate immune cells recruited to the injury site during wound healing, eliminate pathogens and necrotic material via phagocytosis, degranulation, and the release of Neutrophil Extracellular Traps (NETs). NETs, composed of mesh-like DNA, histones, and various granular proteins, are released by activated neutrophils to limit bacterial spread and kill pathogens, a process considered a protective mechanism [212]. However, accumulating evidence indicates that excessive NET formation may contribute to tissue damage [213]. The release of NETs is stringently dependent on glucose and partially reliant on glycolysis [214]. In 2015, Wong et al. were the first to propose that diabetes markedly enhances NET formation in wounds, consequently delaying wound healing in diabetic patients [215].

Current research has confirmed that MSCs and their derived exosomes have a significant regulatory effect on the release of NETs. Upon activation, MSCs engage with the host's innate immune response, which encompasses the induction of NET formation and the augmentation of bacterial phagocytosis. When MSCs are used in conjunction with antibiotic therapy, the administration of activated MSCs into mice with established Staphylococcus aureus biofilm infections results in a marked reduction in bacterial load at the wound site, thus facilitating wound healing [216]. hAM-MSCs mediate their immunomodulatory effects via prostaglandin E2 (PGE2), effectively inhibiting NET release. This discovery unveils a novel mechanism through which hAM-MSCs modulate innate immune responses and suppress NET release [217]. Intravenous administration of mesenchymal stem cell-derived exosomes can significantly mitigate spinal cord injury, a process mediated through the regulation of NET formation by miR-125a-3p contained within the exosomes [218]. Luo et al.'s research was the first to observe that the terminal complement activation complex C5b-9 can initiate NET release and stimulate interleukin-17 production. Conversely, exosomes secreted by MSCs can inhibit the assembly of C5b-9 and subsequently suppress neutrophil-induced NET and IL-17 production [219].

9.11. Inhibition of ferroptosis

Ferroptosis has been closely implicated in the pathogenesis of various diseases. Evidence suggests a strong correlation between the progression of type 2 diabetes and iron overload [220]. As previously discussed, keratinocytes play a critical role in promoting epithelial regeneration during wound healing. Research indicates that the inhibition of ferroptosis in keratinocytes can markedly reduce the expression of inflammatory cytokines such as TNF-α and IL-6, thereby effectively mitigating inflammation in psoriasis [221]. During the resolution phase of inflammation, M1 macrophages gradually transition into M2 macrophages, thereby facilitating wound healing. Zhou et al.'s research revealed that iron overload markedly upregulates the expression of M1 macrophage markers, including IL-1β, TGF-α, and CD86, whereas the expression levels of M2 macrophage markers such as arginase-1 (Arg-1) and CD206 remain largely unaffected. This observation underscores the strong link between iron overload and macrophage polarization. Further mechanistic insights suggest that iron overload substantially promotes ROS release and concurrently activates p53 acetylation, thereby inducing M1 macrophage polarization [222]. High-mobility group box 1 (HMGB1) is a non-histone chromatin-binding protein present in the nuclei of eukaryotic cells, playing a pivotal role in DNA damage repair and the maintenance of genomic stability [223]. Ferroptotic cells can trigger the release of HMGB1, thereby promoting the polarization of pro-inflammatory macrophages [224]. Vascular complications represent another critical factor contributing to delayed wound healing in diabetes. Evidence suggests that iron overload in endothelial cells leads to increased ROS production, which is accompanied by a marked release of the chemokine monocyte chemoattractant protein-1 (MCP-1). Elevated MCP-1 levels enhance the recruitment of monocytes. These monocytes subsequently differentiate into macrophages, which engulf oxidized low-density lipoprotein (Ox-LDL), ultimately transforming into foam cells and exacerbating atherosclerosis [225]. Ferrostatin-1 (Fer-1), a well-known ferroptosis inhibitor, prevents ferroptosis by mitigating lipid peroxidation. Bai et al. discovered that endothelial nitric oxide synthase (eNOS) expression levels were significantly elevated in Fer-1-treated mice. eNOS plays a pivotal role in NO synthesis, thereby promoting vasodilation, inhibiting platelet activation, and reducing immune cell adhesion, which collectively mitigate systemic inflammation [226]. Furthermore, investigations on endothelial cells and fibroblasts cultured under high glucose conditions reveal a reduction in VEGF expression levels in endothelial cells, inhibition of fibroblast migration, and a decrease in activity in both cell types. The diminished ability of endothelial cells to support fibroblast synthesis under high glucose conditions can be restored by Fer-1 [227]. Moreover, Fer-1 facilitates a decrease in ferroptosis-related markers GXP4 and SLC7A11 in endothelial cells and fibroblasts under high glucose conditions, while enhancing TFR1 expression at both the transcriptional and translational levels [227]. Administration of Fer-1 significantly accelerates wound healing in diabetic rats [228]. Deferoxamine (DFO) is a clinically utilized iron chelator for treating iron overload, demonstrated to reduce oxidative stress and stimulate hypoxia-inducible factor-1 alpha (HIF-1α) activation, thereby promoting angiogenesis and improving chronic wound healing [229]. Duscher et al.'s study employing an advanced drug delivery system (TDDS) for delivering DFO to the skin demonstrated significant improvements in wound healing rates in diabetic mice, accompanied by increased neovascularization and enhanced dermal thickness [230].

Research has shown that miRNA-17-92 is highly expressed and enriched in exosomes secreted by MSCs. This miRNA promotes proliferation and migration of HUVECs in vitro, accelerating cell proliferation, migration, angiogenesis, and resistance to ferroptosis. Knockdown of miR-17-92 significantly diminishes its effect on wound healing. These findings indicate that miR-17-92 in MSC-derived exosomes plays a crucial role in protecting HUVECs from ferroptosis [231]. Additionally, exosomes derived from MSCs significantly inhibit the expression of NETs. NETs, in turn, regulate ferroptosis by inhibiting the PI3K/AKT pathway, thereby significantly enhancing diabetic wound healing [232]. Additionally, in vitro studies have shown that BMSC-derived exosomes carrying miR-367-3p significantly inhibit ferroptosis in microglia. Mechanistic analysis reveals that miR-367-3p binds to enhancer of zeste homolog 2 (EZH2) and inhibits its expression, leading to the overexpression of solute carrier family 7 member 11 (SLC7A11). Moreover, the overexpression of SLC7A11 leads to the activation of glutathione peroxidase 4 (GPX4) and suppression of ferroptosis [233]. Exosomes derived from hUC-MSCs inhibit lipid peroxidation (LPO) and ferroptosis through miR-129-5p targeting of acyl-CoA synthetase long-chain family member 4 (ACSL4), thereby significantly reducing inflammation and promoting tissue repair [234].

9.12. Regulation of signaling pathways

Various signaling pathways have been reported to facilitate the pathophysiological mechanisms regulating DFU, thereby promoting its healing. BM-MSCs secrete a variety of growth factors including VEGF-1α, ANG, HIF-1α, and MMP-9. These growth factors elevate the expression levels of VEGF-1α, MMP-9, pAKT, and VEGF-R in endothelial cells (HUVECs), thus activating the VEGF/AKT signaling pathway. This activation leads to enhanced proliferation, migration, and tube formation capabilities of HUVECs, while concurrently suppressing their apoptosis [235]. Treatment with MSC-conditioned medium engineered to overexpress VEGF (MSC-VEGF-CM) activates the PI-3K/AKT/mTOR/eNOS and p38/MAPK signaling pathways. This activation leads to enhanced activity and migratory capacity of pancreatic islet microvascular endothelial cells [236]. The utilization of conditioned medium derived from BM-MSCs markedly enhances the migratory and proliferative capabilities of human umbilical vein endothelial cells. Importantly, this process has been experimentally verified to operate independently of the extracellular signal-regulated kinase (ERK) signaling pathway, with the AKT signaling pathway playing a pivotal role [237]. The protein-serine-threonine kinase (AKT) stands out as a ubiquitous serine/threonine kinase, assuming a central role as a signaling hub across diverse cellular functions in the organism. The Pi3-dependent activation of AKT emerges as a regulatory mechanism governing the activity, proliferation, migratory prowess, and vascularization functionality of MSCs, underscoring the pivotal role this pathway plays in the modulation of MSCs behavior [238]. Given its remarkable evolutionary conservation, the Wnt signaling pathway assumes a pivotal role in embryonic development and the intricate processes of stem cell differentiation. Indeed, mounting evidence underscores the correlation between dysregulated Wnt signaling and the pathogenesis of diverse diseases [239]. The investigation implemented hUC-MSCs-CCLDADM scaffold materials on skin ulcer wounds in both normal and diabetic rats, accompanied by the administration of a Wnt signaling agonist or antagonist at the scaffold periphery. The findings demonstrated a notable enhancement in the healing speed of diabetic rat skin wounds upon activation of the Wnt signaling pathway, whereas the suppression of this pathway resulted in a substantial deceleration in wound healing [88]. Amniotic fluid-derived MSCs (AF-MSCs) notably elevate the levels of endogenous vascular growth factors, thereby fostering wound healing, while also substantially augmenting the regenerative potential of epidermal cells. Under hypoxic conditions, AF-MSCs bolster the proliferation and migration capacities of dermal fibroblasts via the transforming growth factor-β (TGF-β)/SMAD2 and PI3K/AKT signaling pathways, thereby fostering the process of wound healing [240]. Studies have demonstrated that the combined application of PRP and rat ADSCs in treating diabetic wounds in rats significantly accelerates wound healing by modulating the Notch signaling pathway to enhance angiogenesis and the proliferation of epidermal stem cells [241].

9.13. Recruitment of other cells

MSCs facilitate the recruitment of other cells to the site of injury, thereby fostering wound healing, a crucial hallmark of their function. There is ample evidence from various studies supporting this pivotal role of MSCs. Treatment of rat keratinocytes with mesenchymal stem cell-conditioned medium leads to a significant reduction in ROS overproduction mediated by HG and/or LPS. Furthermore, it demonstrates effective regulation of the HG and/or LPS-induced phosphorylation of MEK1/2 and Erk/2, consequently enhancing the proliferation and migratory capacities of keratinocytes under high-glucose environments [242]. The in vitro application of MSCs also facilitates ulcer healing by activating the β2-AR signaling pathway to enhance the migration of keratinocytes around the wound site [243]. Moreover, exosomes released by MSCs activate the Akt, ERK, and STAT3 pathways while inducing the expression of HGF, IGF1, NGF, and SDF1, thereby promoting the proliferation and migratory abilities of fibroblasts. Furthermore, it stimulates angiogenesis, ultimately contributing to the facilitation of chronic wound healing [244].

9.14. Secretion of growth factors

The process of wound healing is intricately woven, requiring the orchestrated interplay of diverse entities such as cells, proteins, growth factors, cytokines, and chemokines. MSCs possess the unique capability to regulate the microenvironment in damaged tissues, consequently fostering the restoration of injured tissues. In the intricate ballet of wound healing, growth factors, in addition to their direct roles in promoting the healing process, function as conductors harmonizing a symphony of cellular activities. The growth factors secreted by MSCs exert their effects through direct secretion or encapsulation within substances such as exosomes. MSCs have the capability to release factors including TGF-β1 and hepatocyte growth factor (HGF). TGF-β1 plays a pivotal role in promoting the differentiation of myofibroblasts and the synthesis of type I and type III collagens [245]. HGF has the ability to augment the expression of MMPs in fibroblasts, leading to the breakdown of ECM molecules such as collagen, laminin, and proteoglycans [246]. Additionally, MSCs have the capacity to secrete prostaglandin E2 (PGE2), which plays a role in inhibiting the differentiation of CD4+ T cells into Th17 cells [247]. MSCs secrete anti-inflammatory cytokines, thereby contributing to the process of wound healing. Moreover, MSCs release the anti-inflammatory factor IL-10, facilitating the transition of macrophages from the M1 phenotype to the M2 phenotype [248], thereby expediting the process of wound healing.

9.15. Production of antimicrobial peptides

MSCs possess the capacity to generate antimicrobial peptides (AMPs), exerting regulatory effects on immune responses and suppressing infections caused by pathogens. MSCs can secrete AMPs with anti-inflammatory activity against a spectrum of microorganisms, including bacteria, fungi, and viruses, comprising hepcidin, cathelicidin LL-37, and β-defensin-2 [249]. Hepcidin mainly functions in antimicrobial activity by reducing extracellular iron availability. This effect has been proven effective against various clinically relevant bacteria. LL-37 is an antimicrobial peptide known as "host defense peptide". It exhibits potent antimicrobial activity against both susceptible and antibiotic-resistant gram-positive and gram-negative bacteria [250]. Furthermore, LL-37 can exert antimicrobial effects by inhibiting the formation of biofilms and eradicating pre-existing biofilms, representing a principal antimicrobial mechanism of LL-37 [251]. LL-37 has the ability to facilitate the migration of immune cells to the site of infection and modulate the activity of immune cells, thus achieving pathogen clearance. β-defensin-2 can induce the substantial production of the classical anti-inflammatory cytokine IL-10 [252]. As reported, defensins can induce the conversion of CD4+T cells into Tregs, thereby exerting immunomodulatory effects [253]. In addition to the functions mentioned above, these AMPs also engage in interactions among MSCs, thus affecting the proliferation, migration, and regeneration of MSCs. These attributes of AMPs have made them a focal point of research in recent years.

9.16. Release of apoptotic vesicles

Research has demonstrated that apoptotic bodies (ABs) released by MSCs facilitate macrophage polarization towards the M2 phenotype, consequently enhancing skin wound healing [254].

10. Bioactive substances in MSCs-exo

Exosomes released by MSCs are capable of transporting a diverse array of bioactive substances over long distances within the body, thereby playing a pivotal role in cellular communication [255]. Exosomes contain a plethora of bioactive substances, including miRNA, lncRNA, circRNA, and proteins. These exosomes released by MSCs aid in the wound healing process of DFU by transporting these active factors(Table .3).

Table 3.

The role of bioactive molecules in MSC-derived exosomes in preclinical studies of DFU.

Source Researchers References
miRNA miR-21 UC-MSCs miR-21 enhances the proliferation and migration capabilities of corneal epithelial cells by inhibiting PTEN, subsequently upregulating the PI3K/Akt signaling pathway, thereby effectively facilitating corneal injury repair and regeneration./miR-21-5p enhances angiogenesis by upregulating VEGFR and activating the AKT and MAPK signaling pathways. Liu, X. et al./Huang, C. et al. [263,265]
miR-23 UC-MSCs miR-23 suppresses the excessive deposition of α-smooth muscle actin and collagen by inhibiting the TGF-β/SMAD2 signaling pathway. S. Fang et al. [267]
miR-126 SMSCs miR-126 enhances the proliferation of human dermal fibroblasts and HMEC-1, as well as the migration of HMEC-1, collagen and tubule formation. Tao, S.C. et al. [269]
miR-146 BM-MSCs miR-146a downregulates the target genes TRAF6 and IRAK1, leading to reduced expression levels of NF-kB, IL-6, and MIP-2, which in turn suppresses the local inflammatory response in the body. Xu, J. et al. [270]
miR-210-3p BM-MSCs miRNA-210-3p enhances the expression of the VEGF gene and activates the expression of pro-angiogenic proteins ERK and AKT, thereby exerting a role in improving microcirculation and promoting angiogenesis. Gangadaran, P. et al. [271]
miR-221-3p BM-MSCs miRNA-221-3p modulates the AKT/eNOS pathway, thereby enhancing the functional activity of endothelial cells. Yu, M. et al. [272]
miR-223 BM-MSCs, JMMSCs miR-223 induces M2 polarization of macrophages by targeting pknox1. He, X. et al. [273]
lncRNA
lncRNA H19 BM-MSCs, HF-MSCs lncRNA H19 orchestrates the healing process of DFU by modulating the PI3K/AKT signaling pathway and fostering a synergistic interplay between miR-152-3p and PTEN./lncRNA H19 inhibits the activation of the NLRP3 inflammasome, thus facilitating HaCaT proliferation, migration, and suppressing cell pyroptosis. Li, B. et al./Yang, H. et al. [274,275]
MALAT1
BM-MSCs
MALAT1 induces a reduction in the expression level of miR-205-5p and enhances the translation level of VEGF protein.
Zhu, L. et al.
[276]
circRNA mmu_circ_0000250 ADSCs mmu_circ_0000250 upregulates SIRT1 expression level while absorbing miR-128-3p. Shi, R. et al. [277]

10.1. miRNA

Within the spectrum of diabetes complications, miR-21 has garnered considerable attention [256]. Its involvement in the diabetic wound healing process spans across diverse mechanisms. Research indicates that miR-21, while reducing the expression levels of phosphatase and tensin homolog (PTEN), stimulates the PI3K/AKT signaling pathway, consequently enhancing dendritic cells (DCs) and facilitating full-thickness wound healing in rat models [257]. Moreover, under high-glucose conditions, cultured fibroblasts exhibit a direct interaction between the NF-κB p65 subunit and the miR-21 promoter upon TGF-β stimulation, facilitating fibroblast migration [258]. This discovery unveils novel therapeutic avenues for DFU management. Through RNA sequencing, studies have identified miRNA-21-5p, known for its capability to promote angiogenesis. Consequently, miRNA-21-5p emerges as a novel mechanism facilitated by MSC-exo to foster angiogenesis and repair ischemic tissue in diabetic foot [259]. Furthermore, the miR-23 family (miR-23a/b/c) targets Sprouty2 and Sema6A, consequently activating angiogenic signals and fostering vascular development within the organism [260]. Exosomes derived from UC-MSCs carrying miR-23 play an active role in suppressing the TGF-β/SMAD2 signaling pathway during the process of wound healing, illustrating a dynamic interplay between cellular components in tissue repair. This inhibition mitigates the excessive deposition of α-SMA and collagen, thereby contributing to the prevention of scar formation during the wound healing process [261]. miR-126 is pivotal in physiological blood vessel formation and inflammation regulation [262,263]. Exosomes derived from synovial mesenchymal stem cells (SMSCs), which overexpress miR-126, not only stimulate the proliferation of human dermal fibroblasts and human microvascular endothelial cells (HMEC-1) but also facilitate HMEC-1 migration, collagen synthesis, and lumen formation. Consequently, they promote the healing of full-thickness skin wounds in diabetic rats [264]. Furthermore, in the exosomes derived from BM-MSCs, the downregulation of miR-146a selectively targets TRAF6 and IRAK1, leading to a decrease in NF-κB, IL-6, and MIP-2 expression levels. This suppression effectively inhibits local inflammatory responses, thereby fostering diabetic wound repair [265]. Within exosomes originating from BM-MSCs, miRNA-210-3p amplifies the expression of the VEGF gene, consequently activating the expression of pro-angiogenic proteins ERK and AKT. Consequently, it facilitates the enhancement of microcirculation and the promotion of angiogenesis [266]. BM-MSCs have the capability to modulate the AKT/eNOS pathway via miRNA-221-3p contained within their exosomes, resulting in heightened functional activity of endothelial cells and contributing to the facilitation of wound healing in individuals with diabetes [267]. miR-223 within exosomes derived from BM-MSCs and JM-MSCs targets pknox1, leading to the polarization of macrophages toward the M2 phenotype, consequently enhancing the healing process of skin wounds [184]. The aforementioned research findings indicate that miRNAs originating from MSCs-exosomes play a pivotal role in facilitating wound healing throughout the DFU healing process.

10.2. lncRNA

Studies suggest that the long non-coding RNA H19 (lncRNA H19) intricately regulates hepatic glucose production and insulin resistance within a high-glucose microenvironment [268]. lncRNA H19 derived from MSCs-exosomes facilitates the healing of DFU by modulating the PI3K/AKT signaling pathway and orchestrating the reciprocal interplay between miR-152-3p and PTEN [269]. Moreover, exosomes derived from hair follicle mesenchymal stem cells (HF-MSCs-Exo) with elevated levels of lncRNA H19 exhibit the capability to dampen the stimulation of NLRP3 inflammasome, consequently fostering the proliferation, migration, and restraining the pyroptosis of HaCaT cells [270]. According to reports, the therapeutic efficacy of VEGF in treating DFU is improved upon the deletion of miR-205-5p within human mesenchymal stem cells. Additionally, lncRNA MALAT1 acts as a competitive endogenous RNA (ceRNA) for miR-205-5p. Consequently, the upregulation of MALAT1 results in a reduction in the expression levels of miR-205-5p and an elevation in the translation levels of VEGF protein, thereby enhancing wound healing in diabetic mice [271].

10.3. circRNA

Circular RNAs (circRNAs) play a pivotal role in modulating the microenvironment during wound healing. The vascular regeneration capacity is hindered under high-glucose conditions, resulting in delayed wound healing. Exosomes derived from ADSCs modified by mmu_circ_0000250 exhibit the ability to counteract this inhibitory effect. Exosomes derived from ADSCs, enriched with mmu_circ_0000250, efficiently sequester miR-128-3p and concomitantly elevate the expression level of SIRT1, consequently manifesting a therapeutic impact on enhancing the healing of diabetic wounds [272].

11. Pretreatment of exosomes

Recent investigations have demonstrated that pretreating MSCs with diverse approaches, including pharmacological agents, cytokines, and physical stimuli, can augment their biological activity and functionality in the context of tissue regeneration [273,278]. According to reports, excessive neutrophil extracellular trap (NET) formation was identified in diabetic wounds, highlighting the pivotal role of NETs in delayed wound healing. Notably, hypoxia-preconditioned MSC-derived exosomes demonstrated enhanced suppression of NETs compared to those secreted by normal MSCs, underscoring the superior advantage of hypoxia-preconditioned MSC-secreted exosomes in the field of regenerative medicine [274](Fig. 6). Furthermore, MSCs pre-treated with red ginseng augment diabetic wound healing by amplifying their paracrine activity [275]. Exosomes derived from pioglitazone-pretreated MSCs expedite diabetic wound healing by stimulating angiogenesis [276]. Exosomes secreted by melatonin-primed MSCs regulate macrophage polarization through the PTEN/AKT pathway, thereby enhancing diabetic wound healing [277]. Exosomes derived from BM-MSCs pre-treated with DMOG facilitate bone regeneration by targeting the AKT/mTOR pathway [279]. Pretreatment with fluoxetine enhances the effectiveness of MSCs in diabetic neuropathy [280]. Modulating the secretion profile of proteins from MSCs through pre-treatment represents a potential new avenue in the study of MSCs and their exosomes.

Fig. 6.

Fig. 6

Low-oxygen pre-treated exosomes expedite the healing of diabetic wounds by attenuating the excessive formation of neutrophil extracellular traps (NETs) in vivo. (a) Nanoparticle tracking analysis (NTA) was employed to evaluate the size and concentration distribution of Normo-sEVs and Hypo-sEVs derived from MSCs. (b) Representative micrographs of Normo-sEVs and Hypo-sEVs were obtained via transmission electron microscopy (TEM). (c) Western blot analysis was performed to evaluate the expression levels of sEV markers, including CD63, CD9, TSG101, and Calnexin. (d) Animals were intravenously injected with PBS, Normo-sEVs, or Hypo-sEVs, and representative images depicting wound closure were captured at days 0, 3, 7, and 14 post-injection, respectively. (e) Illustrative diagrams depicting the wound healing process. (f) Wound closure rates were quantitatively assessed; N = 6. (g) Representative images illustrating H&E staining at various time points of the wound sites were captured. The scale bar represents 2 mm. (h) Quantitative analysis was conducted to assess wound length; N = 6. (i) Immunofluorescence staining was employed to evaluate the NETs area in the wound sites of each group on the third day post-injection in the animal model (H3Cit: green; MPO: red; DAPI: blue); N = 6. The scale bar indicates 100 μm. Significance levels are denoted as **p < 0.01 and ***p < 0.001. Reproduced under the terms of the CC-BY license [274]. Copyright 2023, the authors.

12. Summary and outlook

In comparison to conventional therapeutic approaches, MSCs along with their secreted exosomes offer considerable advantages in the treatment of DFU. MSCs can facilitate DFU wound healing through diverse mechanisms, such as modulating macrophage polarization, optimizing the local microenvironment, and augmenting angiogenesis. These processes constitute a complex cascade involving multiple signaling pathways. Furthermore, the paracrine activity of MSCs, resulting in the secretion of a myriad of bioactive substances, also exerts a pivotal role in the wound healing process. Within the spectrum of bioactive substances secreted by MSCs, exosomes have been empirically shown to transport a broad array of materials, encompassing miRNA, lncRNA, circRNA, proteins, among others, across long distances within the body. This attribute also bestows considerable advantages in fostering DFU healing. Present conclusions from studies on MSCs and their secreted exosomes indicate substantial therapeutic promise in addressing DFU. Nonetheless, the utilization of MSCs and their exosomes in practice presents specific constraints and hurdles.

The differentiation potential, proliferation capacity, and reparative ability of MSCs diminish gradually with both increasing passage number and age. Hence, to optimize the advantageous application of MSCs, it is imperative for us to standardize their treatment in future research endeavors. Various clinical studies have assessed the safety and efficacy of MSCs in treating DFUs. Standardizing the therapeutic outcomes of MSCs prior to commencing clinical trials is paramount. Despite the preliminary confirmation of the efficacy and safety of MSCs therapy for DFU, there is a need for further investigation into treatment mechanisms, efficacy evaluation, individualized selection of MSCs, and application protocols. In conclusion, MSCs therapy presents a promising, relatively safe, and effective treatment modality for DFUs.

In comparison to the solitary use of MSCs-exo, the utilization of engineered exosomes, achieved through co-administration with biomaterial scaffolds or additional modifications to their surface and interior, exhibits enhanced efficacy in wound healing. This innovation also offers a novel avenue for ongoing investigations into MSCs-exo's role in promoting DFU recovery. Nonetheless, limitations exist in the application of MSCs-exo due to their susceptibility to expansion and inactivation at room temperature, as well as the low yields obtained during extraction. Presently, investigations into the efficacy of MSCs-exo for DFU primarily revolve around preclinical studies, including cell experiments and animal models. Notably, clinical trials validating the therapeutic potential of exosomes remain absent. To surmount the transition from preclinical to clinical application, future research endeavors should prioritize understanding the mechanistic underpinnings of MSCs-exo and instituting standardized production protocols. Although facing myriad challenges in the clinical application of MSCs-exo, the undeniable potential of MSCs and their exosomes in DFU therapy underscores the necessity for deeper exploration.

CRediT authorship contribution statement

ShuHui Wu: Writing – original draft, Conceptualization. ZhongSheng Zhou: Writing – original draft, Conceptualization. Yang Li: Investigation. Jinlan Jiang: Funding acquisition, Conceptualization.

Declaration of competing interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

References

  • 1.Tan S.Y., et al. Type 1 and 2 diabetes mellitus: a review on current treatment approach and gene therapy as potential intervention. Diabetes Metabol. Syndr. 2019;13(1):364–372. doi: 10.1016/j.dsx.2018.10.008. [DOI] [PubMed] [Google Scholar]
  • 2.Du S., Zeugolis D.I., O'Brien T. Scaffold-based delivery of mesenchymal stromal cells to diabetic wounds. Stem Cell Res. Ther. 2022;13(1):426. doi: 10.1186/s13287-022-03115-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Yamagishi S., Fukami K., Matsui T. Crosstalk between advanced glycation end products (AGEs)-receptor RAGE axis and dipeptidyl peptidase-4-incretin system in diabetic vascular complications. Cardiovasc. Diabetol. 2015;14:2. doi: 10.1186/s12933-015-0176-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Khalid M., Petroianu G., Adem A. Advanced glycation end products and diabetes mellitus: mechanisms and perspectives. Biomolecules. 2022;12(4) doi: 10.3390/biom12040542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Jais S. Various types of wounds that diabetic patients can develop: a narrative review. Clin Pathol. 2023;16 doi: 10.1177/2632010X231205366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Soria B., et al. Cell therapy of vascular and neuropathic complications of diabetes: can we avoid limb amputation? Int. J. Mol. Sci. 2023;24(24) doi: 10.3390/ijms242417512. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Raghav A., et al. Financial burden of diabetic foot ulcers to world: a progressive topic to discuss always. Ther Adv Endocrinol Metab. 2018;9(1):29–31. doi: 10.1177/2042018817744513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Lopes L., et al. Stem cell therapy for diabetic foot ulcers: a review of preclinical and clinical research. Stem Cell Res. Ther. 2018;9(1):188. doi: 10.1186/s13287-018-0938-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Pérez-Panero A.J., et al. Prevention, assessment, diagnosis and management of diabetic foot based on clinical practice guidelines: a systematic review. Medicine (Baltim.) 2019;98(35) doi: 10.1097/MD.0000000000016877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Huang Y.Z., et al. Mesenchymal stem cells for chronic wound healing: current status of preclinical and clinical studies. Tissue Eng., Part B. 2020;26(6):555–570. doi: 10.1089/ten.TEB.2019.0351. [DOI] [PubMed] [Google Scholar]
  • 11.Li Q., et al. Hyperglycemia-induced accumulation of advanced glycosylation end products in fibroblast-like synoviocytes promotes knee osteoarthritis. Exp. Mol. Med. 2021;53(11):1735–1747. doi: 10.1038/s12276-021-00697-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Zheng W., et al. Research advances on the damage mechanism of skin glycation and related inhibitors. Nutrients. 2022;14(21) doi: 10.3390/nu14214588. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rhee S.Y., Kim Y.S. The role of advanced glycation end products in diabetic vascular complications. Diabetes Metab J. 2018;42(3):188–195. doi: 10.4093/dmj.2017.0105. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Wasim R., et al. Aftermath of AGE-RAGE Cascade in the pathophysiology of cardiovascular ailments. Life Sci. 2022;307 doi: 10.1016/j.lfs.2022.120860. [DOI] [PubMed] [Google Scholar]
  • 15.Baltzis D., Eleftheriadou I., Veves A. Pathogenesis and treatment of impaired wound healing in diabetes mellitus: new insights. Adv. Ther. 2014;31(8):817–836. doi: 10.1007/s12325-014-0140-x. [DOI] [PubMed] [Google Scholar]
  • 16.Zierfuss B., et al. Saturated very long-chain fatty acids regulate macrophage plasticity and invasiveness. J. Neuroinflammation. 2022;19(1):305. doi: 10.1186/s12974-022-02664-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Eming S.A., Martin P., Tomic-Canic M. Wound repair and regeneration: mechanisms, signaling, and translation. Sci. Transl. Med. 2014;6(265):265sr6. doi: 10.1126/scitranslmed.3009337. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Pari B., et al. Insight on infections in diabetic setting. Biomedicines. 2023;11(3) doi: 10.3390/biomedicines11030971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Kruk J., et al. Oxidative stress in biological systems and its relation with pathophysiological functions: the effect of physical activity on cellular redox homeostasis. Free Radic. Res. 2019;53(5):497–521. doi: 10.1080/10715762.2019.1612059. [DOI] [PubMed] [Google Scholar]
  • 20.Eftekharpour E., Fernyhough P. Oxidative stress and mitochondrial dysfunction associated with peripheral neuropathy in type 1 diabetes. Antioxidants Redox Signal. 2022;37(7–9):578–596. doi: 10.1089/ars.2021.0152. [DOI] [PubMed] [Google Scholar]
  • 21.Chevalier J., et al. Obstruction of small arterioles in patients with critical limb ischemia due to partial endothelial-to-mesenchymal transition. iScience. 2020;23(6) doi: 10.1016/j.isci.2020.101251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Theocharidis G., Veves A. Autonomic nerve dysfunction and impaired diabetic wound healing: the role of neuropeptides. Auton. Neurosci. 2020;223 doi: 10.1016/j.autneu.2019.102610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Tellechea A., et al. Mast cells regulate wound healing in diabetes. Diabetes. 2016;65(7):2006–2019. doi: 10.2337/db15-0340. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yu X., et al. Function and mechanism of mesenchymal stem cells in the healing of diabetic foot wounds. Front. Endocrinol. 2023;14 doi: 10.3389/fendo.2023.1099310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Ramachandran V., et al. Physiological and pathophysiological aspects of diabetic foot ulcer and its treatment strategies. Curr. Diabetes Rev. 2023;19(8) doi: 10.2174/1573399819666221103141715. [DOI] [PubMed] [Google Scholar]
  • 26.Nourian Dehkordi A., et al. Skin tissue engineering: wound healing based on stem-cell-based therapeutic strategies. Stem Cell Res. Ther. 2019;10(1):111. doi: 10.1186/s13287-019-1212-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Wilkinson H.N., Hardman M.J. Wound healing: cellular mechanisms and pathological outcomes. Open Biol. 2020;10(9) doi: 10.1098/rsob.200223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Veith A.P., et al. Therapeutic strategies for enhancing angiogenesis in wound healing. Adv. Drug Deliv. Rev. 2019;146:97–125. doi: 10.1016/j.addr.2018.09.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Dalton C.J., Lemmon C.A. Fibronectin: molecular structure, fibrillar structure and mechanochemical signaling. Cells. 2021;10(9) doi: 10.3390/cells10092443. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Pensalfini M., Tepole A.B. Mechano-biological and bio-mechanical pathways in cutaneous wound healing. PLoS Comput. Biol. 2023;19(3) doi: 10.1371/journal.pcbi.1010902. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Larouche J., et al. Immune regulation of skin wound healing: mechanisms and novel therapeutic targets. Adv. Wound Care. 2018;7(7):209–231. doi: 10.1089/wound.2017.0761. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Liew P.X., Kubes P. The neutrophil's role during health and disease. Physiol. Rev. 2019;99(2):1223–1248. doi: 10.1152/physrev.00012.2018. [DOI] [PubMed] [Google Scholar]
  • 33.Hassanshahi A., et al. Macrophage-mediated inflammation in skin wound healing. Cells. 2022;11(19) doi: 10.3390/cells11192953. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Qiang L., et al. Keratinocyte autophagy enables the activation of keratinocytes and fibroblastsand facilitates wound healing. Autophagy. 2021;17(9):2128–2143. doi: 10.1080/15548627.2020.1816342. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Moretti L., et al. The interplay of fibroblasts, the extracellular matrix, and inflammation in scar formation. J. Biol. Chem. 2022;298(2) doi: 10.1016/j.jbc.2021.101530. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Papachristoforou E., et al. Association of glycemic indices (hyperglycemia, glucose variability, and hypoglycemia) with oxidative stress and diabetic complications. J. Diabetes Res. 2020;2020 doi: 10.1155/2020/7489795. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Olsen L., et al. A mathematical model for the capillary endothelial cell-extracellular matrix interactions in wound-healing angiogenesis. IMA J. Math. Appl. Med. Biol. 1997;14(4):261–281. [PubMed] [Google Scholar]
  • 38.Pastar I., et al. Molecular pathophysiology of chronic wounds: current state and future directions. Cold Spring Harbor Perspect. Biol. 2023;15(4) doi: 10.1101/cshperspect.a041243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Garima, et al. Oxygenation therapies for improved wound healing: current trends and technologies. J. Mater. Chem. B. 2022;10(39):7905–7923. doi: 10.1039/d2tb01498j. [DOI] [PubMed] [Google Scholar]
  • 40.Desmet C.M., Préat V., Gallez B. Nanomedicines and gene therapy for the delivery of growth factors to improve perfusion and oxygenation in wound healing. Adv. Drug Deliv. Rev. 2018;129:262–284. doi: 10.1016/j.addr.2018.02.001. [DOI] [PubMed] [Google Scholar]
  • 41.Bassiouni W., Ali M.A.M., Schulz R. Multifunctional intracellular matrix metalloproteinases: implications in disease. FEBS J. 2021;288(24):7162–7182. doi: 10.1111/febs.15701. [DOI] [PubMed] [Google Scholar]
  • 42.Ayuk S.M., Abrahamse H., Houreld N.N. The role of matrix metalloproteinases in diabetic wound healing in relation to photobiomodulation. J. Diabetes Res. 2016;2016 doi: 10.1155/2016/2897656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Serra R. Matrix metalloproteinases in health and disease. Biomolecules. 2020;10(8) doi: 10.3390/biom10081138. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Lin H., Xu P., Huang M. Structure-based molecular insights into matrix metalloproteinase inhibitors in cancer treatments. Future Med. Chem. 2022;14(1):35–51. doi: 10.4155/fmc-2021-0246. [DOI] [PubMed] [Google Scholar]
  • 45.Kandhwal M., et al. Role of matrix metalloproteinase in wound healing. Am J Transl Res. 2022;14(7):4391–4405. [PMC free article] [PubMed] [Google Scholar]
  • 46.Luo H.C., et al. Efficiency and safety of dextran-PAMAM/siMMP-9 complexes for decreasing matrix metalloproteinase-9 expression and promoting wound healing in diabetic rats. Bioconjug Chem. 2022;33(12):2398–2410. doi: 10.1021/acs.bioconjchem.2c00487. [DOI] [PubMed] [Google Scholar]
  • 47.Louiselle A.E., et al. Macrophage polarization and diabetic wound healing. Transl. Res. 2021;236:109–116. doi: 10.1016/j.trsl.2021.05.006. [DOI] [PubMed] [Google Scholar]
  • 48.Wu X., et al. Macrophage polarization in diabetic wound healing. Burns Trauma. 2022;10:tkac051. doi: 10.1093/burnst/tkac051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Wolf S.J., Melvin W.J., Gallagher K. Macrophage-mediated inflammation in diabetic wound repair. Semin. Cell Dev. Biol. 2021;119:111–118. doi: 10.1016/j.semcdb.2021.06.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Goggins E., et al. Reprogramming of VEGF-mediated extracellular matrix changes through autocrine signaling. Cancer Biol. Ther. 2023;24(1) doi: 10.1080/15384047.2023.2184145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Kucharzewski M., et al. Novel trends in application of stem cells in skin wound healing. Eur. J. Pharmacol. 2019;843:307–315. doi: 10.1016/j.ejphar.2018.12.012. [DOI] [PubMed] [Google Scholar]
  • 52.Li G., et al. A small molecule HIF-1α stabilizer that accelerates diabetic wound healing. Nat. Commun. 2021;12(1):3363. doi: 10.1038/s41467-021-23448-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Dissanayake A., et al. Does intensive glycaemic control promote healing in diabetic foot ulcers? - a feasibility study. BMJ Open. 2020;10(1) doi: 10.1136/bmjopen-2019-029009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Jia L., et al. Incidence and risk factors for developing infection in patients presenting with uninfected diabetic foot ulcers. PLoS One. 2017;12(5) doi: 10.1371/journal.pone.0177916. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Dayya D., et al. Debridement of diabetic foot ulcers. Adv. Wound Care. 2022;11(12):666–686. doi: 10.1089/wound.2021.0016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.I N.A., et al. Antibacterial-integrated collagen wound dressing for diabetes-related foot ulcers: an evidence-based review of clinical studies. Polymers. 2020;12(9) doi: 10.3390/polym12092168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Shi C., et al. Selection of appropriate wound dressing for various wounds. Front. Bioeng. Biotechnol. 2020;8:182. doi: 10.3389/fbioe.2020.00182. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Gao D., et al. Functional hydrogels for diabetic wound management. APL Bioeng. 2021;5(3) doi: 10.1063/5.0046682. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Li W., et al. Synthesis and fabrication of a keratin-conjugated insulin hydrogel for the enhancement of wound healing. Colloids Surf. B Biointerfaces. 2019;175:436–444. doi: 10.1016/j.colsurfb.2018.12.020. [DOI] [PubMed] [Google Scholar]
  • 60.Li X., et al. Functionalized silk fibroin dressing with topical bioactive insulin release for accelerated chronic wound healing. Mater. Sci. Eng., C. 2017;72:394–404. doi: 10.1016/j.msec.2016.11.085. [DOI] [PubMed] [Google Scholar]
  • 61.Zhu J., et al. Rapid gelation of oxidized hyaluronic acid and succinyl chitosan for integration with insulin-loaded micelles and epidermal growth factor on diabetic wound healing. Mater. Sci. Eng., C. 2020;117 doi: 10.1016/j.msec.2020.111273. [DOI] [PubMed] [Google Scholar]
  • 62.Zhao L., et al. pH and glucose dual-responsive injectable hydrogels with insulin and fibroblasts as bioactive dressings for diabetic wound healing. ACS Appl. Mater. Interfaces. 2017;9(43):37563–37574. doi: 10.1021/acsami.7b09395. [DOI] [PubMed] [Google Scholar]
  • 63.Zhao X., et al. Antibacterial anti-oxidant electroactive injectable hydrogel as self-healing wound dressing with hemostasis and adhesiveness for cutaneous wound healing. Biomaterials. 2017;122:34–47. doi: 10.1016/j.biomaterials.2017.01.011. [DOI] [PubMed] [Google Scholar]
  • 64.Zhu D.Y., et al. Injectable thermo-sensitive and wide-crack self-healing hydrogel loaded with antibacterial anti-inflammatory dipotassium glycyrrhizate for full-thickness skin wound repair. Acta Biomater. 2022;143:203–215. doi: 10.1016/j.actbio.2022.02.041. [DOI] [PubMed] [Google Scholar]
  • 65.Huang L., et al. Antibacterial poly (ethylene glycol) diacrylate/chitosan hydrogels enhance mechanical adhesiveness and promote skin regeneration. Carbohydr. Polym. 2019;225 doi: 10.1016/j.carbpol.2019.115110. [DOI] [PubMed] [Google Scholar]
  • 66.Yang Y., et al. Mussel-inspired adhesive antioxidant antibacterial hemostatic composite hydrogel wound dressing via photo-polymerization for infected skin wound healing. Bioact. Mater. 2022;8:341–354. doi: 10.1016/j.bioactmat.2021.06.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Cheng H., et al. Sprayable hydrogel dressing accelerates wound healing with combined reactive oxygen species-scavenging and antibacterial abilities. Acta Biomater. 2021;124:219–232. doi: 10.1016/j.actbio.2021.02.002. [DOI] [PubMed] [Google Scholar]
  • 68.Chen X., et al. In situ forming ROS-scavenging hybrid hydrogel loaded with polydopamine-modified fullerene nanocomposites for promoting skin wound healing. J. Nanobiotechnol. 2023;21(1):129. doi: 10.1186/s12951-023-01879-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Lin Z., et al. Histatin1-modified thiolated chitosan hydrogels enhance wound healing by accelerating cell adhesion, migration and angiogenesis. Carbohydr. Polym. 2020;230 doi: 10.1016/j.carbpol.2019.115710. [DOI] [PubMed] [Google Scholar]
  • 70.Mohandas A., et al. Chitosan-hyaluronic acid/VEGF loaded fibrin nanoparticles composite sponges for enhancing angiogenesis in wounds. Colloids Surf. B Biointerfaces. 2015;127:105–113. doi: 10.1016/j.colsurfb.2015.01.024. [DOI] [PubMed] [Google Scholar]
  • 71.Theocharidis G., et al. Murine macrophages or their secretome delivered in alginate dressings enhance impaired wound healing in diabetic mice. Biomaterials. 2022;288 doi: 10.1016/j.biomaterials.2022.121692. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Tan W.S., et al. Improvement of diabetic wound healing by topical application of Vicenin-2 hydrocolloid film on Sprague Dawley rats. BMC Complement Altern Med. 2019;19(1):20. doi: 10.1186/s12906-018-2427-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Everett E., Mathioudakis N. Update on management of diabetic foot ulcers. Ann. N. Y. Acad. Sci. 2018;1411(1):153–165. doi: 10.1111/nyas.13569. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Sahu B., Prusty A., Tudu B. Total contact casting versus traditional dressing in diabetic foot ulcers. J. Orthop. Surg. 2018;26(3) doi: 10.1177/2309499018802486. [DOI] [PubMed] [Google Scholar]
  • 75.Bus S.A., et al. Guidelines on offloading foot ulcers in persons with diabetes (IWGDF 2019 update) Diabetes Metab Res Rev. 2020;36(Suppl 1):e3274. doi: 10.1002/dmrr.3274. [DOI] [PubMed] [Google Scholar]
  • 76.Li H.Z., et al. Negative pressure wound therapy for surgical site infections: a systematic review and meta-analysis of randomized controlled trials. Clin. Microbiol. Infect. 2019;25(11):1328–1338. doi: 10.1016/j.cmi.2019.06.005. [DOI] [PubMed] [Google Scholar]
  • 77.Karam R.A., et al. Effect of negative pressure wound therapy on molecular markers in diabetic foot ulcers. Gene. 2018;667:56–61. doi: 10.1016/j.gene.2018.05.032. [DOI] [PubMed] [Google Scholar]
  • 78.Tejada S., et al. Therapeutic effects of hyperbaric oxygen in the process of wound healing. Curr Pharm Des. 2019;25(15):1682–1693. doi: 10.2174/1381612825666190703162648. [DOI] [PubMed] [Google Scholar]
  • 79.Sharma R., et al. Efficacy of hyperbaric oxygen therapy for diabetic foot ulcer, a systematic review and meta-analysis of controlled clinical trials. Sci. Rep. 2021;11(1):2189. doi: 10.1038/s41598-021-81886-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zhang Z., et al. Efficacy of hyperbaric oxygen therapy for diabetic foot ulcers: an updated systematic review and meta-analysis. Asian J. Surg. 2022;45(1):68–78. doi: 10.1016/j.asjsur.2021.07.047. [DOI] [PubMed] [Google Scholar]
  • 81.Moore M.C., et al. Biological characterization of dehydrated amniotic membrane allograft: mechanisms of action and implications for wound care. J. Biomed. Mater. Res. B Appl. Biomater. 2020;108(8):3076–3083. doi: 10.1002/jbm.b.34635. [DOI] [PubMed] [Google Scholar]
  • 82.Aamar E., et al. Hair-follicle mesenchymal stem cell activity during homeostasis and wound healing. J. Invest. Dermatol. 2021;141(12):2797–2807.e6. doi: 10.1016/j.jid.2021.05.023. [DOI] [PubMed] [Google Scholar]
  • 83.Shu X., et al. Efficiency of stem cell based therapy in the treatment of diabetic foot ulcer: a meta-analysis. Endocr. J. 2018;65(4):403–413. doi: 10.1507/endocrj.EJ17-0424. [DOI] [PubMed] [Google Scholar]
  • 84.Tanaka K., et al. Role and function of mesenchymal stem cells on fibroblast in cutaneous wound healing. Biomedicines. 2022;10(6) doi: 10.3390/biomedicines10061391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Shi R., et al. Role and effect of vein-transplanted human umbilical cord mesenchymal stem cells in the repair of diabetic foot ulcers in rats. Acta Biochim. Biophys. Sin. 2020;52(6):620–630. doi: 10.1093/abbs/gmaa039. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Liu Y., et al. Human umbilical cord-derived mesenchymal stem cells not only ameliorate blood glucose but also protect vascular endothelium from diabetic damage through a paracrine mechanism mediated by MAPK/ERK signaling. Stem Cell Res. Ther. 2022;13(1):258. doi: 10.1186/s13287-022-02927-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Dhoke N.R., Kaushik K., Das A. Cxcr6-Based mesenchymal stem cell gene therapy potentiates skin regeneration in murine diabetic wounds. Mol. Ther. 2020;28(5):1314–1326. doi: 10.1016/j.ymthe.2020.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Han Y., et al. Human umbilical cord mesenchymal stem cells implantation accelerates cutaneous wound healing in diabetic rats via the Wnt signaling pathway. Eur. J. Med. Res. 2019;24(1):10. doi: 10.1186/s40001-019-0366-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Wei L., et al. Mesenchymal stem cells promote wound healing and effects on expression of matrix metalloproteinases-8 and 9 in the wound tissue of diabetic rats. Stem Cell. Dev. 2023;32(1–2):25–31. doi: 10.1089/scd.2021.0218. [DOI] [PubMed] [Google Scholar]
  • 90.Rubio G.A., et al. Mesenchymal stromal cells prevent bleomycin-induced lung and skin fibrosis in aged mice and restore wound healing. J. Cell. Physiol. 2018;233(8):5503–5512. doi: 10.1002/jcp.26418. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Liu Z., et al. Human umbilical cord mesenchymal stem cells improve irradiation-induced skin ulcers healing of rat models. Biomed. Pharmacother. 2018;101:729–736. doi: 10.1016/j.biopha.2018.02.093. [DOI] [PubMed] [Google Scholar]
  • 92.Wang K., et al. LPS-pretreatment adipose-derived mesenchymal stromal cells promote wound healing in diabetic rats by improving angiogenesis. Injury. 2022;53(12):3920–3929. doi: 10.1016/j.injury.2022.09.041. [DOI] [PubMed] [Google Scholar]
  • 93.Prockop D.J., et al. Defining the risks of mesenchymal stromal cell therapy. Cytotherapy. 2010;12(5):576–578. doi: 10.3109/14653249.2010.507330. [DOI] [PubMed] [Google Scholar]
  • 94.Ankrum J.A., Ong J.F., Karp J.M. Mesenchymal stem cells: immune evasive, not immune privileged. Nat. Biotechnol. 2014;32(3):252–260. doi: 10.1038/nbt.2816. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Nickerson D.S., Yamasaki D.S. Objective evidence that nerve decompression surgery reduces neuropathic DFU recurrence risk to less than 5. Adv. Wound Care. 2024;13(7):363–374. doi: 10.1089/wound.2023.0199. [DOI] [PubMed] [Google Scholar]
  • 96.L P.K., et al. The mesenchymal stem cell secretome: a new paradigm towards cell-free therapeutic mode in regenerative medicine. Cytokine Growth Factor Rev. 2019;46:1–9. doi: 10.1016/j.cytogfr.2019.04.002. [DOI] [PubMed] [Google Scholar]
  • 97.Galland S., Stamenkovic I. Mesenchymal stromal cells in cancer: a review of their immunomodulatory functions and dual effects on tumor progression. J. Pathol. 2020;250(5):555–572. doi: 10.1002/path.5357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Xunian Z., Kalluri R. Biology and therapeutic potential of mesenchymal stem cell-derived exosomes. Cancer Sci. 2020;111(9):3100–3110. doi: 10.1111/cas.14563. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Guiotto M., et al. Human platelet lysate to substitute fetal bovine serum in hMSC expansion for translational applications: a systematic review. J. Transl. Med. 2020;18(1):351. doi: 10.1186/s12967-020-02489-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Guillamat-Prats R. The role of MSC in wound healing, scarring and regeneration. Cells. 2021;10(7) doi: 10.3390/cells10071729. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Huang Y., Wu Q., Tam P.K.H. Immunomodulatory mechanisms of mesenchymal stem cells and their potential clinical applications. Int. J. Mol. Sci. 2022;23(17) doi: 10.3390/ijms231710023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Lin Y.H., et al. The effects of microenvironment on wound healing by keratinocytes derived from mesenchymal stem cells. Ann. Plast. Surg. 2013;71(Suppl 1):S67–S74. doi: 10.1097/SAP.0000000000000045. [DOI] [PubMed] [Google Scholar]
  • 103.Dos Santos J.F., et al. Mesenchymal stem cells differentiate into keratinocytes and express epidermal kallikreins: towards an in vitro model of human epidermis. J. Cell. Biochem. 2019;120(8):13141–13155. doi: 10.1002/jcb.28589. [DOI] [PubMed] [Google Scholar]
  • 104.Khaki M., et al. Mesenchymal stem cells differentiate to endothelial cells using recombinant vascular endothelial growth factor -A. Rep Biochem Mol Biol. 2018;6(2):144–150. [PMC free article] [PubMed] [Google Scholar]
  • 105.Ikhapoh I.A., Pelham C.J., Agrawal D.K. Synergistic effect of angiotensin II on vascular endothelial growth factor-A-mediated differentiation of bone marrow-derived mesenchymal stem cells into endothelial cells. Stem Cell Res. Ther. 2015;6(1):4. doi: 10.1186/scrt538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Wang C., et al. Efficient differentiation of bone marrow mesenchymal stem cells into endothelial cells in vitro. Eur. J. Vasc. Endovasc. Surg. 2018;55(2):257–265. doi: 10.1016/j.ejvs.2017.10.012. [DOI] [PubMed] [Google Scholar]
  • 107.Zhang Y., Yu H., Li J. microRNA-181a-5p promotes fibroblast differentiation of mesenchymal stem cells in rats with pelvic floor dysfunction. Clinics. 2024;79 doi: 10.1016/j.clinsp.2024.100428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Mishra P.J., Banerjee D. Visualizing activated myofibroblasts resulting from differentiation of mesenchymal stem cells. Methods Mol. Biol. 2023;2593:83–92. doi: 10.1007/978-1-0716-2811-9_5. [DOI] [PubMed] [Google Scholar]
  • 109.Mishra P.J., Mishra P.J., Banerjee D. Keratinocyte induced differentiation of mesenchymal stem cells into dermal myofibroblasts: a role in effective wound healing. Int J Transl Sci. 2016;2016(1):5–32. doi: 10.13052/ijts2246-8765.2016.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.de Castro L.L., et al. Current understanding of the immunosuppressive properties of mesenchymal stromal cells. J. Mol. Med. (Berl.) 2019;97(5):605–618. doi: 10.1007/s00109-019-01776-y. [DOI] [PubMed] [Google Scholar]
  • 111.van Megen K.M., et al. Activated mesenchymal stromal cells process and present antigens regulating adaptive immunity. Front. Immunol. 2019;10:694. doi: 10.3389/fimmu.2019.00694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Abbasi B., et al. Mesenchymal stem cells and natural killer cells interaction mechanisms and potential clinical applications. Stem Cell Res. Ther. 2022;13(1):97. doi: 10.1186/s13287-022-02777-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Hu C.D., et al. Differential immunomodulatory effects of human bone marrow-derived mesenchymal stromal cells on natural killer cells. Stem Cell. Dev. 2019;28(14):933–943. doi: 10.1089/scd.2019.0059. [DOI] [PubMed] [Google Scholar]
  • 114.Podestà M.A., Remuzzi G., Casiraghi F. Mesenchymal stromal cell therapy in solid organ transplantation. Front. Immunol. 2020;11 doi: 10.3389/fimmu.2020.618243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Di Matteo S., et al. Neuroblastoma tumor-associated mesenchymal stromal cells regulate the cytolytic functions of NK cells. Cancers. 2022;15(1) doi: 10.3390/cancers15010019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Rezaei Kahmini F., Shahgaldi S., Moazzeni S.M. Mesenchymal stem cells alter the frequency and cytokine profile of natural killer cells in abortion-prone mice. J. Cell. Physiol. 2020;235(10):7214–7223. doi: 10.1002/jcp.29620. [DOI] [PubMed] [Google Scholar]
  • 117.Smith E.J., et al. Tumour necrosis factor alpha, interleukin 1 beta and interferon gamma have detrimental effects on equine tenocytes that cannot be rescued by IL-1RA or mesenchymal stromal cell-derived factors. Cell Tissue Res. 2023;391(3):523–544. doi: 10.1007/s00441-022-03726-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Grech L., et al. Possible role of circulating bone marrow mesenchymal progenitors in modulating inflammation and promoting wound repair. Int. J. Mol. Sci. 2021;23(1) doi: 10.3390/ijms23010078. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Bian D., et al. The application of mesenchymal stromal cells (MSCs) and their derivative exosome in skin wound healing: a comprehensive review. Stem Cell Res. Ther. 2022;13(1):24. doi: 10.1186/s13287-021-02697-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Matsuzaka Y., Yashiro R. Therapeutic strategy of mesenchymal-stem-cell-derived extracellular vesicles as regenerative medicine. Int. J. Mol. Sci. 2022;23(12) doi: 10.3390/ijms23126480. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Han Y., et al. Mesenchymal stem cells for regenerative medicine. Cells. 2019;8(8) doi: 10.3390/cells8080886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Nooshabadi V.T., et al. The extracellular vesicles-derived from mesenchymal stromal cells: a new therapeutic option in regenerative medicine. J. Cell. Biochem. 2018;119(10):8048–8073. doi: 10.1002/jcb.26726. [DOI] [PubMed] [Google Scholar]
  • 123.Sid-Otmane C., Perrault L.P., Ly H.Q. Mesenchymal stem cell mediates cardiac repair through autocrine, paracrine and endocrine axes. J. Transl. Med. 2020;18(1):336. doi: 10.1186/s12967-020-02504-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Arya S.B., Collie S.P., Parent C.A. The ins-and-outs of exosome biogenesis, secretion, and internalization. Trends Cell Biol. 2024;34(2):90–108. doi: 10.1016/j.tcb.2023.06.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Wang S., et al. Targeted therapy for inflammatory diseases with mesenchymal stem cells and their derived exosomes: from basic to clinics. Int. J. Nanomed. 2022;17:1757–1781. doi: 10.2147/IJN.S355366. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Rani S., et al. Mesenchymal stem cell-derived extracellular vesicles: toward cell-free therapeutic applications. Mol. Ther. 2015;23(5):812–823. doi: 10.1038/mt.2015.44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Shen Z., et al. Effects of mesenchymal stem cell-derived exosomes on autoimmune diseases. Front. Immunol. 2021;12 doi: 10.3389/fimmu.2021.749192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Liu H., et al. Immunomodulatory effects of mesenchymal stem cells and mesenchymal stem cell-derived extracellular vesicles in rheumatoid arthritis. Front. Immunol. 2020;11:1912. doi: 10.3389/fimmu.2020.01912. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Doyle L.M., Wang M.Z. Overview of extracellular vesicles, their origin, composition, purpose, and methods for exosome isolation and analysis. Cells. 2019;8(7) doi: 10.3390/cells8070727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Zhang L., Yu D. Exosomes in cancer development, metastasis, and immunity. Biochim. Biophys. Acta Rev. Canc. 2019;1871(2):455–468. doi: 10.1016/j.bbcan.2019.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 131.Yang B., Chen Y., Shi J. Exosome biochemistry and advanced nanotechnology for next-generation theranostic platforms. Adv. Mater. 2019;31(2) doi: 10.1002/adma.201802896. [DOI] [PubMed] [Google Scholar]
  • 132.Wortzel I., et al. Exosome-mediated metastasis: communication from a distance. Dev. Cell. 2019;49(3):347–360. doi: 10.1016/j.devcel.2019.04.011. [DOI] [PubMed] [Google Scholar]
  • 133.Hessvik N.P., Llorente A. Current knowledge on exosome biogenesis and release. Cell. Mol. Life Sci. 2018;75(2):193–208. doi: 10.1007/s00018-017-2595-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Zhang Y., et al. Recent advances in exosome-mediated nucleic acid delivery for cancer therapy. J. Nanobiotechnol. 2022;20(1):279. doi: 10.1186/s12951-022-01472-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Abhange K., et al. Small extracellular vesicles in cancer. Bioact. Mater. 2021;6(11):3705–3743. doi: 10.1016/j.bioactmat.2021.03.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Hu S., Hu Y., Yan W. Extracellular vesicle-mediated interorgan communication in metabolic diseases. Trends Endocrinol Metab. 2023;34(9):571–582. doi: 10.1016/j.tem.2023.06.002. [DOI] [PubMed] [Google Scholar]
  • 137.Chen J., et al. Review on strategies and technologies for exosome isolation and purification. Front. Bioeng. Biotechnol. 2021;9 doi: 10.3389/fbioe.2021.811971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138.Yang X.X., et al. New insight into isolation, identification techniques and medical applications of exosomes. J. Contr. Release. 2019;308:119–129. doi: 10.1016/j.jconrel.2019.07.021. [DOI] [PubMed] [Google Scholar]
  • 139.Li M., et al. Exosomes from different cells: characteristics, modifications, and therapeutic applications. Eur. J. Med. Chem. 2020;207 doi: 10.1016/j.ejmech.2020.112784. [DOI] [PubMed] [Google Scholar]
  • 140.Chen G., et al. Mesenchymal stem cell-derived exosomal miR-143-3p suppresses myocardial ischemia-reperfusion injury by regulating autophagy. Life Sci. 2021;280 doi: 10.1016/j.lfs.2021.119742. [DOI] [PubMed] [Google Scholar]
  • 141.Lin Z., et al. Mesenchymal stem cell-derived exosomes in cancer therapy resistance: recent advances and therapeutic potential. Mol. Cancer. 2022;21(1):179. doi: 10.1186/s12943-022-01650-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Qiu H., et al. Prospective application of exosomes derived from adipose-derived stem cells in skin wound healing: a review. J. Cosmet. Dermatol. 2020;19(3):574–581. doi: 10.1111/jocd.13215. [DOI] [PubMed] [Google Scholar]
  • 143.Tutuianu R., et al. Human mesenchymal stromal cell-derived exosomes promote in vitro wound healing by modulating the biological properties of skin keratinocytes and fibroblasts and stimulating angiogenesis. Int. J. Mol. Sci. 2021;22(12) doi: 10.3390/ijms22126239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144.Hassanzadeh A., et al. Mesenchymal stem/stromal cell-derived exosomes in regenerative medicine and cancer; overview of development, challenges, and opportunities. Stem Cell Res. Ther. 2021;12(1):297. doi: 10.1186/s13287-021-02378-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Qiu G., et al. Mesenchymal stem cell-derived extracellular vesicles affect disease outcomes via transfer of microRNAs. Stem Cell Res. Ther. 2018;9(1):320. doi: 10.1186/s13287-018-1069-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146.Bunggulawa E.J., et al. Recent advancements in the use of exosomes as drug delivery systems. J. Nanobiotechnol. 2018;16(1):81. doi: 10.1186/s12951-018-0403-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Zhang B., et al. Mesenchymal stem cell-derived extracellular vesicles in tissue regeneration. Cell Transplant. 2020;29 doi: 10.1177/0963689720908500. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148.Ha D.H., et al. Mesenchymal stem/stromal cell-derived exosomes for immunomodulatory therapeutics and skin regeneration. Cells. 2020;9(5) doi: 10.3390/cells9051157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Watanabe Y., Tsuchiya A., Terai S. The development of mesenchymal stem cell therapy in the present, and the perspective of cell-free therapy in the future. Clin. Mol. Hepatol. 2021;27(1):70–80. doi: 10.3350/cmh.2020.0194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Zhou C., 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Ding J.Y., et al. Mesenchymal stem cell-derived extracellular vesicles in skin wound healing: roles, opportunities and challenges. Mil Med Res. 2023;10(1):36. doi: 10.1186/s40779-023-00472-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Zhang W., et al. Extracellular vesicles derived from mesenchymal stem cells promote wound healing and skin regeneration by modulating multiple cellular changes: a brief review. Genes. 2023;14(8) doi: 10.3390/genes14081516. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Chen L., et al. Paracrine factors of mesenchymal stem cells recruit macrophages and endothelial lineage cells and enhance wound healing. PLoS One. 2008;3(4):e1886. doi: 10.1371/journal.pone.0001886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Kato J., et al. Mesenchymal stem cells ameliorate impaired wound healing through enhancing keratinocyte functions in diabetic foot ulcerations on the plantar skin of rats. J. Diabet. Complicat. 2014;28(5):588–595. doi: 10.1016/j.jdiacomp.2014.05.003. [DOI] [PubMed] [Google Scholar]
  • 155.Tefft J.B., Chen C.S., Eyckmans J. Reconstituting the dynamics of endothelial cells and fibroblasts in wound closure. APL Bioeng. 2021;5(1) doi: 10.1063/5.0028651. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Omorphos N.P., et al. Understanding angiogenesis and the role of angiogenic growth factors in the vascularisation of engineered tissues. Mol. Biol. Rep. 2021;48(1):941–950. doi: 10.1007/s11033-020-06108-9. [DOI] [PubMed] [Google Scholar]
  • 157.Zhang S., et al. Umbilical cord-matrix stem cells induce the functional restoration of vascular endothelial cells and enhance skin wound healing in diabetic mice via the polarized macrophages. Stem Cell Res. Ther. 2020;11(1):39. doi: 10.1186/s13287-020-1561-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Zhang B., et al. Human umbilical cord mesenchymal stem cell exosomes enhance angiogenesis through the Wnt4/β-catenin pathway. Stem Cells Transl Med. 2015;4(5):513–522. doi: 10.5966/sctm.2014-0267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Hu Y., 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Zhang L., et al. Exosomes from bone marrow mesenchymal stem cells enhance fracture healing through the promotion of osteogenesis and angiogenesis in a rat model of nonunion. Stem Cell Res. Ther. 2020;11(1):38. doi: 10.1186/s13287-020-1562-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Zhang Y., et al. Exosomes derived from adipose mesenchymal stem cells promote diabetic chronic wound healing through SIRT3/SOD2. Cells. 2022;11(16) doi: 10.3390/cells11162568. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Li X., 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):1–14. doi: 10.1038/s12276-018-0058-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Yan C., 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 doi: 10.3389/fbioe.2022.829868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Wawrzyniak O., et al. Protein-related circular RNAs in human pathologies. Cells. 2020;9(8) doi: 10.3390/cells9081841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Liang Z.H., et al. UCMSCs-derived exosomal circHIPK3 promotes ulcer wound angiogenesis of diabetes mellitus via miR-20b-5p/Nrf2/VEGFA axis. Diabet. Med. 2023;40(2) doi: 10.1111/dme.14968. [DOI] [PubMed] [Google Scholar]
  • 166.Zhang J., et al. Exosomes released from human induced pluripotent stem cells-derived MSCs facilitate cutaneous wound healing by promoting collagen synthesis and angiogenesis. J. Transl. Med. 2015;13:49. doi: 10.1186/s12967-015-0417-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Ren S., et al. Exosomes from adipose stem cells promote diabetic wound healing through the eHSP90/LRP1/AKT Axis. Cells. 2022;11(20) doi: 10.3390/cells11203229. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Yadav H., et al. Protection from obesity and diabetes by blockade of TGF-β/Smad3 signaling. Cell Metabol. 2011;14(1):67–79. doi: 10.1016/j.cmet.2011.04.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Hsu H.H., et al. Therapeutic potential of exosomes derived from diabetic adipose stem cells in cutaneous wound healing of db/db mice. Pharmaceutics. 2022;14(6) doi: 10.3390/pharmaceutics14061206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Wang J., 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 171.de Mayo T., et al. The role of bone marrow mesenchymal stromal cell derivatives in skin wound healing in diabetic mice. PLoS One. 2017;12(6) doi: 10.1371/journal.pone.0177533. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Hu L., et al. Exosomes derived from human adipose mensenchymal stem cells accelerates cutaneous wound healing via optimizing the characteristics of fibroblasts. Sci. Rep. 2016;6 doi: 10.1038/srep32993. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Pu C.M., et al. Adipose-derived stem cells protect skin flaps against ischemia/reperfusion injury via IL-6 expression. J. Invest. Dermatol. 2017;137(6):1353–1362. doi: 10.1016/j.jid.2016.12.030. [DOI] [PubMed] [Google Scholar]
  • 174.Zhang J., et al. Exosome and exosomal microRNA: trafficking, sorting, and function. Dev. Reprod. Biol. 2015;13(1):17–24. doi: 10.1016/j.gpb.2015.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Shen C., 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Tao H., et al. Mesenchymal stem cell-derived extracellular vesicles for corneal wound repair. Stem Cell. Int. 2019;2019 doi: 10.1155/2019/5738510. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Gu J., et al. Nanog interact with CDK6 to regulates astrocyte cells proliferation following spinal cord injury. Biochem. Biophys. Res. Commun. 2016;469(4):1097–1103. doi: 10.1016/j.bbrc.2015.12.053. [DOI] [PubMed] [Google Scholar]
  • 178.Nagarajan A., et al. PEA15 regulates the DNA damage-induced cell cycle checkpoint and oncogene-directed transformation. Mol. Cell Biol. 2014;34(12):2264–2282. doi: 10.1128/MCB.01542-13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Liao X., et al. Adipose mesenchymal stem cell sheets-derived extracellular vesicles-microRNA-10b promote skin wound healing by elevating expression of CDK6. Biomater. Adv. 2022;136 doi: 10.1016/j.bioadv.2022.212781. [DOI] [PubMed] [Google Scholar]
  • 180.Ma T., et al. Adipose mesenchymal stem cell-derived exosomes promote cell proliferation, migration, and inhibit cell apoptosis via Wnt/β-catenin signaling in cutaneous wound healing. J. Cell. Biochem. 2019;120(6):10847–10854. doi: 10.1002/jcb.28376. [DOI] [PubMed] [Google Scholar]
  • 181.Süntar I., et al. Regulatory role of Nrf2 signaling pathway in wound healing process. Molecules. 2021;26(9) doi: 10.3390/molecules26092424. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 182.Hseu Y.C., et al. Chalcone flavokawain A attenuates TGF-β1-induced fibrotic pathology via inhibition of ROS/Smad3 signaling pathways and induction of Nrf2/ARE-mediated antioxidant genes in vascular smooth muscle cells. J. Cell Mol. Med. 2019;23(2):775–788. doi: 10.1111/jcmm.13973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Mansuroğlu B., et al. Protective effect of chemically modified SOD on lipid peroxidation and antioxidant status in diabetic rats. Int. J. Biol. Macromol. 2015;72:79–87. doi: 10.1016/j.ijbiomac.2014.07.039. [DOI] [PubMed] [Google Scholar]
  • 184.He X., et al. MSC-derived exosome promotes M2 polarization and enhances cutaneous wound healing. Stem Cell. Int. 2019;2019 doi: 10.1155/2019/7132708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Locati M., Curtale G., Mantovani A. Diversity, mechanisms, and significance of macrophage plasticity. Annu. Rev. Pathol. 2020;15:123–147. doi: 10.1146/annurev-pathmechdis-012418-012718. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Wang C., et al. Macrophage polarization and its role in liver disease. Front. Immunol. 2021;12 doi: 10.3389/fimmu.2021.803037. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Abdelaziz M.H., et al. Alternatively activated macrophages; a double-edged sword in allergic asthma. J. Transl. Med. 2020;18(1):58. doi: 10.1186/s12967-020-02251-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Huang S.M., et al. High glucose environment induces M1 macrophage polarization that impairs keratinocyte migration via TNF-α: an important mechanism to delay the diabetic wound healing. J. Dermatol. Sci. 2019;96(3):159–167. doi: 10.1016/j.jdermsci.2019.11.004. [DOI] [PubMed] [Google Scholar]
  • 189.Yin Y., et al. Human umbilical cord-derived mesenchymal stem cells direct macrophage polarization to alleviate pancreatic islets dysfunction in type 2 diabetic mice. Cell Death Dis. 2018;9(7):760. doi: 10.1038/s41419-018-0801-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 190.Philipp D., et al. Preconditioning of bone marrow-derived mesenchymal stem cells highly strengthens their potential to promote IL-6-dependent M2b polarization. Stem Cell Res. Ther. 2018;9(1):286. doi: 10.1186/s13287-018-1039-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 191.Qin X., et al. 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 doi: 10.3389/fimmu.2023.1256687. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Landén N.X., Li D., Ståhle M. Transition from inflammation to proliferation: a critical step during wound healing. Cell. Mol. Life Sci. 2016;73(20):3861–3885. doi: 10.1007/s00018-016-2268-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 193.Zhu D., et al. Macrophage M2 polarization induced by exosomes from adipose-derived stem cells contributes to the exosomal proangiogenic effect on mouse ischemic hindlimb. Stem Cell Res. Ther. 2020;11(1):162. doi: 10.1186/s13287-020-01669-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 194.Zhang S., et al. MSC exosomes mediate cartilage repair by enhancing proliferation, attenuating apoptosis and modulating immune reactivity. Biomaterials. 2018;156:16–27. doi: 10.1016/j.biomaterials.2017.11.028. [DOI] [PubMed] [Google Scholar]
  • 195.Chamberlain C.S., et al. Extracellular vesicle-educated macrophages promote early achilles tendon healing. Stem Cell. 2019;37(5):652–662. doi: 10.1002/stem.2988. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Nakamura H., Takada K. Reactive oxygen species in cancer: current findings and future directions. Cancer Sci. 2021;112(10):3945–3952. doi: 10.1111/cas.15068. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Luo Q., et al. Antioxidant activity of mesenchymal stem cell-derived extracellular vesicles restores hippocampal neurons following seizure damage. Theranostics. 2021;11(12):5986–6005. doi: 10.7150/thno.58632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Al-Massri K.F., Ahmed L.A., El-Abhar H.S. Mesenchymal stem cells therapy enhances the efficacy of pregabalin and prevents its motor impairment in paclitaxel-induced neuropathy in rats: role of Notch1 receptor and JAK/STAT signaling pathway. Behav. Brain Res. 2019;360:303–311. doi: 10.1016/j.bbr.2018.12.013. [DOI] [PubMed] [Google Scholar]
  • 199.Ortiz-Virumbrales M., et al. Human adipose mesenchymal stem cells modulate myeloid cells toward an anti-inflammatory and reparative phenotype: role of IL-6 and PGE2. Stem Cell Res. Ther. 2020;11(1):462. doi: 10.1186/s13287-020-01975-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Zhou Y., et al. Bone marrow mesenchymal stem cell-derived exosomes ameliorate aging-induced BTB impairment in porcine testes by activating autophagy and inhibiting ROS/NLRP3 inflammasomes via the AMPK/mTOR signaling pathway. Antioxidants. 2024;13(2) doi: 10.3390/antiox13020183. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 201.Zhu F., et al. Hypoxic mesenchymal stem cell-derived exosomes alleviate ulcerative colitis injury by limiting intestinal epithelial cells reactive oxygen species accumulation and DNA damage through HIF-1α. Int. Immunopharm. 2022;113(Pt A) doi: 10.1016/j.intimp.2022.109426. [DOI] [PubMed] [Google Scholar]
  • 202.Hu W.S., et al. Mesenchymal stem cell transplantation increases antioxidant protein expression and ameliorates GP91/ROS/inflammasome signals in diabetic cardiomyopathy. J Cardiovasc Dev Dis. 2022;9(11) doi: 10.3390/jcdd9110381. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 203.Lu H., et al. Human mesenchymal stem cells-derived exosome mimetic vesicles regulation of the MAPK pathway and ROS levels inhibits glucocorticoid-induced apoptosis in osteoblasts. Stem Cell. Int. 2023;2023 doi: 10.1155/2023/5537610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 204.Qin A., et al. hMSCs treatment attenuates murine herpesvirus-68 (MHV-68) pneumonia through altering innate immune response via ROS/NLRP3 signaling pathway. Mol Biomed. 2023;4(1):27. doi: 10.1186/s43556-023-00137-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Barros J.F., et al. Role of chemokine receptor CCR4 and regulatory T cells in wound healing of diabetic mice. J. Invest. Dermatol. 2019;139(5):1161–1170. doi: 10.1016/j.jid.2018.10.039. [DOI] [PubMed] [Google Scholar]
  • 206.Hazrati A., et al. Therapeutic and immunomodulatory potentials of mesenchymal stromal/stem cells and immune checkpoints related molecules. Biomark. Res. 2024;12(1):35. doi: 10.1186/s40364-024-00580-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Prakhar P., et al. The small intestine epithelium exempts Foxp3+ Tregs from their IL-2 requirement for homeostasis and effector function. JCI Insight. 2021;6(21) doi: 10.1172/jci.insight.149656. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Sivasami P., et al. Obesity-induced dysregulation of skin-resident PPARγ(+) Treg cells promotes IL-17A-mediated psoriatic inflammation. Immunity. 2023;56(8):1844–1861.e6. doi: 10.1016/j.immuni.2023.06.021. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 209.Dikiy S., Rudensky A.Y. Principles of regulatory T cell function. Immunity. 2023;56(2):240–255. doi: 10.1016/j.immuni.2023.01.004. [DOI] [PubMed] [Google Scholar]
  • 210.Dong C. Cytokine regulation and function in T cells. Annu. Rev. Immunol. 2021;39:51–76. doi: 10.1146/annurev-immunol-061020-053702. [DOI] [PubMed] [Google Scholar]
  • 211.Li A., et al. Mesenchymal stem cell therapy: hope for patients with systemic lupus erythematosus. Front. Immunol. 2021;12 doi: 10.3389/fimmu.2021.728190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 212.Poli V., Zanoni I. Neutrophil intrinsic and extrinsic regulation of NETosis in health and disease. Trends Microbiol. 2023;31(3):280–293. doi: 10.1016/j.tim.2022.10.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 213.Zhu S., et al. The emerging roles of neutrophil extracellular traps in wound healing. Cell Death Dis. 2021;12(11):984. doi: 10.1038/s41419-021-04294-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Rodríguez-Espinosa O., et al. Metabolic requirements for neutrophil extracellular traps formation. Immunology. 2015;145(2):213–224. doi: 10.1111/imm.12437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Wong S.L., et al. Diabetes primes neutrophils to undergo NETosis, which impairs wound healing. Nat. Med. 2015;21(7):815–819. doi: 10.1038/nm.3887. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 216.Chow L., et al. Antibacterial activity of human mesenchymal stem cells mediated directly by constitutively secreted factors and indirectly by activation of innate immune effector cells. Stem Cells Transl Med. 2020;9(2):235–249. doi: 10.1002/sctm.19-0092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Estúa-Acosta G.A., et al. Human amniotic membrane mesenchymal stem cell-synthesized PGE(2) exerts an immunomodulatory effect on neutrophil extracellular trap in a PAD-4-dependent pathway through EP2 and EP4. Cells. 2022;11(18) doi: 10.3390/cells11182831. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Morishima Y., et al. Intravenous administration of mesenchymal stem cell-derived exosome alleviates spinal cord injury by regulating neutrophil extracellular trap formation through exosomal miR-125a-3p. Int. J. Mol. Sci. 2024;25(4) doi: 10.3390/ijms25042406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 219.Loh J.T., et al. Mechanism for the attenuation of neutrophil and complement hyperactivity by MSC exosomes. Cytotherapy. 2022;24(7):711–719. doi: 10.1016/j.jcyt.2021.12.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 220.Liu J., et al. Iron metabolism and type 2 diabetes mellitus: a meta-analysis and systematic review. J Diabetes Investig. 2020;11(4):946–955. doi: 10.1111/jdi.13216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 221.Shou Y., et al. Inhibition of keratinocyte ferroptosis suppresses psoriatic inflammation. Cell Death Dis. 2021;12(11):1009. doi: 10.1038/s41419-021-04284-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Zhou Y., et al. Iron overloaded polarizes macrophage to proinflammation phenotype through ROS/acetyl-p53 pathway. Cancer Med. 2018;7(8):4012–4022. doi: 10.1002/cam4.1670. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 223.Ge Y., Huang M., Yao Y.M. The effect and regulatory mechanism of high mobility group box-1 protein on immune cells in inflammatory diseases. Cells. 2021;10(5) doi: 10.3390/cells10051044. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 224.Feng Z., et al. Inhibition of ferroptosis rescues M2 macrophages and alleviates arthritis by suppressing the HMGB1/TLR4/STAT3 axis in M1 macrophages. Redox Biol. 2024;75 doi: 10.1016/j.redox.2024.103255. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 225.Vinchi F., et al. Atherosclerosis is aggravated by iron overload and ameliorated by dietary and pharmacological iron restriction. Eur. Heart J. 2020;41(28):2681–2695. doi: 10.1093/eurheartj/ehz112. [DOI] [PubMed] [Google Scholar]
  • 226.Bai T., et al. Inhibition of ferroptosis alleviates atherosclerosis through attenuating lipid peroxidation and endothelial dysfunction in mouse aortic endothelial cell. Free Radic. Biol. Med. 2020;160:92–102. doi: 10.1016/j.freeradbiomed.2020.07.026. [DOI] [PubMed] [Google Scholar]
  • 227.Luo E.F., et al. Role of ferroptosis in the process of diabetes-induced endothelial dysfunction. World J. Diabetes. 2021;12(2):124–137. doi: 10.4239/wjd.v12.i2.124. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Li S., et al. Diabetic ferroptosis plays an important role in triggering on inflammation in diabetic wound. Am. J. Physiol. Endocrinol. Metab. 2021;321(4):E509–e520. doi: 10.1152/ajpendo.00042.2021. [DOI] [PubMed] [Google Scholar]
  • 229.Shen H., et al. Application of deferoxamine in tissue regeneration attributed to promoted angiogenesis. Molecules. 2024;29(9) doi: 10.3390/molecules29092050. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 230.Duscher D., et al. Optimization of transdermal deferoxamine leads to enhanced efficacy in healing skin wounds. J. Contr. Release. 2019;308:232–239. doi: 10.1016/j.jconrel.2019.07.009. [DOI] [PubMed] [Google Scholar]
  • 231.Nie W., et al. Exosomal miR-17-92 derived from human mesenchymal stem cells promotes wound healing by enhancing angiogenesis and inhibiting endothelial cell ferroptosis. Tissue Cell. 2023;83 doi: 10.1016/j.tice.2023.102124. [DOI] [PubMed] [Google Scholar]
  • 232.Lu W., et al. Mesenchymal stem cell-derived extracellular vesicles accelerate diabetic wound healing by inhibiting NET-induced ferroptosis of endothelial cells. Int. J. Biol. Sci. 2024;20(9):3515–3529. doi: 10.7150/ijbs.97150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Fan J., et al. Mesenchymal stem cell-derived exosomal microRNA-367-3p alleviates experimental autoimmune encephalomyelitis via inhibition of microglial ferroptosis by targeting EZH2. Biomed. Pharmacother. 2023;162 doi: 10.1016/j.biopha.2023.114593. [DOI] [PubMed] [Google Scholar]
  • 234.Wei Z., et al. Human umbilical cord mesenchymal stem cells derived exosome shuttling mir-129-5p attenuates inflammatory bowel disease by inhibiting ferroptosis. J. Nanobiotechnol. 2023;21(1):188. doi: 10.1186/s12951-023-01951-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 235.Liu J., et al. Hypoxia pretreatment of bone marrow mesenchymal stem cells facilitates angiogenesis by improving the function of endothelial cells in diabetic rats with lower ischemia. PLoS One. 2015;10(5) doi: 10.1371/journal.pone.0126715. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 236.Xu T., et al. Vascular endothelial growth factor over-expressed mesenchymal stem cells-conditioned media ameliorate palmitate-induced diabetic endothelial dysfunction through PI-3K/AKT/m-TOR/eNOS and p38/MAPK signaling pathway. Biomed. Pharmacother. 2018;106:491–498. doi: 10.1016/j.biopha.2018.06.129. [DOI] [PubMed] [Google Scholar]
  • 237.Hou C., et al. The effect of heme oxygenase-1 complexed with collagen on MSC performance in the treatment of diabetic ischemic ulcer. Biomaterials. 2013;34(1):112–120. doi: 10.1016/j.biomaterials.2012.09.022. [DOI] [PubMed] [Google Scholar]
  • 238.Chen J., et al. The key regulatory roles of the PI3K/Akt signaling pathway in the functionalities of mesenchymal stem cells and applications in tissue regeneration. Tissue Eng., Part B. 2013;19(6):516–528. doi: 10.1089/ten.TEB.2012.0672. [DOI] [PubMed] [Google Scholar]
  • 239.Logan C.Y., Nusse R. The Wnt signaling pathway in development and disease. Annu. Rev. Cell Dev. Biol. 2004;20:781–810. doi: 10.1146/annurev.cellbio.20.010403.113126. [DOI] [PubMed] [Google Scholar]
  • 240.Jun E.K., et al. Hypoxic conditioned medium from human amniotic fluid-derived mesenchymal stem cells accelerates skin wound healing through TGF-β/SMAD2 and PI3K/Akt pathways. Int. J. Mol. Sci. 2014;15(1):605–628. doi: 10.3390/ijms15010605. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 241.Ebrahim N., et al. Adipose mesenchymal stem cells combined with platelet-rich plasma accelerate diabetic wound healing by modulating the Notch pathway. Stem Cell Res. Ther. 2021;12(1):392. doi: 10.1186/s13287-021-02454-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 242.Li M., et al. Mesenchymal stem cell-conditioned medium improves the proliferation and migration of keratinocytes in a diabetes-like microenvironment. Int. J. Low. Extrem. Wounds. 2015;14(1):73–86. doi: 10.1177/1534734615569053. [DOI] [PubMed] [Google Scholar]
  • 243.Huo J., et al. Bone marrow-derived mesenchymal stem cells promoted cutaneous wound healing by regulating keratinocyte migration via β(2)-adrenergic receptor signaling. Mol. Pharm. 2018;15(7):2513–2527. doi: 10.1021/acs.molpharmaceut.7b01138. [DOI] [PubMed] [Google Scholar]
  • 244.Shabbir A., et al. Mesenchymal stem cell exosomes induce proliferation and migration of normal and chronic wound fibroblasts, and enhance angiogenesis in vitro. Stem Cell. Dev. 2015;24(14):1635–1647. doi: 10.1089/scd.2014.0316. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 245.Ma J., et al. Hepatocyte growth factor secreted from human adipose-derived stem cells inhibits fibrosis in hypertrophic scar fibroblasts. Curr. Mol. Med. 2020;20(7):558–571. doi: 10.2174/1566524020666200106095745. [DOI] [PubMed] [Google Scholar]
  • 246.Wang W., et al. MicroRNA-129 and -335 promote diabetic wound healing by inhibiting sp1-mediated MMP-9 expression. Diabetes. 2018;67(8):1627–1638. doi: 10.2337/db17-1238. [DOI] [PubMed] [Google Scholar]
  • 247.Zhou C., et al. Immunomodulatory effect of urine-derived stem cells on inflammatory bowel diseases via downregulating Th1/Th17 immune responses in a PGE2-dependent manner. J Crohns Colitis. 2020;14(5):654–668. doi: 10.1093/ecco-jcc/jjz200. [DOI] [PubMed] [Google Scholar]
  • 248.El-Sayed M., et al. Immunomodulatory effect of mesenchymal stem cells: cell origin and cell quality variations. Mol. Biol. Rep. 2019;46(1):1157–1165. doi: 10.1007/s11033-018-04582-w. [DOI] [PubMed] [Google Scholar]
  • 249.Silva-Carvalho A., et al. Dissecting the relationship between antimicrobial peptides and mesenchymal stem cells. Pharmacol. Ther. 2022;233 doi: 10.1016/j.pharmthera.2021.108021. [DOI] [PubMed] [Google Scholar]
  • 250.Radic M., Muller S. LL-37, a multi-faceted amphipathic peptide involved in NETosis. Cells. 2022;11(15) doi: 10.3390/cells11152463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 251.Memariani H., Memariani M. Antibiofilm properties of cathelicidin LL-37: an in-depth review. World J. Microbiol. Biotechnol. 2023;39(4):99. doi: 10.1007/s11274-023-03545-z. [DOI] [PubMed] [Google Scholar]
  • 252.Routsias J.G., et al. Serum β-defensin 2, A novel biomarker for the diagnosis of acute infections. Diagnostics. 2023;13(11) doi: 10.3390/diagnostics13111885. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 253.Damour A., et al. Eczema herpeticum: clinical and pathophysiological aspects. Clin. Rev. Allergy Immunol. 2020;59(1):1–18. doi: 10.1007/s12016-019-08768-3. [DOI] [PubMed] [Google Scholar]
  • 254.Liu J., et al. Apoptotic bodies derived from mesenchymal stem cells promote cutaneous wound healing via regulating the functions of macrophages. Stem Cell Res. Ther. 2020;11(1):507. doi: 10.1186/s13287-020-02014-w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 255.Nazarenko I. Extracellular vesicles: recent developments in Technology and perspectives for cancer liquid biopsy. Recent Results Cancer Res. 2020;215:319–344. doi: 10.1007/978-3-030-26439-0_17. [DOI] [PubMed] [Google Scholar]
  • 256.Tsuji K., Kitamura S., Wada J. Immunomodulatory and regenerative effects of mesenchymal stem cell-derived extracellular vesicles in renal diseases. Int. J. Mol. Sci. 2020;21(3) doi: 10.3390/ijms21030756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 257.Liu X., et al. Umbilical cord mesenchymal stem cell-derived small extracellular vesicles deliver miR-21 to promote corneal epithelial wound healing through PTEN/PI3K/Akt pathway. Stem Cell. Int. 2022;2022 doi: 10.1155/2022/1252557. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 258.Madhyastha R., et al. NFkappaB activation is essential for miR-21 induction by TGFβ1 in high glucose conditions. Biochem. Biophys. Res. Commun. 2014;451(4):615–621. doi: 10.1016/j.bbrc.2014.08.035. [DOI] [PubMed] [Google Scholar]
  • 259.Huang C., et al. Human mesenchymal stem cells promote ischemic repairment and angiogenesis of diabetic foot through exosome miRNA-21-5p. Stem Cell Res. 2021;52 doi: 10.1016/j.scr.2021.102235. [DOI] [PubMed] [Google Scholar]
  • 260.Zhou Q., et al. Regulation of angiogenesis and choroidal neovascularization by members of microRNA-23∼27∼24 clusters. Proc. Natl. Acad. Sci. U. S. A. 2011;108(20):8287–8292. doi: 10.1073/pnas.1105254108. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 261.Fang S., et al. Umbilical cord-derived mesenchymal stem cell-derived exosomal MicroRNAs suppress myofibroblast differentiation by inhibiting the transforming growth factor-β/SMAD2 pathway during wound healing. Stem Cells Transl Med. 2016;5(10):1425–1439. doi: 10.5966/sctm.2015-0367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 262.Shen G., et al. Role of ADAM9 and miR-126 in the development of abdominal aortic aneurysm. Atherosclerosis. 2020;297:47–54. doi: 10.1016/j.atherosclerosis.2020.01.014. [DOI] [PubMed] [Google Scholar]
  • 263.Sun Y., et al. Platelet-derived exosomes affect the proliferation and migration of human umbilical vein endothelial cells via miR-126. Curr. Vasc. Pharmacol. 2019;17(4):379–387. doi: 10.2174/1570161116666180313142139. [DOI] [PubMed] [Google Scholar]
  • 264.Tao S.C., et al. Chitosan wound dressings incorporating exosomes derived from MicroRNA-126-overexpressing synovium mesenchymal stem cells provide sustained release of exosomes and heal full-thickness skin defects in a diabetic rat model. Stem Cells Transl Med. 2017;6(3):736–747. doi: 10.5966/sctm.2016-0275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 265.Xu J., et al. The role of microRNA-146a in the pathogenesis of the diabetic wound-healing impairment: correction with mesenchymal stem cell treatment. Diabetes. 2012;61(11):2906–2912. doi: 10.2337/db12-0145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 266.Gangadaran P., et al. Extracellular vesicles from mesenchymal stem cells activates VEGF receptors and accelerates recovery of hindlimb ischemia. J. Contr. Release. 2017;264:112–126. doi: 10.1016/j.jconrel.2017.08.022. [DOI] [PubMed] [Google Scholar]
  • 267.Yu M., 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 268.Zhang N., et al. Elevated hepatic expression of H19 long noncoding RNA contributes to diabetic hyperglycemia. JCI Insight. 2018;3(10) doi: 10.1172/jci.insight.120304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 269.Li B., et al. The MSC-derived exosomal lncRNA H19 promotes wound healing in diabetic foot ulcers by upregulating PTEN via MicroRNA-152-3p. Mol. Ther. Nucleic Acids. 2020;19:814–826. doi: 10.1016/j.omtn.2019.11.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Yang H., et al. Hair follicle mesenchymal stem cell exosomal lncRNA H19 inhibited NLRP3 pyroptosis to promote diabetic mouse skin wound healing. Aging (Albany NY) 2023;15(3):791–809. doi: 10.18632/aging.204513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 271.Zhu L., et al. Improved therapeutic effects on diabetic foot by human mesenchymal stem cells expressing MALAT1 as a sponge for microRNA-205-5p. Aging (Albany NY) 2019;11(24):12236–12245. doi: 10.18632/aging.102562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 272.Shi R., et al. Exosomes derived from mmu_circ_0000250-modified adipose-derived mesenchymal stem cells promote wound healing in diabetic mice by inducing miR-128-3p/SIRT1-mediated autophagy. Am. J. Physiol. Cell Physiol. 2020;318(5):C848–c856. doi: 10.1152/ajpcell.00041.2020. [DOI] [PubMed] [Google Scholar]
  • 273.Hu C., Li L. Preconditioning influences mesenchymal stem cell properties in vitro and in vivo. J. Cell Mol. Med. 2018;22(3):1428–1442. doi: 10.1111/jcmm.13492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 274.Chu Z., et al. Novel neutrophil extracellular trap-related mechanisms in diabetic wounds inspire a promising treatment strategy with hypoxia-challenged small extracellular vesicles. Bioact. Mater. 2023;27:257–270. doi: 10.1016/j.bioactmat.2023.04.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 275.Ariyanti A.D., et al. Salidroside-pretreated mesenchymal stem cells enhance diabetic wound healing by promoting paracrine function and survival of mesenchymal stem cells under hyperglycemia. Stem Cells Transl Med. 2019;8(4):404–414. doi: 10.1002/sctm.18-0143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 276.Hu Y., et al. Exosomes derived from pioglitazone-pretreated MSCs accelerate diabetic wound healing through enhancing angiogenesis. J. Nanobiotechnol. 2021;19(1):150. doi: 10.1186/s12951-021-00894-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Liu W., 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Ferreira J.R., et al. Mesenchymal stromal cell secretome: influencing therapeutic potential by cellular pre-conditioning. Front. Immunol. 2018;9:2837. doi: 10.3389/fimmu.2018.02837. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 279.Liang B., et al. Dimethyloxaloylglycine-stimulated human bone marrow mesenchymal stem cell-derived exosomes enhance bone regeneration through angiogenesis by targeting the AKT/mTOR pathway. Stem Cell Res. Ther. 2019;10(1):335. doi: 10.1186/s13287-019-1410-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 280.Abdelrahman S.A., Samak M.A., Shalaby S.M. Fluoxetine pretreatment enhances neurogenic, angiogenic and immunomodulatory effects of MSCs on experimentally induced diabetic neuropathy. Cell Tissue Res. 2018;374(1):83–97. doi: 10.1007/s00441-018-2838-6. [DOI] [PubMed] [Google Scholar]

Articles from Heliyon are provided here courtesy of Elsevier

RESOURCES