Open Access

Cell migration in diabetic wound healing: Molecular mechanisms and therapeutic strategies (Review)

  • Authors:
    • Jielin Song
    • Tong Zhao
    • Chuanfu Wang
    • Xu Sun
    • Junchao Sun
    • Zhaohui Zhang
  • View Affiliations

  • Published online on: June 16, 2025     https://doi.org/10.3892/ijmm.2025.5567
  • Article Number: 126
  • Copyright: © Song et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Diabetic wounds are among the most prevalent forms of chronic wound and are a prominent clinical challenge in contemporary healthcare. Impaired cell migration represents one of the key mechanisms underlying the difficulty in diabetic wound healing, involving multiple cell types including neutrophils, macrophages, keratinocytes, endothelial cells and fibroblasts. Under the influence of pathological factors, including hyperglycemia, chronic inflammation, oxidative stress and an abnormal microenvironment, the cell migration becomes impaired, leading to delayed wound healing. Key signaling pathways including Rho GTPase, PI3K/Akt, TGF‑β/Smad and Wnt/β‑catenin are involved in the regulation of cell migration. Non‑coding RNAs exert a pivotal influence on diabetic wound healing by modulating these signaling pathways or their downstream targets. Notably, stem cells and their exosomes, growth factor therapy, drug‑loaded dressings and traditional Chinese medicine can modulate cell migration via non‑coding RNAs and associated signaling pathways, thereby establishing a therapeutic regulatory axis. This review systematically consolidates advances in this field, providing novel insight into the mechanisms of cell migration in diabetic wounds and facilitating the development of innovative therapeutic strategies.

Introduction

Diabetic wounds are a prevalent and severe complication of diabetes mellitus, characterized by impaired healing, increased infection risk and failure to achieve complete closure (1,2). Among reported cases of diabetes mellitus, ~25% are at risk of developing diabetic wounds (3). Diabetes disrupts wound healing, potentially leading to chronic foot ulcers, lower extremity amputation and increased mortality (4,5). Moreover, the high treatment costs of diabetic wounds place an economic burden on healthcare systems (6). The global prevalence of diabetes was estimated at 529 million cases in 2021, with projections indicating a potential surge to 1.31 billion by 2050 (7). With the increasing prevalence of diabetes, the incidence of diabetic wounds is expected to rise, posing a challenge to public health systems.

The difficulty in diabetic wound healing primarily arises from a multifactorial combination of neuropathy, vasculopathy and infection, which collectively exacerbate the complexity and challenges of wound management (8). At the cellular level, a key factor influencing wound healing is the efficient and rapid migration of cells to the wound center (9). This involves multiple cell types, including neutrophils, macrophages, keratinocytes, fibroblasts and endothelial cells (ECs) (10-14). The diabetic microenvironment impairs the migratory capacity of cells via multiple mechanisms, thereby contributing to delayed wound healing (15,16). Consequently, understanding of cell migration regulatory mechanisms is key to formulate novel therapeutic interventions for diabetic wounds.

The present study aimed to summarize cell migration dysfunction in diabetic wounds and the mechanisms underlying cell migration impairment induced by hyperglycemia, chronic inflammation, oxidative stress and an abnormal microenvironment. Based on key signaling pathways, non-coding RNAs (ncRNAs) and their associated molecular networks that regulate cell migration in diabetic wounds, the present study further explores innovative therapeutic strategies to enhance cell migration, as well as therapeutic challenges and future research directions, offering innovative perspectives to advance diabetic wound clinical management.

Cell migration

Cell migration is a synchronized and dynamic phenomenon that generally commences with the polarization of cells (Fig. 1) (17). Cell polarization occurs when migrating cells detect chemotactic signals such as chemokines, growth factors or extracellular matrix (ECM) cues. This process involves coordinated activity of key molecular players, including Rho GTPases, integrins, phosphoinositide 3-kinase (PI3K), microtubules and vesicular transport systems, to establish front-rear polarity and generate distinct functional patterns between the leading and trailing edges (18). Actin polymerization at the leading edge drives membrane protrusion, forming pseudopodial structures such as lamellipodia and filopodia, facilitating cell extension. The extended pseudopodia adhere to the ECM through adhesion molecules such as integrins, forming focal adhesions that mechanically link the cytoskeleton to the extracellular environment (19). Through myosin-mediated contractile forces, the cell body is propelled forward, driving translocation. Disassembly of adhesion complexes at the trailing edge permits tail detachment from the substrate, finalizing the de-adhesion process (20). The coordinated actions of the anterior and posterior regions complete one cycle of migration.

Cell migration in normal wound healing

Cell migration is a critical and continuous process in wound healing, integral to every phase of repair. Various cell types facilitate tissue regeneration, notably the migration of neutrophils, macrophages, keratinocytes, vascular ECs and fibroblasts. Through precise migration and functional modulation, these cells contribute to pathogen clearance, angiogenesis promotion, ECM secretion and epidermal barrier reconstruction. Their migration is regulated by signaling molecules and microenvironmental factors to ensure efficient repair (21,22). Investigating these migration mechanisms not only enhances understanding of the physiological principles governing wound healing but also offers a solid theoretical framework for potential therapeutic strategies for managing chronic wounds and impaired repair.

Neutrophil migration

Neutrophils are the initial immune responders during wound healing, rapidly mobilizing to injury sites via intricate migratory mechanisms (Fig. 2A). Neutrophils detect 'find-me' signals, including damage-associated molecular patterns (DAMPs), hydrogen peroxide, lipid mediators and chemokines, via surface receptors such as G protein-coupled receptors (GPCRs), integrins, Fc receptors and pattern recognition receptors. These signals, released from injured tissues, drive neutrophil directed migration to the site of damage and trigger inflammatory responses (23,24). The migration process is coordinated by ~30 distinct neutrophil receptors and multiple signaling pathways. Neutrophils recognize fMet-Leu-Phe (fMLP) released by damaged cells and bacteria through specific formyl peptide receptors (25). Mast cells augment vascular permeability and facilitate neutrophil infiltration via the release of histamine, chemokines and inflammatory mediators. Macrophages identify DAMPs or pathogen-associated molecular patterns, leading to their activation and guidance of neutrophils to the site of injury (26). Local cells and neutrophils release chemokines, including C-C motif ligand 3 (CCL3) and C-X-C motif ligands 1/2 (CXCL1/2). These chemokines interact with their receptors (CCR1, CXCR1, and CXCR2) to guide neutrophil migration (21). Furthermore, neutrophils produce leukotriene B4, which binds leukotriene B4 receptor 1 to drive neutrophil recruitment from distant tissue (27). During migration, neutrophils utilize adhesion molecules (such as CD11b) and downstream signaling pathways (such as Src and Rho GTPases) to drive actin remodeling and membrane extension, enabling transendothelial migration and deep tissue infiltration through ECM degradation (23,28). At the wound site, neutrophils perform essential antimicrobial functions through various mechanisms, including the secretion of toxic granules, production of reactive oxygen species, phagocytosis of invading pathogens and the formation of neutrophil extracellular traps (29,30). Additionally, they secrete proteases to remodel the ECM, recruit immune cells and facilitate tissue repair. While these functions are essential for combating infections, they can also lead to bystander effects, causing tissue damage, particularly in chronic inflammatory conditions (31,32). Upon completion of their task, neutrophils are cleared by macrophages or re-enter the vasculature via reverse migration, thereby contributing to the resolution of inflammation (33). Excessive neutrophil retention or migration defects may sustain inflammatory responses and hinder healing processes, contributing to chronic wound pathogenesis (34). The CXCL12-CXCR4 axis serves a pivotal role in neutrophil retention at inflammatory sites. Inhibition of this pathway may enhance inflammation resolution by inducing neutrophil reverse migration (35).

Figure 2

Cell migration and function during normal wound healing. (A) Neutrophil migration in wound healing. Neutrophils are regulated by immune signals from macrophages and mast cells, as well as positive autocrine feedback, leading to migration to the wound site to initiate the inflammatory response. After fulfilling their role, neutrophils are phagocytosed by macrophages or re-enter circulation through reverse migration. (B) Monocyte/macrophage recruitment and polarization. Platelets, mast cells and endogenous chemokines recruit monocytes to the wound site, where they differentiate into M1 macrophages, mediating pro-inflammatory and phagocytic responses. Upon inflammation resolution, these macrophages shift to the M2 phenotype, promoting tissue repair and regeneration. (C) Keratinocyte migration and epithelialization. Inflammatory mediators released by macrophages, neutrophils and mast cells, along with growth factors secreted by fibroblasts, facilitate the migration of keratinocytes towards the wound site. MMPs facilitate ECM removal, thereby creating pathways that promote cell migration and re-epithelialization. (D) Endothelial cell migration-driven angiogenesis. Vascular endothelial cells, influenced by growth factors such as VEGF secreted by macrophages and fibroblasts, migrate both locally and systemically toward the wound site, thereby facilitating angiogenesis. (E) Fibroblast migration and tissue remodeling. Keratinocytes, macrophages and platelets secrete cytokines and growth factors that recruit fibroblasts to the wound site. These fibroblasts synthesize collagen to generate granulation tissue, differentiate into myofibroblasts to induce wound contraction and drive scar maturation through ECM remodeling. Created with BioRender.com. ECM, extracellular matrix; NET, neutrophil extracellular trap; CCL, C-C motif ligand; CXCL, C-X-C motif ligand; LTB, leukotriene B; CXCR, CXC chemokine receptor; BLT, leukotriene B4 receptor; CCR, CC chemokine receptor; HMGB, high-mobility group box; SDF, stromal cell-derived factor; HIF, hypoxia-inducible factor; KGF, keratinocyte growth factor; HGF, hepatocyte growth factor; SMA, smooth muscle actin.

Monocyte/macrophage migration

Following tissue injury, a coordinated chemokine cascade drives monocyte and macrophage trafficking to the wound site (Fig. 2B). Damaged cells release calcium waves that activate NADPH oxidase, producing hydrogen peroxide, which, together with calcium, serves as an early signal to mobilize immune cells to the injury site (36-38). Additionally, DAMPs such as high-mobility group box 1 and ATP, along with inflammatory cytokines such as IL-1 and IL-33, activate resident macrophages, prompting the release of pro-inflammatory factors that establish a localized inflammatory environment (39). During the initial injury phase, monocytes expressing CCR2 migrate in response to CCL2 signaling. These monocytes simultaneously express and respond to CCL7, promoting the recruitment of myeloid cells (monocytes and macrophages) to the injury site (21). Furthermore, the degranulation of platelets and mast cells releases chemokines such as stromal cell-derived factor 1/CXCL12, while hypoxia-inducible factors (HIFs) amplify chemotactic signals, promoting the recruitment of monocytes (23). The clotting mechanism initiated by blood vessel damage further liberates compounds such as unbound heme, exacerbating the inflammatory reaction and drawing monocytes to the site of injury (40). Upon reaching the wound site, monocytes develop into macrophages and serve essential roles throughout the healing process. Initially, M1-polarized macrophages dominate, displaying potent antimicrobial and inflammatory properties (41). As healing progresses, macrophages transition to the M2 phenotype, facilitating inflammation resolution and angiogenesis while orchestrating collagen deposition and ECM remodeling to promote tissue regeneration and functional recovery (42,43).

Keratinocyte migration

Keratinocyte movement is key for successful wound closure, facilitating re-epithelialization and barrier function recovery (Fig. 2C). Following tissue injury, edge-located keratinocytes respond to inflammatory mediators (IL-1, TNF-α) by adopting a flattened morphology, cellular elongation and forming membrane protrusions such as lamellipodia and filopodia (22). These changes are driven by cytoskeletal reorganization, which provides the structural support and mechanical force required for migration. Fibroblasts promote keratinocyte proliferation, migration and differentiation by secreting growth factors such as keratinocyte and hepatocyte growth factors, thereby driving the re-epithelialization of wounds (44). During motility, keratinocytes engage with the ECM via integrin receptors such as αvβ5 and α5β1 while releasing MMPs to remodel temporary matrix proteins (fibrin, fibronectin), thereby promoting cellular migration (45-48). Dynamic regulation of cell-cell and cell-ECM connections, along with increased gap junction communication, ensures coordinated and efficient migration (49). Keratinocytes employ multiple mechanisms for migration, including the 'leapfrog' and 'sliding' models and suprabasal cell dedifferentiation in collaboration with basal cells (22). The leapfrog mechanism proposes that suprabasal cells roll over the leading edge basal cells, undergo dedifferentiation, and subsequently form new migratory leaders within the cohesive epidermal tongue (50). In the sliding mechanism, keratinocytes from the basal layer move forward as a cohesive block at the leading edge, while the overlying cluster of superficial cells is passively dragged along (51). Suprabasal cell dedifferentiation refers to the reversal of differentiation in committed suprabasal keratinocytes, which regain migratory and proliferative capacity to directly contribute to epidermal regeneration. During migration, keratinocytes proliferate and differentiate to form new epithelium, reestablishing the skin barrier. They also regulate local inflammation, promote ECM remodeling and coordinate the activity of neighboring cells through autocrine and paracrine signaling (52). Additionally, keratinocytes adapt their migratory behavior and functional characteristics to the dynamic wound microenvironment, providing key flexibility and support for effective wound repair.

EC migration

New blood vessel formation (angiogenesis) peaks in the proliferative stage of repair, delivering oxygen and nutrients critical for tissue regeneration and functional restoration (53). EC movement is key for vascular restructuring and necessary for new blood vessel formation, primarily mediated by chemotactic, haptotactic and mechanotactic cues (Fig. 2D) (54). Chemotaxis in angiogenesis involves the directional migration of ECs along chemical gradients of attractants such as VEGF and basic fibroblast growth factor. This migration is initiated when VEGF binds to VEGF receptor (R)-2, activating downstream effectors such as PI3K/Akt and Rho GTPases to remodel the cytoskeleton. VEGFR-2 serves as the primary regulator of this process (55). Through paracrine signaling, VEGF is secreted by repair-associated macrophages, keratinocytes and fibroblasts, driving EC expansion and motility to enhance blood vessel formation (56,57). Haptotaxis refers to the directional movement of ECs during angiogenesis, driven by the interaction of integrins (such as αvβ3 and αvβ5) with ECM components along a ligand gradient (58). Integrin activation triggers downstream signaling pathways, including Ras-related C3 botulinum toxin substrate (Rac) and cell division cycle 42 (Cdc42), which regulate cytoskeletal remodeling and mechanical force generation to propel cell movement (59). Additionally, integrins synergize with growth factor signaling pathways to enhance migratory efficiency. Mechanotaxis is the process by which ECs undergo directed migration in response to mechanical forces, such as shear stress, mediated by integrin activation and cytoskeletal remodeling (60).

Fibroblast migration

As key mediators of tissue regeneration, fibroblasts generate and remodel ECM proteins, including collagen, elastin, fibronectin and laminin, to support structural integrity (61). Chemokines and cytokines secreted by platelets and inflammatory cells guide fibroblasts to the injury site during early wound healing (62,63). Keratinocytes support this process by releasing IL-1 and transforming growth factor (TGF)-β, which enhance fibroblast migration and activation (Fig. 2E) (44). Fibroblasts recruited to the wound site synthesize and secrete ECM components, such as collagen, promoting granulation tissue formation. Under mechanical tension and cytokine stimulation (TGF-β), these fibroblasts differentiate into myofibroblasts. This transition is marked by α-smooth muscle actin expression, which forms stress fibers that generate contractile forces to draw the wound edges together, aiding in closure (64). Additionally, a small population of bone marrow-derived circulating fibroblasts migrates to the wound bed in response to cytokines such as IL-4, IL-13 and IFN-γ and differentiates into myofibroblasts (65); however, their contribution to skin wound healing is relatively minor (66). Persistent fibroblast overproliferation and hyperactivation drive excessive ECM deposition, primarily collagen, promoting hypertrophic scar or keloid formation (67,68). In the final remodeling phase, fibroblasts deposit type I collagen to replace type III collagen, reinforcing the ECM and forming a mature, mechanically stable scar. This process may persist for months to years (69).

In wound healing, although different cell types exhibit distinct migration characteristics, their movement is regulated by signaling molecules such as chemokines and cytokines, as well as microenvironmental factors including hypoxia, mechanical forces and ECM composition (21,23,44,56). These cells sense external cues via surface receptors, facilitate migration through cytoskeletal reorganization and dynamic adhesion molecule interactions and maintain coordinated motility via cell-cell and cell-matrix communications (23,28). Collectively, these mechanisms establish an efficient and orderly repair network that drives the wound healing process.

Mechanisms of cellular migration impairment in diabetic wound healing

Diabetic patients exhibit impairment in cell migration function during wound repair (70,71). This impairment may be linked to the interplay of multiple factors, including hyperglycemia, chronic inflammatory responses, oxidative stress and an aberrant wound microenvironment. These factors influence cell migration via complex biological pathways, ultimately leading to the obstruction of wound healing and potentially resulting in prolonged refractory states (Fig. 3).

Effects of high glucose on cell migration

High glucose may impair cell migration by promoting the formation of unstable protrusions, decreasing adhesion maturation, altering RhoA activity to disrupt migration regulation and enhancing glucose uptake and metabolism to activate the mTOR pathway (72). These mechanisms are implicated in various cell types such as neutrophils, macrophages, keratinocytes and endothelial cells. Chronic elevation of blood glucose levels results in the buildup of advanced glycation end products (AGEs) within tissue. AGE-modified proteins disrupt chemotactic signaling, impairing the migration of neutrophils to wound sites (73). Under chronic hyperglycemia, monocyte motility is compromised, resulting in inadequate macrophage infiltration, decreased phagocytic capacity and dysregulated polarization from M1 to M2 phenotypes (74). Inadequate migration of neutrophils and macrophages to the wound site increases the risk of wound infection (75). Additionally, high glucose upregulates STING expression and activates the interferon regulatory factor 3 and NF-κB signaling pathway, thereby inhibiting EC migration and delaying the healing of diabetic wounds (70). Under physiological conditions, keratinocyte proliferation and migration are key for re-epithelialization during cutaneous wound repair. Phosphorylated focal adhesion kinase (p125FAK) is a key regulator of keratinocyte migration. However, hyperglycemia attenuates p125FAK phosphorylation, compromising keratinocyte migration in diabetic wound healing. (76). High glucose also inhibits the PI3K signaling pathway, impairing the function of ClC-2 chloride channels and suppressing the migratory capacity of keratinocytes. Conversely, hyperglycemia-induced upregulation of keratin 17 activates c-MYB/PI3K/AKT signaling, promoting excessive keratinocyte proliferation and migration. This dysregulation contributes to hyperkeratosis and impairs wound healing (77). Thus, high glucose conditions exert a dual effect, both inhibiting and promoting cell migration through distinct mechanisms, leading to delayed diabetic wound healing.

Effects of chronic inflammation on cell migration

Chronic inflammation is a key pathological mechanism underlying diabetic wound healing disorder, exerting its effects on cell migration and delaying the wound repair process through a variety of mechanisms (78-80). Hyperglycemia impedes macrophage polarization from the M1 to M2 phenotype, leading to persistent secretion of pro-inflammatory mediators including IL-6, IL-1 and TNF-α, alongside decreased production of anti-inflammatory factors such as IL-10 and TGF-β (57,80). This persistent pro-inflammatory microenvironment markedly impairs the motility of keratinocytes, fibroblasts and vascular ECs, extending the inflammatory phase and impairing wound repair (81-83). Overproduction of TNF-α from M1-polarized macrophages elevates tissue inhibitor of metalloproteinases-1 (TIMP-1) expression in keratinocytes, suppressing motility and ultimately impairing diabetic wound repair (71). IL-1β is a key contributor to maintaining a pro-inflammatory state. It activates the p38 MAPK pathway to upregulate MMP2 and MMP9 expression while downregulating TIMP1 and TIMP2, thereby altering the levels of ECM remodeling proteins. This suppresses proliferation and motility of dermal fibroblasts, thereby delaying wound repair in diabetic individuals (84).

Effects of oxidative stress on cell migration

Oxidative stress reflects a disrupted equilibrium where oxidation predominates over antioxidant defenses. Tissue in diabetic hyperglycemic wound microenvironments exhibits heightened vulnerability to this oxidative imbalance (85). Reactive oxygen species (ROS) serve a dual role in wound healing: Moderate levels promote tissue repair, while excess accumulation impairs wound closure and delays regenerative processes (86). High concentrations of ROS inhibit cell migration by oxidizing related proteins such as actin and myosin II, disrupting the structure and function of the cytoskeleton and impairing cell contractility (87). High glucose exacerbates oxidative stress, resulting in excessive Rac1 activation. This overactivation promotes the formation of unstable protrusions, disrupts cell polarity and impairs adhesion maturation, collectively decreasing cell migration speed and directionality, ultimately contributing to defective wound healing (72). A study has shown that hyperglycemia exacerbates oxidative stress, impairing the migratory and proliferative capacity of keratinocytes, thereby impairing wound repair in diabetic conditions (88). Moreover, oxidative stress decreases nuclear Nrf2 levels and manganese-superoxide dismutase (Mn-SOD) expression, thereby weakening the cellular antioxidant defense system, exacerbating ROS accumulation, impairing EC proliferation and migration and ultimately delaying tissue regeneration (89). MMP9 impedes diabetic wound repair (90,91). In human keratinocytes, ROS activate NF-κB, upregulating MMP-9 and suppressing keratinocyte migration, thereby delaying wound closure (92,93).

Effects of wound microenvironment on cell migration

Alterations in the wound microenvironment, such as deficiencies in key growth factors, changes in chemokine receptors and abnormal ECM remodeling, disrupt cell migration and impede wound healing. Diminished growth factor secretion in diabetic wounds disrupts cellular migration. During early wound repair, platelet-derived growth factor (PDGF) recruits fibroblasts, neutrophils and monocytes to the injury site (94). However, in diabetic wounds, PDGF and its receptor expression are downregulated, compromising cell migration and delaying wound closure (95). Similarly, hyperglycemia suppresses VEGF secretion by macrophages, fibroblasts and keratinocytes, impairing EC and keratinocyte migration, thereby hindering vascularization and re-epithelialization and delaying wound healing (94,96). TGF-β3 has been shown to facilitate the migration of fibroblasts and keratinocytes; considering the marked downregulation of TGF-β3 in diabetic wounds, restoring its activity locally may represent a viable approach to improving wound regeneration in diabetic patients (97). The alteration of chemokine receptors is also a key factor in impaired cell migration in diabetic wounds. Compared with healthy individuals, neutrophils from diabetic patients exhibit a substantial decrease in chemotaxis towards the chemokines CXCL8/IL-8, platelet-activating factor and fMLP (98). This attenuated chemotactic response may hinder cellular migration to the wound site, disrupting healing progression. Moreover, in chronic diabetic wounds, an imbalance in the regulation of MMPs disrupts ECM remodeling, impairing cell migration and delaying tissue repair (99,100). Normal function of MMP1 in keratinocytes is key for their migration on type I collagen (101). Hyperglycemia may impair keratinocyte migration via inhibition of the p-Stat-1 pathway and α2β1 integrin-dependent MMP1 activation, contributing to delayed diabetic wound healing (102). Hyperglycemia upregulates FOXO1, increasing MMP-9 while decreasing TGF-β1, thereby disrupting ECM homeostasis, impairing keratinocyte migration and delaying diabetic wound healing (100,103,104).

Other effects on cell migration

Complications of diabetes, including vasculopathy and neuropathy, are key pathological factors that impair wound healing by affecting cellular migration. Vascular disease in diabetic patients contributes to the delayed migration of white blood cells to injury sites (105). Diabetic neuropathy may disrupt keratinocyte and immune cell migration by altering neuropeptide release (substance P and calcitonin gene-related peptide), thereby impairing wound healing (106,107). Bacterial biofilms are highly structured, surface-associated microbial aggregates encased in a self-secreted extracellular polymeric substance, which provides mechanical stability and protects against environmental stresses (108). Diabetic wounds exhibit heightened susceptibility to infection and biofilm formation due to hyperglycemia-induced immunosuppression. The presence of these bacteria and their associated biofilms hinders cellular migration and disrupts the normal wound healing process (18,109). In diabetic wounds, aberrant mechanical signals may contribute to impaired cell migration function. Beyond its structural scaffolding role, the ECM also serves as a platform for initiating and integrating mechanotransduction signals. Under diabetic conditions, fibroblasts secrete a thicker and less porous ECM, hindering the migration of normal fibroblasts. Diabetic fibroblasts exhibit increased cellular stiffness yet generate markedly reduced traction and contractile forces within collagen matrices (110). These pathological changes may impair cell migration, disrupting wound contraction and delaying healing. Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) are the central effectors of the Hippo pathway and serve as the primary nuclear sensors for a variety of extracellular and intrinsic mechanical signals (111,112). Downregulation of Agrin (a key component of the ECM) in diabetic wounds may decrease MMP12 expression by inhibiting the nuclear localization of YAP/TAZ and its positive feedback regulation in keratinocytes (113). This weakens the cell ability to respond to mechanical stress, impairs their migration efficiency and ultimately delays wound healing. Mitochondria serve a crucial role in cell migration by providing ATP and maintaining calcium homeostasis (114). Mitochondrial dysfunction in diabetic wounds may be a key factor impairing cell migration. Sirtuin 3 (SIRT3), a key mitochondrial deacetylase, regulates energy metabolism and oxidative stress (115,116). SIRT3 deficiency in diabetic wounds disrupts mitochondrial structure and function, leading to oxidative stress, necroptosis, impaired migration of skin fibroblasts and delayed wound healing (117).

The impaired wound healing in diabetes arises from multifaceted interactions of various factors that compromise cellular migratory function. Research has predominantly focused on conventional mechanisms including metabolic dysregulation, oxidative stress, inflammatory responses and microenvironmental alterations (70,79,88,95). However, knowledge remains limited regarding regulatory pathways such as neural modulation, mechanical signals, mitochondrial dynamics, epigenetic modification and microbial community regulation (106,113,117). Future investigations should broaden the research scope to elucidate the precise roles of these factors in cellular migration, thereby providing theoretical foundations for developing targeted therapeutic strategies for diabetic wounds.

Key signaling pathways regulating cell migration to promote wound healing

During wound healing, cell migration is controlled by multiple signaling cascades, including Rho GTPase, PI3K/Akt, TGF-β/Smad and Wnt/β-catenin pathways (118-121). These signaling axes regulate key migratory processes such as cytoskeletal dynamics, cell polarization and force generation through distinct molecular mechanisms (122-125).

Rho GTPase signaling pathway

Rho GTPases serve as master regulators of cell migration, controlling cytoskeletal dynamics to facilitate directional cell movement (Fig. 4) (122). Rho GTPases are a distinct subclass within the Ras superfamily, serving as molecular switches that cycle between their biologically active GTP-bound conformation and inactive GDP-bound state. The Rho GTPase regulatory cycle is orchestrated by three protein families: Guanine nucleotide exchange factors, which promote GDP-to-GTP exchange to activate Rho GTPases; GTPase-activating proteins, which enhance GTP hydrolysis, inducing inactivation, and GDP dissociation inhibitors, which stabilize the inactive GDP-bound form and sequester Rho GTPases from membranes, thus maintaining quiescence (126).

Recent investigations into cell migration have largely centered on three key Rho GTPases: RhoA, Rac1 and Cdc42 (127-129). RhoA exhibits activity at both the leading and trailing edges of migrating cells, where it orchestrates actomyosin contractility via Rho-associated coiled-coil forming protein kinase (ROCK) and modulates actin polymerization via formin family nucleators (130). Rac1 initiates cytoskeletal remodeling by sequential activation of downstream effectors, first stimulating PAK kinase and the Wiskott-Aldrich syndrome protein (WASP) family verprolin-homologous protein (WAVE) regulatory complex. This signaling cascade induces WAVE-mediated direct activation of the actin-related protein (Arp) 2/3 complex, resulting in the nucleation of highly branched actin filament networks that mechanically drive lamellipodial membrane extension (122). Cdc42 orchestrates a distinct morphological response by specifically activating WASP family proteins, which serve as molecular scaffolds to facilitate precise Arp2/3 complex assembly and generation of parallel actin bundles, thereby promoting filopodial protrusion.

PI3K/Akt signaling pathway

The PI3K/Akt pathway is a central regulator of fundamental cellular functions, such as proliferative signaling, migratory behavior and immune modulation (Fig. 5) (131). Initiation of this pathway occurs through upstream membrane-associated receptors, such as receptor tyrosine kinases, integrins, antigen/cytokine receptors and GPCRs, which trigger PI3K activation (132). Following stimulation, PI3K mediates the phosphorylation of phosphatidylinositol-4,5-bisphosphate, generating the second messenger phosphatidylinositol-3,4,5-trisphosphate (PIP3). This lipid product recruits Akt to the membrane via interaction with its pleckstrin homology domain, enabling dual phosphorylation (Thr308/Ser473) and consequent functional activation (133). Once activated, Akt phosphorylates multiple downstream substrates in both the cytoplasm and nucleus, thereby modulating cellular motility.

Previous studies have suggested that the PI3K/Akt axis may contribute to wound repair by modulating the migratory dynamics of target cells (134,135). Through modulation of cytoskeletal component equilibrium, this pathway drives cell migration. Specifically, the PI3K/Akt-mediated signaling cascade activates Rho family small GTPases, including Rac1 and Cdc42, as well as actin-polymerization-promoting factors such as WASP/WAVE-Arp2/3 complexes (136). These components enhance actin nucleation and branched assembly at the advancing front, fostering lamellipodia growth and other protrusions that support cell motility. Secondly, the PI3K/Akt pathway modulates migratory capacity by regulating the dynamics of cell adhesion. Akt kinase activity enhances the formation and turnover of integrin-mediated focal adhesions, thereby allowing migrating cells to maintain traction at the leading edge while efficiently releasing adhesions at the rear (137). This mechanism ensures continuous and coordinated cell movement. Furthermore, this pathway exerts a key influence on chemotactic cell migration. When chemokines bind to GPCRs, the Gβγ subunit rapidly activates PI3K, particularly the PI3Kγ isoform, leading to increased PIP3 accumulation at the cell membrane leading edge. Subsequently, PIP3 recruits and activates Akt at the cell front, locally eliciting downstream pro-migratory effects and guiding the cell toward areas with higher chemokine concentrations (138).

TGF-β/Smad signaling pathway

The TGF-β superfamily signaling cascade regulates a wide range of cell processes, including proliferation, migration and ECM synthesis and reorganization (139). Central to this pathway is the TGF-β/Smad axis, which involves TGF-β ligands, cognate receptors (TGFβRI and TGFβRII) and downstream Smad mediators (Fig. 6). Upon ligand binding, TGFβRII interacts with TGF-β to assemble a heteromeric receptor complex, facilitating the recruitment and phosphorylation of TGFβRI (140). Activated TGFβRI exhibits kinase function, specifically phosphorylating Smad2 and Smad3. These phosphorylated Smads dimerize with Smad4, forming a transcriptionally active oligomeric complex that translocates into the nucleus to modulate target gene expression (141).

The TGF-β/Smad signaling pathway activates ROCK through the stimulation of RhoA (142). ROCK facilitates cell migration by modulating actin cytoskeletal rearrangement and myosin contractility. TGF-β signaling promotes keratinocyte migration and facilitates wound healing through the transcriptional regulation of key ECM-associated molecules. This includes the upregulation of integrin subunits (α5, αv and β5) as well as specific MMPs, particularly MMP3 and MMP9 (143,144). The TGF-β/Smad pathway serves as a key regulator of epithelial-mesenchymal transition (EMT) by downregulating E-cadherin and tight junction proteins, including occludin, claudins and zona occludens-1 (145). Simultaneously, elevated expression of mesenchymal markers, including N-cadherin and vimentin, reinforces cell motility, promoting the advancement of EMT.

Wnt/β-catenin signaling pathway

The Wnt/β-catenin signaling pathway, an evolutionarily conserved regulatory mechanism, governs a spectrum of biological functions including cellular proliferation, differentiation, apoptotic regulation and migratory behavior (Fig. 7) (146). In the absence of Wnt signaling, cytosolic β-catenin undergoes sequential phosphorylation mediated by the multiprotein degradation complex, comprising adenomatous polyposis coli, axin, casein kinase 1 and glycogen synthase kinase-3β, leading to its proteasomal degradation and thus ensuring minimal intracellular concentrations (147). Activation occurs upon binding of Wnt ligands (Wnt3a) to the Frizzled receptor family and its coreceptor low-density lipoprotein receptor-related protein 5/6, which disrupts the destabilization complex (148). This stabilization allows β-catenin to accumulate in the cytoplasm, undergo nuclear translocation and form complexes with T cell factor/lymphoid enhancer factor transcription factors, driving transcription of downstream target genes (149).

Wnt signaling induces localized activation of Rho family GTPases, including Rac1, at the leading edge of migrating cells. This stimulates actin polymerization, which is essential for driving forward cell motility (150). Wnt signaling promotes EMT through the suppression of E-cadherin and concurrent upregulation of mesenchymal markers such as vimentin and N-cadherin. This shift disrupts intercellular adhesion and augments the migratory potential of cells. In addition, Wnt/β-catenin signaling can upregulate the expression of MMPs, including MMP2, MMP7 and MMP9, thereby promoting the degradation of the ECM and facilitating cell migration (151,152). Activation of Wnt downstream targets serves a crucial role in cell migration, which significantly enhances wound healing processes. For example, Wnt1-inducible signaling pathway protein 1 stimulates proliferation and directional movement of dermal fibroblasts (153), while epidermal growth factor receptor (EGFR) activation is essential for keratinocyte recruitment to wound sites (154). Additionally, VEGF promotes mitogenic activity and chemotactic migration of ECs during neovascularization (155).

Crosstalk between signaling pathways may regulate cell migration during wound healing. Hypoxic conditioned medium from human amniotic fluid-derived mesenchymal stem cells (MSCs) promotes fibroblast migration and accelerates wound healing by modulating the TGF-β/SMAD2 and PI3K/Akt signaling pathways (156). Toraldo et al (157) demonstrated that topical androgen antagonism accelerates keratinocyte migration and promotes skin wound healing by inhibiting β-catenin nuclear translocation and its crosstalk with TGF-β signaling in keratinocytes. Furthermore, the Rho GTPase signaling pathway may serve as a common downstream node for other pathways to regulate cell migration (136,142,150). The types of cell migration regulated by different pathways exhibit distinct characteristics (Table I) (118-121,158-166).

Table I

Signaling pathways regulating migration of distinct cell types during diabetic wound healing.

Table I

Signaling pathways regulating migration of distinct cell types during diabetic wound healing.

First author/s, yearWound typeSignaling pathwayType of cell(Refs.)
Johan et al, 2024DiabeticRho GTPase (ROCK)Fibroblast(118)
Wang et al, 2022Diabetic foot ulcerRho GTPase (ROCK1)Fibroblast, endothelial cell(158)
Yu et al, 2020DiabeticRho GTPase (Rac1, ROCK)Fibroblast(159)
Li et al, 2025DiabeticPI3K/AktFibroblast(119)
Huang et al, 2025DiabeticPI3K/AktKeratinocyte, endothelial cell(160)
Xu et al, 2024DiabeticPI3K/AktEndothelial cell(161)
Gao et al, 2022DiabeticTGF-β/SmadKeratinocyte(120)
Peng et al, 2019DiabeticTGF-β1/SmadFibroblast(162)
Oyebode and Houreld, 2022DiabeticTGF-β1/SmadFibroblast(163)
Dong et al, 2025DiabeticWnt/β-cateninFibroblast(121)
Wang et al, 2024DiabeticWnt/β-cateninFibroblast(164)
Liu et al, 2023DiabeticWnt/β-cateninEndothelial cell(165)
Lv et al, 2020Diabetic cutaneousWnt/β-cateninKeratinocyte(166)

[i] ROCK, Rho-associated coiled-coil forming protein kinase; Rac, Ras-related C3 botulinum toxin substrate.

ncRNAs of cell migration in diabetic wounds

ncRNAs represent a diverse class of RNA transcripts that lack protein-coding potential yet serve key roles in modulating gene expression and orchestrating cell processes (167). Among these transcripts, distinct subcategories, including long ncRNAs (lncRNAs), microRNAs (miRNAs or miRs) and circular RNAs (circRNAs), have been identified and functionally characterized (168). Emerging evidence underscores the significance of ncRNAs in controlling migratory behaviors of cells and their impact on diabetic wound repair (Table II) (169-171). Given their regulatory versatility, targeting ncRNAs may offer novel therapeutic avenues for improving wound outcomes in diabetic patients.

Table II

ncRNAs regulating cell migration in diabetic wounds.

Table II

ncRNAs regulating cell migration in diabetic wounds.

First author/s, yearncRNAExosomeTargetType of cellEffect on cell migration(Refs.)
Hong et al, 2024lncRNA XISTN/A miR-126-3p/EGFRKeratinocytePromotion(174)
Chen et al, 2023lncRNA SNHG16N/AmiR-31-5pHuman dermal fibroblastInhibition(169)
He et al, 2022lncRNA CASC2N/AmiR-155/HIF-1αFibroblastPromotion(175)
Li et al, 2021lncRNA H19N/AmiR-29b, FBN1FibroblastPromotion(176)
Hu et al, 2020lnc-URIDSN/APlod1FibroblastInhibition(177)
Li et al, 2020lncRNA H19MSC-exosmiR-152-3p, PTENFibroblastPromotion(179)
Han et al, 2022lncRNA KLF3-AS1BMSCs-exomiR-383, VEGFAHUVECPromotion(180)
Fu et al, 2022LINC01435 Keratinocyte-exosYY1, HDAC8HUVECInhibition(181)
Peng et al, 2024miR-155N/AHIF-1α, SOX2, EGFR/MEK/ERKKeratinocyteInhibition(170)
Tsai et al, 2024miR-3138N/APALM2-AKAP2, SNX30, ZNF365KeratinocytePromotion(184)
Tsai et al, 2024miR-3679-5pN/ADMXL1, PPP2R2A, TTC39CKeratinocyteInhibition(184)
Zhao et al, 2023miR-204-3pN/AKLF6KeratinocytePromotion(185)
Zhang et al, 2022miR-146aN/AAKAP12KeratinocytePromotion(186)
Li et al, 2023miR-182-5pEPC-exosPPARGKeratinocytePromotion(188)
Lv, et al 2020miR-21-5phASC-exosWnt/β-cateninKeratinocytePromotion(166)
Wang et al, 2018miR-129, miR-335N/ASp1, MMP9KeratinocytePromotion(187)
Song et al, 2025miR-204-5pADSC-exosTGF-β1/SmadFibroblastPromotion(275)
Zheng et al, 2024miR-132-3pInsig1-exosMMP9, PDGF, VEGFDermal fibroblastPromotion(195)
Wang et al, 2024miR-145-5pN/APDGFDHuman foreskin fibroblastInhibition(192)
Wang et al, 2023miR-185-5pN/AIL-6, TNF-α, ICAM-1Human skin fibroblastPromotion(189)
Wu et al, 2023miR-16-5pN/ASP5Rat fibroblastPromotion(190)
Zhao et al, 2022miR-103N/ARCAN1Dermal fibroblastInhibition(193)
Zhang et al, 2020miR-27-3pN/ANOVA1FibroblastInhibition(194)
Wu et al, 2020miR-21-3pN/ASPRY1FibroblastPromotion(191)
Wang et al, 2022miR-199a-5pN/AVEGFA, ROCK1Human, foreskin fibroblast HUVECInhibition(158)
Zuo et al, 2024miR-488-3pN/AMeCP2, CYP1B1, Wnt4/β-cateninHUVECPromotion(196)
Qiu et al, 2024miR-221-3pBMSC-exosFOXP1/SPRY1HUVECPromotion(198)
Guo et al, 2024miR-125bADSC-exosCD34, Ki-67, VEGF, TGFβ1HUVECPromotion(200)
Che et al, 2024miR-146a-5pADSC-exosJAZF1HUVECPromotion(201)
Zhou et al, 2024miR-146a-5pBMSC-exosTRAF6HUVECPromotion(199)
Huang et al, 2023miR-204-3pN/AHIPK2HUVECPromotion(197)
Ge et al, 2023miR-132ADSC-exosNF-κBHUVECPromotion(202)
Yan et al, 2022miR-31-5pMilk-exosHIF1ANHUVECPromotion(205)
Huang et al, 2024circCDK13sEVsIGF2BP3, CD44, c-MYCHuman dermal fibroblast, human epidermal keratinocytePromotion(213)
Tian et al, 2023circ_072697N/AmiR-3150a-3p/KDM2A, MAPKKeratinocyteInhibition(171)
Fu et al, 2023circ_0080968N/AmiR-326, miR-766-3pKeratinocyteInhibition(209)
Han et al, 2021circ_PRKDCN/AmiR-31/FBN1KeratinocyteInhibition(210)
Wang et al, 2020 hsa_circ_0084443N/API3K, EGFR and ERK pathwaysKeratinocyteInhibition(211)
Wang et al, 2024circMYO9BMSCs-exos hnRNPU/CBL/KDM1A/VEGFAHUVECPromotion(285)
Liang et al, 2022 mmu_circ_0001052ADSCs-exosFGF4/p38MAPK pathwayHUVECPromotion(212)

[i] FBN1, fibrillin 1; YY1, yin yang 1; HDAC8, histone deacetylase 8; Plod1, procollagen-lysine, 2-oxoglutarate 5-dioxygenase 1; SOX2, sex determining region Y-box 2; PALM2-AKAP2, paralemmin 2-A kinase anchoring protein 2; SNX30, sorting nexin family member 30; ZNF365, zinc finger protein 365; DMXL1, Dmx-like 1; PPP2R2A, protein phosphatase 2; TTC39C, tetratricopeptide repeat domain 39C; KLF6, Kruppel-like factor 6; AKAP12, A-kinase-anchoring protein 12; PPARG, peroxisome proliferator activated receptor γ; Sp1, specificity protein-1; ICAM-1, intercellular adhesion molecule 1; SP5, transacting transcription factor 5; RCAN1, regulator of calcineurin; NOVA1, neuro-oncological ventral antigen 1; SPRY1, protein sprout homolog 1; ROCK1, Rho-associated kinase 1; MeCP2, methyl-CpG-binding protein 2; CYP1B1, cytochrome P450 1B1; FOXP1, forkhead box P1; JAZF1, juxtaposed with another zinc finger 1; TRAF6, tumor necrosis factor receptor-associated factor 6; HIPK2, homeodomain-interacting protein kinase 2; HIF1AN, hypoxia-inducible factor 1 subunit α inhibitor. IGF2BP3, insulin-like growth factor 2 mRNA binding protein 3; KDM2A, lysine demethylase 2 A; N/A, no information available; miR, microRNA; lnc, long non-coding; circ, circular.

lncRNAs

lncRNAs are ncRNAs >200 nucleotides in length (172). These molecules serve as master regulators in key biological activities, spanning cellular proliferation, lineage specification, embryogenesis, programmed cell death and metastatic dissemination. lncRNAs frequently function via the competitive endogenous RNA (ceRNA) network, where they sequester miRNAs to modulate the abundance of miRNA target transcripts (173). Hong et al (174) demonstrated that decreased lncRNA XIST expression in diabetic wounds elevates miR-126-3p levels, which subsequently suppresses EGFR (174). This inhibition impairs keratinocyte proliferation and migration in high-glucose environments, contributing to delayed wound healing. These observations indicate that XIST could serve as a promising therapeutic target to enhance keratinocyte motility during wound repair. lncRNAs CASC2 and H19 accelerate fibroblast migration in diabetic wounds through miRNA modulation, ultimately altering the expression of downstream effector genes (175,176). By contrast, lncRNA SNHG16 and lnc-URIDS have been found to inhibit fibroblast migration and impair wound healing (169,177). Exosomes represent a distinct subclass of extracellular vesicles that serve a pivotal role in cell-to-cell signaling by transporting bioactive cargo, including proteins, lipids and nucleic acids. lncRNAs, which are expressed at low levels in cells, tend to be enriched in exosome secretion (178). lncRNA H19 encapsulated in MSC-derived exosomes alleviates fibroblast apoptosis and inflammatory responses by attenuating miR-152-3p-dependent suppression of PTEN, enhancing fibroblast proliferation and motility to accelerate diabetic wound repair (179). Exosomal lncRNA KLF3-AS1 secreted by bone marrow MSCs (BMSCs) enhances EC proliferation, migration and angiogenic tube formation via the miR-383/VEGFA axis, where KLF3-AS1 competitively sponges miR-383 to decrease its suppression of VEGFA, ultimately accelerating diabetic cutaneous wound regeneration (180). Fu et al revealed that keratinocyte-derived exosomal LINC01435 inhibits the migration of human umbilical vein ECs (HUVECs) via the Yin Yang 1 (YY1)/histone deacetylase 8 (HDAC8) pathway, thereby suppressing angiogenesis and hindering diabetic wound healing (181).

miRNAs

miRNAs, a class of evolutionarily conserved small ncRNAs averaging 21-25 nucleotides in length (182), serve as critical mediators of post-transcriptional regulation, exerting their effects through either mRNA destabilization or translational suppression upon target binding (183). Certain miRNAs, including miR-3138, miR-204-3p, miR-146a, miR-129, and miR-335, promote keratinocyte migration through regulation of downstream targets or key signaling pathways, thereby accelerating wound healing in diabetic models (184-187). By contrast, elevated expression of miR-155 and miR-3679-5p suppresses keratinocyte migratory capacity, contributing to impaired re-epithelialization and protracted wound closure in diabetic conditions (170,184). Exosomes derived from endothelial progenitor cells deliver miR-182-5p, which downregulates PPARG expression, enhances keratinocyte proliferation and migration under hyperglycemic conditions, decreases apoptotic activity and ultimately facilitates diabetic wound repair (188). In addition, loading miR-21-5p into human adipose stem cell-derived exosomes facilitates keratinocyte proliferation and migration by activating the Wnt/β-catenin signaling pathway. This improves diabetic wound repair by concurrently stimulating re-epithelialization, optimizing collagen deposition and organization, boosting neovascularization and fostering functional vascular network maturation (166).

In a high-glucose environment, miR-185-5p, miR-16-5p, and miR-21-3p enhance the migratory capacity of fibroblasts (189-191). By contrast, miR-145-5p, miR-103, miR-27-3p, and miR-199a-5p exert a pronounced inhibitory effect on fibroblast migration in diabetic wounds (192-194,158). Recent research demonstrates that exosomes derived from insulin-induced gene 1 (Insig1)-overexpressing BMSCs (Insig1-exos), which are highly enriched with miR-132-3p, markedly enhance dermal fibroblast migration, proliferation and angiogenic activity under hyperglycemic conditions (195). Mechanistically, Insig1-exos modulate key wound repair mediators, including MMP9, PDGF and VEGF, thereby improving diabetic wound closure in murine models. These results underscore the key contribution of miR-132-3p in facilitating cellular motility and tissue regeneration.

Recent studies have demonstrated that miR-488-3p, miR-199a-5p and miR-204-3p facilitate diabetic wound repair by enhancing EC migration under hyperglycemia (158,196,197). Moreover, BMSCs release miR-221-3p and miR-146a-5p, while exosomes from adipose-derived SCs (ADSCs) deliver miR-125b, miR-146a-5p and miR-132, all of which collaboratively enhance EC migration, proliferation and angiogenic functions. These effects contribute to accelerated wound healing in diabetic models, underscoring their therapeutic promise (198-202). Milk-derived exosomes, obtained from both bovine colostrum and mature milk, present benefits such as excellent safety profiles, cost-effectiveness and high production yields (203). These natural vesicles exhibit remarkable drug-loading capacity and robust biological functionality across both in vitro and in vivo systems, highlighting their potential for pharmaceutical delivery and immunomodulatory applications (204). Notably, Yan et al (205) revealed that milk exosome-encapsulated miR-31-5p exhibits enhanced stability and cellular internalization (compared with free miR-31-5p mimics), stimulates EC proliferation, migration and neovascularization through hypoxia-inducible factor 1 subunit α inhibitor suppression and ultimately promotes diabetic wound repair.

circRNAs

circRNAs are a unique class of ncRNA molecules distinguished by their covalently closed-loop conformation, which confers resistance to exonuclease-mediated degradation (206). circRNAs function as 'sponges' to inhibit specific miRNAs, preventing their binding to target mRNAs, thereby serving as endogenous miRNA regulators (inhibitors) (207). Moreover, circRNAs modulate post-transcriptional gene expression and transcriptional processes through interactions with transcription factors and RNA-binding proteins (208). circ_072697, circ_0080968, circ_PRKDC and hsa_ circ_0084443 exert inhibitory effects on keratinocyte migration and impair diabetic wound healing by suppressing miRNAs or directly modulating target gene expression (171,209-211). ADSC-derived exosomes carrying mmu_circ_0001052 downregulate miR-106a-5p, which elevates fibroblast growth factor 4 (FGF4) levels, triggers the FGF4/p38MAPK signaling cascade and stimulates HUVEC proliferation, migration and angiogenic activity in high-glucose environments, collectively improving diabetic wound healing (212). Huang et al (213) reported that engineered small extracellular vesicles (sEVs) with circCDK13 overexpression bind insulin-like growth factor 2 mRNA-binding protein 3, stabilizing CD44 and c-MYC transcripts to enhance keratinocyte and fibroblast motility and division, thus accelerating wound closure in diabetic mouse models.

ncRNAs modulate cell migration via signaling pathways or downstream targets, presenting a promising intervention strategy for diabetic wounds. Both lncRNAs and circRNAs serve as ceRNAs, sequestering specific miRNAs to prevent their suppressive effects on target mRNAs, thereby indirectly regulating gene expression (171,174). By contrast, miRNAs directly bind to mRNAs to induce degradation or translational inhibition, forming an integrated miRNA-mRNA-functional protein regulatory network (170,186). Future studies should delineate the dynamic ncRNA-mediated regulatory networks in diabetic wound healing to develop precision-based therapeutic approaches for clinical translation.

Regulation of cell migration for the treatment of diabetic wounds

SCs and derived exosomes

SCs are undifferentiated cells characterized by their capacity for self-renewal and pluripotency, allowing differentiation into specialized lineages (214). Additionally, they secrete bioactive factors, modulate inflammatory responses, stimulate angiogenesis and enhance tissue remodeling, highlighting their therapeutic potential in regenerative medicine and tissue repair (Table III) (215). Their low immunogenicity and diverse sources enhance clinical potential. A recent investigation revealed that perinatal tissue-derived MSCs potentiate keratinocyte and EC proliferation and migration via PI3K/Akt pathway activation, culminating in accelerated healing of diabetic wounds (160). Furthermore, SCs can serve as carriers for drugs or bioactive molecules, facilitating sustained release while protecting them from degradation. Administration of genetically modified umbilical cord MSCs expressing angiopoietin-1 improves wound vascularization in diabetic murine models by stimulating EC migration and tubulogenesis, leading to faster healing kinetics (216).

Table III

SCs and exos regulating cell migration in diabetic wounds.

Table III

SCs and exos regulating cell migration in diabetic wounds.

First author/s, yearType of SCExosTargetType of cell migration(Refs.)
Huang et al, 2025MSCs derived from perinatal tissueN/API3K/AktKeratinocyte, HUVEC(160)
Deng et al, 2024MSCs derived from human umbilical cordsN/AANG1HUVEC(216)
He et al, 2024ADSCADSC-exosTRIM32/STINGHUVEC(218)
Liu et al, 2023GMSCGMSC-exosWnt/β-cateninHUVEC(165)

[i] TRIM32, tripartite motif-containing 32; STING, stimulator of interferon genes; N/A, no information available; MSC, mesenchymal stem cell; exo, exosome; ADSC, adipose-derived stem cell; GMSC, gingival mesenchymal stem cell; ANG, angiopoietin; HUVEC, human umbilical vein endothelial cell.

The application of SCs is limited by several factors, including a low survival rate, difficulty in controlling differentiation direction and the potential for immune rejection (217). By contrast, SC-derived exosomes, characterized by strong targeting ability, well-defined mechanisms of action and high safety profiles, offer a more precise and controllable cell-free therapeutic strategy for tissue repair. Beyond ncRNA-mediated regulation, SC-derived exosomes also promote cell migration and improve diabetic wound repair by targeting key signaling pathways. For example, ADSC-exos enhance EC migration and angiogenesis via the Tripartite motif-containing protein 32 (TRIM32)/STING axis, expediting wound closure in diabetic models (218). Liu et al (165) reported that exosomes isolated from gingival MSCs stimulate EC proliferation, migration and tube formation by activating the Wnt/β-catenin cascade, offering a potential therapeutic strategy for diabetic wound management.

SC therapy has demonstrated substantial therapeutic potential in diabetic wound repair; however, limitations persist (215). The precise mechanistic pathways governing its efficacy remain incompletely characterized, underscoring the need for further exploration of its molecular and cellular regulatory networks. Safety concerns need to be addressed by defining optimal dosages and administration routes to minimize potential adverse effects. Additionally, the complex processes involved in SC collection and pretreatment require simplification to enhance clinical feasibility and efficiency. The preparation and quality control of exosomes require the establishment of a unified standard (219). Additionally, due to the low molecular concentration of natural exosomes and their limited repair capabilities, exploring pretreatment methods, genetically engineered exosomes and the integration of exosomes with biomaterials may represent promising directions for development (220).

Growth factor therapy

Growth factor therapy for diabetic wound healing is based on its ability to coordinate key cellular responses and molecular pathways governing tissue regeneration. Key growth factors, including PDGF, VEGF, EGF, FGF and TGF, demonstrate potent pro-healing effects by stimulating cell migration and other regenerative processes that collectively accelerate wound repair (221). FGF-21 markedly improves EC proliferation, migration and angiogenic tube formation under hyperglycemic conditions, accelerating diabetic wound repair and underscoring its potential as a therapeutic agent for diabetic wounds (222). Tang et al (223) reported that PDGF-loaded nanocapsules with sustained release properties efficiently regulate fibroblast migration, proliferation and neovascularization, contributing to enhanced wound repair in diabetic models. Jeong et al (224) found that EGF encapsulated within gelatin-alginate coacervates enhances keratinocyte migration in vitro and accelerates wound closure in diabetic mice.

Growth factor therapy exhibits potential for promoting tissue regeneration; however, its clinical translation is hindered by rapid degradation, poor diffusion efficiency and insufficient local retention (221). Future studies should prioritize design of advanced biocompatible biomaterials with tunable degradation kinetics, alongside optimization of nanoscale delivery platforms to enable spatiotemporal control of drug release, targeted accumulation at wound sites and improved pharmacokinetic profile, which are key for expanding therapeutic utility in clinical settings.

Drug-loaded dressings
Hydrogel-based drug therapy

Hydrogels are three-dimensional polymeric networks distinguished by high hydration capacity and structural integrity, formed via cross-linked polymer chains. Owing to their biocompatibility, low immunogenicity and ability to retain moisture, hydrogels have gained prominence as an ideal biomaterial for diabetic wound management (Table IV) (225,226). These materials are capable of absorbing excess wound exudate, sustaining a moist microenvironment and preventing anaerobic bacterial proliferation through enhanced oxygen diffusion, facilitating cellular migration, tissue repair mechanisms and an accelerated healing trajectory (227,228). Recent studies have demonstrated that drug-loaded hydrogel dressings accelerate diabetic wound closure by facilitating cell motility and tissue remodeling (229,230). Notably, PDGF and cytokines stimulate ECM production, neovascularization and directed cellular movement, contributing to enhanced tissue repair (231). Xu et al (232) demonstrated that platelet-rich plasma-loaded multifunctional hydrogels exhibit dual therapeutic effects, suppressing excessive inflammation while shifting macrophage differentiation in favor of the regenerative M2 subset. This phenotypical modulation enhances migratory activity in both fibroblasts and vascular ECs, contributing to accelerated wound repair. This approach presents a clinically viable therapeutic paradigm for enhancing diabetic wound repair mechanisms. In addition, the incorporation of engineered sEVs into hydrogels prolongs their residence within the wound microenvironment, establishing them as optimal vehicles for bioactive molecule delivery. Wei et al (233) revealed that miR-17-5p-modified sEVs encapsulated in gelatin methacryloyl hydrogels enhance ECM remodeling via PTEN/p21 pathway modulation, thus stimulating both EC and fibroblast motility. Such a therapeutic strategy markedly improves the healing kinetics of diabetic wounds.

Table IV

Modulation of cell migration by drug-loaded dressings for diabetic wound healing.

Table IV

Modulation of cell migration by drug-loaded dressings for diabetic wound healing.

First author/s, yearDressing typeNameComponentsType of cell migration(Refs.)
Bei et al, 2024HydrogelPAN/Ag-PLG hydrogelGallic acid; functionalized polylysine; Ag-PLG, oxidized HA, cross-linked polyacrylic acid grafted with N-hydrosuccinimide esterHUVEC(229)
Li et al, 2022HydrogelHA-DA/MXene@ PDA hydrogelHA-DA; PDA; Ti3C2 MXene nanosheetsHUVEC(230)
Xu et al, 2023HydrogelPRP loaded multifunctional hydrogelPRP; DA-grafted alginate; 6-aminobenzo[c][1,2] oxaborol-1(3H)-ol-conjugated HAFibroblast, HUVEC(232)
Liu et al, 2022HydrogelB-G hydrogel Methacryloyl-substituted B; GFibroblast(286)
Wu et al, 2022HydrogelInjectable conductive and angiogenic hydrogelQuaternized chitosan; polyaniline; four-armed aldehyde-terminated polyethylene glycol; deferoxamineHUVEC(287)
Wei et al, 2024HydrogelGelMA hydrogel loaded with sEVs17−OE sEVs17−OE; GelMAFibroblast, HUVEC(233)
Wu et al, 2024AerogelTDNP functionalized aerogelTDNPsFibroblast(242)
John et al, 2023AerogelNanofiber aerogels with precision macrochannels and LL-37-mimic peptides Poly(glycolide-co-lactide) (90:10 glycolide: lactide); gelatin; poly-p-dioxanone; LL-37-mimic peptide W379Keratinocyte, fibroblast(245)
Yin et al, 2021Microneedle patchMN-MOF-GO-AgMg-MOF; poly(γ-glutamic acid) hydrogel; gallic acid; GO-Ag; Mg2+HUVEC(248)
Wang et al, 2023Microneedle patch MN-MgH2MgH2; poly(lactic-co-glycolic acid)Fibroblast, HUVEC(249)
Liu et al, 2023Microneedle patchDouble-layer drug-loaded microneedles (DMN@TH/rh-EGF)TH; rh-EGF; HA; carboxymethyl chitosan; gelatinHUVEC(288)

[i] PAN, polyacrylic acid grafted with N-hydrosuccinimide ester; PLG, polylysine-gallic acid; HUVEC, human umbilical vein endothelial cell; HA, hyaluronic acid; DA, dopamine; PDA, polydopamine; MXene, two-dimensional transition metal carbides, carbonitrides, and nitrides; PRP, platelet rich plasma; G, gelatin; B, Bletilla Striata polysaccharide; GelMA, Gelatin methacryloyl; sEVs17−OE, miR-17-5p-engineered small extracellular vesicles; miR, microRNA; TDNP, Turmeric-derived nanoparticle; LL-37, human cathelicidin antimicrobial peptide; MN, microneedle; MOF, magnesium organic framework; GO, graphene oxide; DMN@TH, double-layer microneedle loaded with tetracycline hydrochloride; rh-EGF, recombinant human epidermal growth factor.

Hydrogels face limitations in their application. Due to their time-dependent viscoelastic properties, long-term structural degradation and stress relaxation under load may occur, which can impair cell adhesion, migration and proliferation (234). Additionally, insufficient mechanical strength, challenges in controlling degradation rate and narrow functionality further restrict their practical use. Future integration of artificial intelligence-based screening with advanced 3D bioprinting platforms may simultaneously optimize the biomechanical performance, biofunctional characteristics and therapeutic applicability of next-generation hydrogels (235,236). Combined with personalized customization, these advancements may provide more efficient and precise solutions for wound healing.

Aerogel-based drug therapy

Aerogels are ultra-lightweight nanoporous materials formed by removing the liquid from gel pores to create interconnected porous structures (237). They exhibit rapid absorption of exudate while maintaining a moist wound environment and facilitating efficient gas exchange (238). Compared with conventional hydrogels and standard wound dressings, aerogels demonstrate superior structural characteristics, including ultralow density, minimal thermal conductivity, interconnected macroporosity and an extensive surface-to-volume ratio, which position them as promising alternatives for advanced wound management (Table IV) (239). Emerging evidence highlights the therapeutic potential of aerogel-based drug delivery systems in accelerating cell migration during wound regeneration (240,241). Wu et al (242) developed a turmeric nanoparticle-embedded aerogel dressing that demonstrates controlled drug release kinetics and potent anti-inflammatory and antioxidant activity, alongside enhanced fibroblast migration and proliferation. This formulation exhibited remarkable effectiveness in treating diabetic ulcers. Furthermore, LL-37, a cathelicidin-derived host defense peptide, serves key biological functions beyond its antimicrobial effects, including the modulation of keratinocyte and fibroblast activity to facilitate cutaneous wound closure (243,244). John et al (245) developed a nanofiber aerogel scaffold engineered with tailored macrochannels and LL-37 biomimetic peptides for diabetic wound therapy. The results demonstrated notable stimulation of both keratinocyte and fibroblast migratory activity and mitotic expansion, alongside marked enhancement in neovascularization and epidermal regeneration.

The application of aerogel requires further enhancement. Its intrinsic porous structure results in inadequate mechanical properties, low mechanical strength and susceptibility to fragmentation (238). Moreover, the intricate preparation process and high production costs hinder large-scale manufacturing and clinical adoption (242). In future, it may be feasible to enhance mechanical strength and flexibility through composite material design (by integrating with polymers), while simultaneously optimizing manufacturing processes, reducing expenses and developing more environmentally friendly and sustainable preparation methodologies.

Drug therapy based on microneedle patches

Microneedles are miniature, spine-like structures made from biocompatible materials, typically measuring from tens to hundreds of microns in size. Microneedle technology facilitates the penetration of the stratum corneum, enabling drug delivery, substance extraction or physical therapy targeting deep skin tissue (Table IV) (246). The microneedle patch integrates microneedle technology with a patch format, using tiny needle-like structures distributed on a substrate for transdermal drug delivery. Given their high exudate absorption capacity, robust bioadhesive performance and sustained drug release kinetics, microneedle patches have emerged as a promising therapeutic modality for chronic wound management (247). Yin et al (248) engineered a microneedle system incorporating magnesium-based organic frameworks, enabling efficient transdermal drug transport in diabetic wounds. This platform significantly augments EC migratory activity, stimulates neovascularization and accelerates tissue repair processes. Wang et al (249) developed a biodegradable poly (lactic-co-glycolic acid) microneedle patch loaded with magnesium hydride. This platform effectively scavenges ROS, induces a shift toward pro-regenerative M2 macrophage phenotypes and stimulates the proliferation and motility of fibroblasts and ECs, enhancing the healing trajectory of diabetic wounds.

Microneedle technology encounters several challenges, including limited drug-loading capacity, high production cost and insufficient stability in complex wound environments (246,250). Future advancements are required, such as optimizing materials and structures for the design of novel microneedles, assessing drug stability and enhancing biocompatibility and safety profiles. Additionally, the combination of micromachining and 3D printing techniques may streamline the manufacturing process and decrease production expenses (250).

Traditional chinese medicine (TCM) treatment

Phytochemicals derived from medicinal plants demonstrate multifunctional bioactive properties, including the stimulation of cellular proliferation and migratory capacity, potent antimicrobial effects and the induction of neovascularization, all of which contribute to enhanced tissue regeneration (Table V) (251,252). Recent research has shown that topical administration of Crocus sativus L. (saffron) petal extract markedly accelerates diabetic wound repair by elevating Collagen type I alpha 1 and VEGF levels, stimulating fibroblast and EC motility and enhancing overall re-epithelialization in mice (253). Ginsenoside Rg1 (Rg1), a principal active component derived from Panax ginseng, exerts pro-angiogenic effects by stimulating the proliferation and migration of ECs, thereby facilitating wound repair in diabetic wounds. Mechanistically, Rg1 downregulates miR-48-3p, elevates Sirt1 expression and triggers the PI3K/AKT/endothelial nitric oxide synthase) cascade, collectively enhancing vascular regeneration (254). Similarly, paeoniflorin, a key monoterpene glycoside isolated from Paeoniae alba radix, was demonstrated by Sun et al to attenuate oxidative damage while promoting keratinocyte proliferation and motility (255). These reparative effects are achieved via Nrf2 pathway activation coupled with increased VEGF and TGF-β1 production, expediting diabetic wound closure in rats.

Table V

Regulation of cell migration in diabetic wound healing by traditional Chinese medicine.

Table V

Regulation of cell migration in diabetic wound healing by traditional Chinese medicine.

First author/s, yearDrug categoryNameTargetType of cell migration(Refs.)
Soheilifar et al, 2024Natural productSaffron (Crocus Sativus L.) petal extractCOL1A1, VEGFFibroblast, HUVEC(253)
Xiong et al, 2024Natural productAstragaloside IVPIK3R2, VEGF/PI3K/AKTHUVEC(289)
Lei et al, 2022Natural productPanax notoginseng saponins GSK-3β/β-catenin/VEGFHUVEC(290)
Huang et al, 2021Natural productGinsenoside Rg1miR-489-3p/Sirt1, PI3K/AKT/eNOSHUVEC(254)
Sun et al, 2020Natural productPaeoniflorinNrf2, VEGF, TGF-β1Keratinocyte(255)
Lu et al, 2021Herbal formulaQuyu Shengji formulaPGTHuman dermal microvascular endothelial cell(291)
Zhang et al, 2024Herbal formulaDang-Gui-Si-Ni decoction AGE/RAGE/TGF-β/Smad2/3Fibroblast(256)
Gong et al, 2022Herbal formulaMoist exposed burn ointmentN/AKeratinocyte(257)
Liu et al, 2023Herbal formulaPien-tze-huangNrf2/AREHUVEC(292)

[i] COL1A1, collagen type I α1; PIK3R2, phosphoinositol-3 kinase regulatory subunit 2; GSK-3β, glycogen synthase kinase-3β; PGT, prostaglandin transporter; ARE, antioxidant response element; N/A, no information available; HUVEC, human umbilical vein endothelial cell; miR, microRNA; Sirt, Sirtuin; eNOS, endothelial nitric oxide synthase; RAGE, receptor for advanced glycation end-products.

Chinese herbal formulas have potential in facilitating cell migration in diabetic wound healing (256,257). Danggui Sini decoction (DSD), a TCM formulation, exhibits multi-target pharmacological actions such as vasodilatory, anti-inflammatory and antioxidant activity (258). Mechanistic study has revealed that DSD facilitates diabetic wound repair by augmenting fibroblast proliferation and migratory capacity, mediated via regulation of the AGE/RAGE (Receptor for advanced glycation end-products)/TGF-β/Smad2/3 signaling axis in diabetic foot ulcer rats (256). Moist exposed burn ointment, a herbal oil-based preparation, is utilized for burn management and chronic refractory wound care due to its clinical effectiveness (259). When applied to diabetic wounds, as demonstrated by Gong et al (257), this formulation accelerates tissue regeneration by stimulating keratinocyte migration, promoting granulation tissue development and collagen reorganization and enhancing re-epithelialization.

While TCM demonstrates therapeutic promise in enhancing diabetic wound repair, several challenges remain to be resolved, including poorly characterized molecular mechanisms, intricate multi-component formulations and restricted administration options. Overcoming these limitations requires systematic research strategies to elucidate fundamental mechanisms, optimize bioactive compound extraction protocols, develop novel delivery systems and validate therapeutic effects through multicenter clinical studies.

Additional treatment options

Diabetic foot ulcers are frequently attributed to inadequate blood supply to the lower limb vessels, leading to localized hypoxia in the wound and consequently impairing the healing process. Hyperbaric oxygen therapy (HBOT) serves as an adjunctive therapy that elevates oxygen concentrations in arterial blood and tissues (260). This therapeutic intervention involves the administration of 100% oxygen in a pressurized chamber, elevating environmental pressure to 2-3 atmospheres absolute (261). Under hyperbaric conditions, tissue hypoxia is alleviated, improving oxygenation for key metabolic processes, cellular proliferation and wound repair. HBOT stimulates fibroblast and EC activity via HIF-1α pathway activation, which enhances vascularization and accelerates healing of diabetic wounds (262).

Negative pressure wound therapy (NPWT) is a non-surgical therapeutic approach utilizing an airtight dressing system to achieve localized sub-atmospheric pressure at the wound bed, facilitating enhanced tissue perfusion and wound closure. This therapy effectively removes wound exudate and necrotic tissue, decreases tissue edema, promotes the growth of granulation tissue and angiogenesis, thereby providing optimal conditions for wound healing (263,264). Huang et al (265) revealed that NPWT promotes human dermal fibroblast proliferation and migration via miR-155 downregulation in diabetic wound granulation tissue, concurrently augmenting FGF7 expression to accelerate wound repair (265). Liu et al (266) demonstrated that NPWT stimulates keratinocyte proliferation and migration by suppressing hsa-miR-203, which elevates p63 protein levels in both peripheral blood and wound edge tissue, contributing to enhanced diabetic wound healing.

Photobiomodulation (PBM), commonly known as low-intensity laser therapy, is a non-interventional treatment approach that employs low-power optical radiation, typically delivered via lasers or light-emitting diodes (267). PBM enhances wound closure and tissue regeneration, with optimal therapeutic outcomes depend on precise selection of wavelength and fluence parameters (268,269). PBM at 830 nm (5 J/cm2 fluence) significantly boosts fibroblast viability, migration and proliferative capacity via activation of the TGF-β1/Smad pathway, leading to accelerated healing of diabetic wounds (163). Cai et al (270) examined dual-wavelength (red/blue) phototherapy in diabetic rats, observing substantial decreases in inflammatory markers and ROS accumulation alongside enhanced EC activity. This combined approach promotes NO synthesis and markedly improves wound closure rates.

Filgrastim, a recombinant human granulocyte colony-stimulating factor analog, promotes both neutrophil progenitor differentiation and functional maturation (271). Additionally, this cytokine directs neutrophil trafficking toward inflammatory and infectious foci, amplifying localized immune defenses via targeted cellular recruitment. A retrospective analysis of patients with infectious diabetic wounds demonstrated that those treated with filgrastim exhibited significantly faster recovery times (272). This indicates the potential therapeutic value of filgrastim in enhancing infection control and promoting wound healing via increased neutrophil production and migration, bolstered immune response and accelerated tissue repair.

There are limitations in the application of the aforementioned therapies. The high cost of treatment and reliance on specialized equipment restrict the widespread adoption of HBOT. Future research should focus on optimizing treatment parameters, decreasing cost and investigating combination therapies. NPWT may induce pain and skin damage, with limited efficacy for infected wounds. Advances in dressing materials and refined control of negative pressure are required to minimize adverse reactions (273). PBM lacks standardized therapeutic parameters, such as wavelength and fluence, and its efficacy varies between individuals (268,269). Large-scale clinical trials are necessary to establish optimal parameters and indications. Filgrastim may lead to overactivation of neutrophils, potentially causing increased inflammation and other adverse reactions (274). Future studies should aim to optimize dosing regimens and develop novel drugs to enhance therapeutic outcomes.

Conclusion

The present review summarizes cell migration dynamics in diabetic wounds, with a focus on cellular mechanisms, signaling cascades, ncRNA-mediated regulation and their translational implications for targeted therapies. Emerging therapies, such as SCs, exosomes, drug-loaded dressings and TCM, enhance cell migration via ncRNA-mediated signaling (160,275). This establishes regulatory axes of drug/therapy-ncRNA-signaling pathway/downstream target-cell migration (166,254). These breakthroughs substantially enhance understanding of diabetic wound pathological mechanisms while establishing a framework for targeted therapeutic development.

Although the mechanisms underlying abnormal cell migration in diabetic wounds and targeted therapeutic approaches have seen advancements, notable gaps remain. The majority of studies emphasize the regulation of individual cell types or specific signaling pathways, with limited exploration of cellular interactions and signaling crosstalk (160,165,166,275). Research on the regulation of cell migration primarily focuses on ncRNAs, whereas other epigenetic modifications, such as DNA methylation and histone modification, warrant further investigation (170,177,192,211). Most studies rely on in vitro experiments or animal models, which differ from the complex pathological environment of the human body, limiting their clinical translation and necessitating further validation (170,253). Despite their growing use, SC/exosome and growth factor therapy, advanced drug-loaded dressings and TCM intervention lack comprehensive clinical trial data to confirm their long-term safety and therapeutic efficacy. Furthermore, given the high heterogeneity of patients, there is a lack of research on personalized treatment approaches in existing studies, which restricts broader clinical application (276-278).

Future research should explore crosstalk between immune cells, fibroblasts, keratinocytes and ECs in diabetic wounds, focusing on key pathways. The application of organoids or 3D-printed tissue models may facilitate the development of more accurate models that closely mimic the human pathological environment (279,280). It is essential to refine the preparation and delivery technologies for SCs and exosomes, enhance the manufacturing processes of drug-loaded dressings and design intelligent dressing delivery systems to improve the precision and control of therapeutic interventions (281-283). The integration of topical TCM agents with advanced wound dressings may enhance therapeutic efficacy, presenting a potential strategy for diabetic wound management. Large-scale, multi-center clinical trials are required to validate the efficacy of existing treatments. Integrating multi-omics techniques with artificial intelligence-based analysis to explore personalized treatment strategies will aid in achieving precise intervention tailored to individual patient characteristics (284). Ultimately, it is essential to enhance multi-disciplinary collaboration between basic research and clinical practice, thereby facilitating the translation of research findings into practical applications and providing more efficient and safer solutions for the treatment of diabetic wounds.

Availability of data and materials

Not applicable.

Authors' contributions

JLS designed the study, wrote the manuscript and constructed figures. TZ and CW performed the literature review and created figures. XS, JCS and ZZ revised the manuscript. JCS and ZZ supervised the study and acquired funding. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

The present study was supported by the Construction Project of the TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine (grant no. 202206), the Construction Project of the Institute of Traditional Chinese Medicine and Integrated Chinese and Western Medicine under the Tianjin Municipal Health Commission (grant no. 202455) and the Research Planning Projects of the Tianjin Municipal Education Commission (grant no. 2024ZD014).

References

1 

Dwivedi J, Sachan P, Wal P, Wal A and Rai AK: Current state and future perspective of diabetic wound healing treatment: Present evidence from clinical trials. Curr Diabetes Rev. 20:e2808232204052024. View Article : Google Scholar

2 

Sun H, Pulakat L and Anderson DW: Challenges and new therapeutic approaches in the management of chronic wounds. Curr Drug Targets. 21:1264–1275. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Burgess JL, Wyant WA, Abujamra BA, Kirsner RS and Jozic I: Diabetic wound-healing science. Medicina (Kaunas). 57:10722021. View Article : Google Scholar : PubMed/NCBI

4 

Vijayakumar V, Samal SK, Mohanty S and Nayak SK: Recent advancements in biopolymer and metal nanoparticle-based materials in diabetic wound healing management. Int J Biol Macromol. 122:137–148. 2019. View Article : Google Scholar

5 

Armstrong DG, Tan TW, Boulton AJM and Bus SA: Diabetic foot ulcers: A review. JAMA. 330:62–75. 2023. View Article : Google Scholar : PubMed/NCBI

6 

Dasari N, Jiang A, Skochdopole A, Chung J, Reece EM, Vorstenbosch J and Winocour S: Updates in diabetic wound healing, inflammation, and scarring. Semin Plast Surg. 35:153–158. 2021. View Article : Google Scholar : PubMed/NCBI

7 

GBD 2021 Diabetes Collaborators: Global, regional, and national burden of diabetes from 1990 to 2021, with projections of prevalence to 2050: A systematic analysis for the global burden of disease study 2021. Lancet. 402:203–234. 2023. View Article : Google Scholar : PubMed/NCBI

8 

Feng J, Yao Y, Wang Q, Han X, Deng X, Cao Y, Chen X, Zhou M and Zhao C: Exosomes: Potential key players towards novel therapeutic options in diabetic wounds. Biomed Pharmacother. 166:1152972023. View Article : Google Scholar : PubMed/NCBI

9 

Peña OA and Martin P: Cellular and molecular mechanisms of skin wound healing. Nat Rev Mol Cell Biol. 25:599–616. 2024. View Article : Google Scholar : PubMed/NCBI

10 

Hanson AJ and Quinn MT: Effect of fibrin sealant composition on human neutrophil chemotaxis. J Biomed Mater Res. 61:474–481. 2002. View Article : Google Scholar : PubMed/NCBI

11 

Liu C, Lu Y, Du P, Yang F, Guo P, Tang X, Diao L and Lu G: Mesenchymal stem cells pretreated with proinflammatory cytokines accelerate skin wound healing by promoting macrophages migration and M2 polarization. Regen Ther. 21:192–200. 2022. View Article : Google Scholar : PubMed/NCBI

12 

Wang S, Wang K, Xin Y and Lv D: Maggot excretions/secretions induces human microvascular endothelial cell migration through AKT1. Mol Biol Rep. 37:2719–2725. 2010. View Article : Google Scholar

13 

Xia W, Li M, Jiang X, Huang X, Gu S, Ye J, Zhu L, Hou M and Zan T: Young fibroblast-derived exosomal microRNA-125b transfers beneficial effects on aged cutaneous wound healing. J Nanobiotechnology. 20:1442022. View Article : Google Scholar : PubMed/NCBI

14 

Fang PH, Lai YY, Chen CL, Wang HY, Chang YN, Lin YC, Yan YT, Lai CH and Cheng B: Cobalt protoporphyrin promotes human keratinocyte migration under hyperglycemic conditions. Mol Med. 28:712022. View Article : Google Scholar : PubMed/NCBI

15 

Huang SM, Wu CS, Chiu MH, Yang HJ, Chen GS and Lan CCE: High-glucose environment induced intracellular O-GlcNAc glycosylation and reduced galectin-7 expression in keratinocytes: Implications on impaired diabetic wound healing. J Dermatol Sci. 87:168–175. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Song Q, An X, Li D, Sodha NR, Boodhwani M, Tian Y, Sellke FW and Li J: Hyperglycemia attenuates angiogenic capability of survivin in endothelial cells. Microvasc Res. 78:257–264. 2009. View Article : Google Scholar : PubMed/NCBI

17 

Huttenlocher A: Cell polarization mechanisms during directed cell migration. Nat Cell Biol. 7:336–337. 2005. View Article : Google Scholar : PubMed/NCBI

18 

Brothers KM, Stella NA, Hunt KM, Romanowski EG, Liu X, Klarlund JK and Shanks RMQ: Putting on the brakes: Bacterial impediment of wound healing. Sci Rep. 5:140032015. View Article : Google Scholar : PubMed/NCBI

19 

Carragher NO, Fincham VJ, Riley D and Frame MC: Cleavage of focal adhesion kinase by different proteases during SRC-regulated transformation and apoptosis. Distinct roles for calpain and caspases. J Biol Chem. 276:4270–4275. 2001. View Article : Google Scholar

20 

Zhao Y, Wang Y, Sarkar A and Wang X: Keratocytes generate high integrin tension at the trailing edge to mediate rear de-adhesion during rapid cell migration. iScience. 9:502–512. 2018. View Article : Google Scholar : PubMed/NCBI

21 

Clayton SM, Shafikhani SH and Soulika AM: Macrophage and neutrophil dysfunction in diabetic wounds. Adv Wound Care (New Rochelle). 13:463–484. 2024. View Article : Google Scholar : PubMed/NCBI

22 

Rousselle P, Braye F and Dayan G: Re-epithelialization of adult skin wounds: Cellular mechanisms and therapeutic strategies. Adv Drug Deliv Rev. 146:344–365. 2019. View Article : Google Scholar

23 

Rodrigues M, Kosaric N, Bonham CA and Gurtner GC: Wound healing: A cellular perspective. Physiol Rev. 99:665–706. 2019. View Article : Google Scholar :

24 

Su Y and Richmond A: Chemokine regulation of neutrophil infiltration of skin wounds. Adv Wound Care (New Rochelle). 4:631–640. 2015. View Article : Google Scholar : PubMed/NCBI

25 

Dahlgren C, Gabl M, Holdfeldt A, Winther M and Forsman H: Basic characteristics of the neutrophil receptors that recognize formylated peptides, a danger-associated molecular pattern generated by bacteria and mitochondria. Biochem Pharmacol. 114:22–39. 2016. View Article : Google Scholar : PubMed/NCBI

26 

Mamun AA, Shao C, Geng P, Wang S and Xiao J: Recent advances in molecular mechanisms of skin wound healing and its treatments. Front Immunol. 15:13954792024. View Article : Google Scholar : PubMed/NCBI

27 

Lämmermann T, Afonso PV, Angermann BR, Wang JM, Kastenmüller W, Parent CA and Germain RN: Neutrophil swarms require LTB4 and integrins at sites of cell death in vivo. Nature. 498:371–375. 2013. View Article : Google Scholar : PubMed/NCBI

28 

Lee WL, Harrison RE and Grinstein S: Phagocytosis by neutrophils. Microbes Infect. 5:1299–1306. 2003. View Article : Google Scholar : PubMed/NCBI

29 

McLeish KR and Fernandes MJ: Understanding inhibitory receptor function in neutrophils through the lens of CLEC12A. Immunol Rev. 314:50–68. 2023. View Article : Google Scholar

30 

Fetz AE and Bowlin GL: Neutrophil extracellular traps: Inflammation and biomaterial preconditioning for tissue engineering. Tissue Eng Part B Rev. 28:437–450. 2022. View Article : Google Scholar

31 

Bratton DL and Henson PM: Neutrophil clearance: When the party is over, clean-up begins. Trends Immunol. 32:350–357. 2011. View Article : Google Scholar : PubMed/NCBI

32 

Guo S and DiPietro LA: Factors affecting wound healing. J Dent Res. 89:219–229. 2010. View Article : Google Scholar : PubMed/NCBI

33 

de Oliveira S, Rosowski EE and Huttenlocher A: Neutrophil migration in infection and wound repair: Going forward in reverse. Nat Rev Immunol. 16:378–391. 2016. View Article : Google Scholar : PubMed/NCBI

34 

Chen WYJ and Rogers AA: Recent insights into the causes of chronic leg ulceration in venous diseases and implications on other types of chronic wounds. Wound Repair Regen. 15:434–449. 2007. View Article : Google Scholar : PubMed/NCBI

35 

Isles HM, Herman KD, Robertson AL, Loynes CA, Prince LR, Elks PM and Renshaw SA: The CXCL12/CXCR4 signaling axis retains neutrophils at inflammatory sites in zebrafish. Front Immunol. 10:17842019. View Article : Google Scholar : PubMed/NCBI

36 

Wood W: Wound healing: Calcium flashes illuminate early events. Curr Biol. 22:R14–R16. 2012. View Article : Google Scholar : PubMed/NCBI

37 

Niethammer P, Grabher C, Look AT and Mitchison TJ: A tissue-scale gradient of hydrogen peroxide mediates rapid wound detection in zebrafish. Nature. 459:996–999. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Razzell W, Evans IR, Martin P and Wood W: Calcium flashes orchestrate the wound inflammatory response through DUOX activation and hydrogen peroxide release. Curr Biol. 23:424–429. 2013. View Article : Google Scholar : PubMed/NCBI

39 

Minutti CM, Knipper JA, Allen JE and Zaiss DM: Tissue-specific contribution of macrophages to wound healing. Semin Cell Dev Biol. 61:3–11. 2017. View Article : Google Scholar

40 

Dutra FF and Bozza MT: Heme on innate immunity and inflammation. Front Pharmacol. 5:1152014. View Article : Google Scholar : PubMed/NCBI

41 

Sindrilaru A and Scharffetter-Kochanek K: Disclosure of the culprits: Macrophages-versatile regulators of wound healing. Adv Wound Care (New Rochelle). 2:357–368. 2013. View Article : Google Scholar

42 

Gurtner GC, Werner S, Barrandon Y and Longaker MT: Wound repair and regeneration. Nature. 453:314–321. 2008. View Article : Google Scholar : PubMed/NCBI

43 

Zhang T, Tai Z, Miao F, Zhao Y, Wang W, Zhu Q and Chen Z: Bioinspired nanovesicles derived from macrophage accelerate wound healing by promoting angiogenesis and collagen deposition. J Mater Chem B. 12:12338–12348. 2024. View Article : Google Scholar : PubMed/NCBI

44 

Werner S, Krieg T and Smola H: Keratinocyte-fibroblast interactions in wound healing. J Invest Dermatol. 127:998–1008. 2007. View Article : Google Scholar : PubMed/NCBI

45 

Rousselle P, Montmasson M and Garnier C: Extracellular matrix contribution to skin wound re-epithelialization. Matrix Biol. 75-76:12–26. 2019. View Article : Google Scholar

46 

Rousselle P and Beck K: Laminin 332 processing impacts cellular behavior. Cell Adh Migr. 7:122–134. 2013. View Article : Google Scholar :

47 

Hamill KJ and McLean WHI: The alpha-3 polypeptide chain of laminin 5: Insight into wound healing responses from the study of genodermatoses. Clin Exp Dermatol. 30:398–404. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Rousselle P, Lunstrum GP, Keene DR and Burgeson RE: Kalinin: An epithelium-specific basement membrane adhesion molecule that is a component of anchoring filaments. J Cell Biol. 114:567–576. 1991. View Article : Google Scholar : PubMed/NCBI

49 

Theveneau E and Mayor R: Collective cell migration of epithelial and mesenchymal cells. Cell Mol Life Sci. 70:3481–3492. 2013. View Article : Google Scholar : PubMed/NCBI

50 

Krawczyk WS: A pattern of epidermal cell migration during wound healing. J Cell Biol. 49:247–263. 1971. View Article : Google Scholar : PubMed/NCBI

51 

Headon D: Reversing stratification during wound healing. Nat Cell Biol. 19:595–597. 2017. View Article : Google Scholar : PubMed/NCBI

52 

Freedberg IM, Tomic-Canic M, Komine M and Blumenberg M: Keratins and the keratinocyte activation cycle. J Invest Dermatol. 116:633–640. 2001. View Article : Google Scholar : PubMed/NCBI

53 

Veith AP, Henderson K, Spencer A, Sligar AD and Baker AB: Therapeutic strategies for enhancing angiogenesis in wound healing. Adv Drug Deliv Rev. 146:97–125. 2019. View Article : Google Scholar :

54 

Velnar T and Gradisnik L: Tissue augmentation in wound healing: The role of endothelial and epithelial cells. Med Arch. 72:444–448. 2018. View Article : Google Scholar

55 

Lamalice L, Le Boeuf F and Huot J: Endothelial cell migration during angiogenesis. Circ Res. 100:782–794. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Al Sadoun H: Macrophage phenotypes in normal and diabetic wound healing and therapeutic interventions. Cells. 11:24302022. View Article : Google Scholar : PubMed/NCBI

57 

Song J, Wu Y, Chen Y, Sun X and Zhang Z: Epigenetic regulatory mechanism of macrophage polarization in diabetic wound healing (Review). Mol Med Rep. 31:22025. View Article : Google Scholar

58 

Morbidelli L, Genah S and Cialdai F: Effect of microgravity on endothelial cell function, angiogenesis, and vessel remodeling during wound healing. Front Bioeng Biotechnol. 9:7200912021. View Article : Google Scholar : PubMed/NCBI

59 

Hsu S, Thakar R, Liepmann D and Li S: Effects of shear stress on endothelial cell haptotaxis on micropatterned surfaces. Biochem Biophys Res Commun. 337:401–409. 2005. View Article : Google Scholar : PubMed/NCBI

60 

Li S, Huang NF and Hsu S: Mechanotransduction in endothelial cell migration. J Cell Biochem. 96:1110–1126. 2005. View Article : Google Scholar : PubMed/NCBI

61 

Lynch MD and Watt FM: Fibroblast heterogeneity: Implications for human disease. J Clin Invest. 128:26–35. 2018. View Article : Google Scholar : PubMed/NCBI

62 

Talbott HE, Mascharak S, Griffin M, Wan DC and Longaker MT: Wound healing, fibroblast heterogeneity, and fibrosis. Cell Stem Cell. 29:1161–1180. 2022. View Article : Google Scholar : PubMed/NCBI

63 

Bussone G: Subjectivity in primary headaches: Insight the causes. Neurol Sci. 38:1–2. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Younesi FS, Miller AE, Barker TH, Rossi FMV and Hinz B: Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat Rev Mol Cell Biol. 25:617–638. 2024. View Article : Google Scholar : PubMed/NCBI

65 

Shao DD, Suresh R, Vakil V, Gomer RH and Pilling D: Pivotal advance: Th-1 cytokines inhibit, and Th-2 cytokines promote fibrocyte differentiation. J Leukoc Biol. 83:1323–1333. 2008. View Article : Google Scholar : PubMed/NCBI

66 

Grieb G, Steffens G, Pallua N, Bernhagen J and Bucala R: Circulating fibrocytes-biology and mechanisms in wound healing and scar formation. Int Rev Cell Mol Biol. 291:1–19. 2011. View Article : Google Scholar

67 

Park JG, Lim DC, Park JH, Park S, Mok J, Kang KW and Park J: Benzbromarone induces targeted degradation of HSP47 protein and improves hypertrophic scar formation. J Invest Dermatol. 144:633–644. 2024. View Article : Google Scholar

68 

Kohlhauser M, Mayrhofer M, Kamolz LP and Smolle C: An update on molecular mechanisms of scarring-A narrative review. Int J Mol Sci. 25:115792024. View Article : Google Scholar : PubMed/NCBI

69 

Hinz B: Formation and function of the myofibroblast during tissue repair. J Invest Dermatol. 127:526–537. 2007. View Article : Google Scholar : PubMed/NCBI

70 

Luo L, An Y, Geng K, Wan S, Zhang F, Tan X, Jiang Z and Xu Y: High glucose-induced endothelial STING activation inhibits diabetic wound healing through impairment of angiogenesis. Biochem Biophys Res Commun. 668:82–89. 2023. View Article : Google Scholar : PubMed/NCBI

71 

Huang SM, Wu CS, Chiu MH, Wu CH, Chang YT, Chen GS and Lan CCE: 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. 96:159–167. 2019. View Article : Google Scholar : PubMed/NCBI

72 

Lamers ML, Almeida MES, Vicente-Manzanares M, Horwitz AF and Santos MF: High glucose-mediated oxidative stress impairs cell migration. PLoS One. 6:e228652011. View Article : Google Scholar : PubMed/NCBI

73 

Brubaker AL, Rendon JL, Ramirez L, Choudhry MA and Kovacs EJ: Reduced neutrophil chemotaxis and infiltration contributes to delayed resolution of cutaneous wound infection with advanced age. J Immunol. 190:1746–1757. 2013. View Article : Google Scholar : PubMed/NCBI

74 

den Dekker A, Davis FM, Kunkel SL and Gallagher KA: Targeting epigenetic mechanisms in diabetic wound healing. Transl Res. 204:39–50. 2019. View Article : Google Scholar :

75 

Kasuya A and Tokura Y: Attempts to accelerate wound healing. J Dermatol Sci. 76:169–172. 2014. View Article : Google Scholar : PubMed/NCBI

76 

Lan CCE, Liu IH, Fang AH, Wen CH and Wu CS: Hyperglycaemic conditions decrease cultured keratinocyte mobility: Implications for impaired wound healing in patients with diabetes. Br J Dermatol. 159:1103–1115. 2008.PubMed/NCBI

77 

Zhou P, Feng H, Qin W and Li Q: KRT17 from skin cells with high glucose stimulation promotes keratinocytes proliferation and migration. Front Endocrinol (Lausanne). 14:12370482023. View Article : Google Scholar : PubMed/NCBI

78 

Worsley AL, Lui DH, Ntow-Boahene W, Song W, Good L and Tsui J: The importance of inflammation control for the treatment of chronic diabetic wounds. Int Wound J. 20:2346–2359. 2023. View Article : Google Scholar :

79 

Li M, Wang T, Tian H, Wei G, Zhao L and Shi Y: Macrophage-derived exosomes accelerate wound healing through their anti-inflammation effects in a diabetic rat model. Artif Cells Nanomed Biotechnol. 47:3793–3803. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Nirenjen S, Narayanan J, Tamilanban T, Subramaniyan V, Chitra V, Fuloria NK, Wong LS, Ramachawolran G, Sekar M, Gupta G, et al: Exploring the contribution of pro-inflammatory cytokines to impaired wound healing in diabetes. Front Immunol. 14:12163212023. View Article : Google Scholar : PubMed/NCBI

81 

Goren I, Müller E, Schiefelbein D, Christen U, Pfeilschifter J, Mühl H and Frank S: Systemic anti-TNFalpha treatment restores diabetes-impaired skin repair in ob/ob mice by inactivation of macrophages. J Invest Dermatol. 127:2259–2267. 2007. View Article : Google Scholar : PubMed/NCBI

82 

Mirza RE, Fang MM, Ennis WJ and Koh TJ: Blocking interleukin-1β induces a healing-associated wound macrophage phenotype and improves healing in type 2 diabetes. Diabetes. 62:2579–2587. 2013. View Article : Google Scholar : PubMed/NCBI

83 

Xu F, Zhang C and Graves DT: Abnormal cell responses and role of TNF-α in impaired diabetic wound healing. Biomed Res Int. 2013:7548022013. View Article : Google Scholar

84 

Dai J, Shen J, Chai Y and Chen H: IL-1β impaired diabetic wound healing by regulating MMP-2 and MMP-9 through the p38 pathway. Mediators Inflamm. 2021:66457662021. View Article : Google Scholar

85 

Lan CCE, Wu CS, Huang SM, Wu IH and Chen GS: High-glucose environment enhanced oxidative stress and increased interleukin-8 secretion from keratinocytes: New insights into impaired diabetic wound healing. Diabetes. 62:2530–2538. 2013. View Article : Google Scholar : PubMed/NCBI

86 

Wang G, Yang F, Zhou W, Xiao N, Luo M and Tang Z: The initiation of oxidative stress and therapeutic strategies in wound healing. Biomed Pharmacother. 157:1140042023. View Article : Google Scholar

87 

Xu Q, Huff LP, Fujii M and Griendling KK: Redox regulation of the actin cytoskeleton and its role in the vascular system. Free Radic Biol Med. 109:84–107. 2017. View Article : Google Scholar : PubMed/NCBI

88 

Long M, de la Vega MR, Wen Q, Wen Q, Bharara M, Jiang T, Zhang R, Zhou S, Wong PK, Wondrak GT, et al: An essential role of NRF2 in diabetic wound healing. Diabetes. 65:780–793. 2016. View Article : Google Scholar : PubMed/NCBI

89 

Zhou X, Ruan Q, Ye Z, Chu Z, Xi M, Li M, Hu W, Guo X, Yao P and Xie W: Resveratrol accelerates wound healing by attenuating oxidative stress-induced impairment of cell proliferation and migration. Burns. 47:133–139. 2021. View Article : Google Scholar

90 

Gao M, Nguyen TT, Suckow MA, Wolter WR, Gooyit M, Mobashery S and Chang M: Acceleration of diabetic wound healing using a novel protease-anti-protease combination therapy. Proc Natl Acad Sci USA. 112:15226–15231. 2015. View Article : Google Scholar : PubMed/NCBI

91 

Jones JI, Nguyen TT, Peng Z and Chang M: Targeting MMP-9 in diabetic foot ulcers. Pharmaceuticals (Basel). 12:792019. View Article : Google Scholar : PubMed/NCBI

92 

Yabluchanskiy A, Ma Y, Iyer RP, Hall ME and Lindsey ML: Matrix metalloproteinase-9: Many shades of function in cardiovascular disease. Physiology (Bethesda). 28:391–403. 2013.PubMed/NCBI

93 

Ambrozova N, Ulrichova J and Galandakova A: Models for the study of skin wound healing. The role of Nrf2 and NF-κB. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub. 161:1–13. 2017. View Article : Google Scholar : PubMed/NCBI

94 

Zubair M and Ahmad J: Role of growth factors and cytokines in diabetic foot ulcer healing: A detailed review. Rev Endocr Metab Disord. 20:207–217. 2019. View Article : Google Scholar : PubMed/NCBI

95 

Papanas N and Maltezos E: Growth factors in the treatment of diabetic foot ulcers: New technologies, any promises? Int J Low Extrem Wounds. 6:37–53. 2007. View Article : Google Scholar : PubMed/NCBI

96 

Ong HT and Dilley RJ: Novel non-angiogenic role for mesenchymal stem cell-derived vascular endothelial growth factor on keratinocytes during wound healing. Cytokine Growth Factor Rev. 44:69–79. 2018. View Article : Google Scholar : PubMed/NCBI

97 

Ou MY, Tan PC, Xie Y, Liu K, Gao YM, Yang XS, Zhou SB and Li QF: Dedifferentiated Schwann cell-derived TGF-β3 is essential for the neural system to promote wound healing. Theranostics. 12:5470–5487. 2022. View Article : Google Scholar :

98 

R AS, Nambi N, Radhakrishnan L, Prasad MK and Ramkumar KM: Neutrophil migration is a crucial factor in wound healing and the pathogenesis of diabetic foot ulcers: Insights into pharmacological interventions. Curr Vasc Pharmacol. 30: View Article : Google Scholar : 2024.

99 

Michopoulou A, Montmasson M, Garnier C, Lambert E, Dayan G and Rousselle P: A novel mechanism in wound healing: Laminin 332 drives MMP9/14 activity by recruiting syndecan-1 and CD44. Matrix Biol. 94:1–17. 2020. View Article : Google Scholar : PubMed/NCBI

100 

Fang WC and Lan CCE: The epidermal keratinocyte as a therapeutic target for management of diabetic wounds. Int J Mol Sci. 24:42902023. View Article : Google Scholar : PubMed/NCBI

101 

Pilcher BK, Dumin JA, Sudbeck BD, Krane SM, Welgus HG and Parks WC: The activity of collagenase-1 is required for keratinocyte migration on a type I collagen matrix. J Cell Biol. 137:1445–1457. 1997. View Article : Google Scholar : PubMed/NCBI

102 

Lan CCE, Wu CS, Kuo HY, Huang SM and Chen GS: Hyperglycaemic conditions hamper keratinocyte locomotion via sequential inhibition of distinct pathways: New insights on poor wound closure in patients with diabetes. Br J Dermatol. 160:1206–1214. 2009. View Article : Google Scholar : PubMed/NCBI

103 

Zhang C, Ponugoti B, Tian C, Xu F, Tarapore R, Batres A, Alsadun S, Lim J, Dong G and Graves DT: FOXO1 differentially regulates both normal and diabetic wound healing. J Cell Biol. 209:289–303. 2015. View Article : Google Scholar : PubMed/NCBI

104 

Wang Y and Graves DT: Keratinocyte function in normal and diabetic wounds and modulation by FOXO1. J Diabetes Res. 2020:37147042020. View Article : Google Scholar : PubMed/NCBI

105 

Kaussikaa S, Prasad MK and Ramkumar KM: Nrf2 activation in keratinocytes: A central role in diabetes-associated wound healing. Exp Dermatol. 33:e151892024. View Article : Google Scholar : PubMed/NCBI

106 

da Silva L, Carvalho E and Cruz MT: Role of neuropeptides in skin inflammation and its involvement in diabetic wound healing. Expert Opin Biol Ther. 10:1427–1439. 2010. View Article : Google Scholar : PubMed/NCBI

107 

Pradhan L, Nabzdyk C, Andersen ND, LoGerfo FW and Veves A: Inflammation and neuropeptides: The connection in diabetic wound healing. Expert Rev Mol Med. 11:e22009. View Article : Google Scholar : PubMed/NCBI

108 

Grande R, Puca V and Muraro R: Antibiotic resistance and bacterial biofilm. Expert Opin Ther Pat. 30:897–900. 2020. View Article : Google Scholar : PubMed/NCBI

109 

Marano RJ, Wallace HJ, Wijeratne D, Fear MW, Wong HS and O'Handley R: Secreted biofilm factors adversely affect cellular wound healing responses in vitro. Sci Rep. 5:132962015. View Article : Google Scholar

110 

Xing H, Huang Y, Kunkemoeller BH, Dahl PJ, Muraleetharan O, Malvankar NS, Murrell MP and Kyriakides TR: Dysregulation of TSP2-Rac1-WAVE2 axis in diabetic cells leads to cytoskeletal disorganization, increased cell stiffness, and dysfunction. Sci Rep. 12:224742022. View Article : Google Scholar : PubMed/NCBI

111 

Dupont S, Morsut L, Aragona M, Enzo E, Giulitti S, Cordenonsi M, Zanconato F, Le Digabel J, Forcato M, Bicciato S, et al: Role of YAP/TAZ in mechanotransduction. Nature. 474:179–183. 2011. View Article : Google Scholar : PubMed/NCBI

112 

Brusatin G, Panciera T, Gandin A, Citron A and Piccolo S: Biomaterials and engineered microenvironments to control YAP/TAZ-dependent cell behaviour. Nat Mater. 17:1063–1075. 2018. View Article : Google Scholar : PubMed/NCBI

113 

Lin MO, Sampath D, Bosykh DA, Wang C, Wang X, Subramaniam T, Han W, Hong W and Chakraborty S: YAP/TAZ drive agrin-matrix metalloproteinase 12-mediated diabetic skin wound healing. J Invest Dermatol. 145:155–170.e2. 2025. View Article : Google Scholar

114 

Majumdar R, Steen K, Coulombe PA and Parent CA: Non-canonical processes that shape the cell migration landscape. Curr Opin Cell Biol. 57:123–134. 2019. View Article : Google Scholar : PubMed/NCBI

115 

Xie L, Feng H, Li S, Meng G, Liu S, Tang X, Ma Y, Han Y, Xiao Y, Gu Y, et al: SIRT3 mediates the antioxidant effect of hydrogen sulfide in endothelial cells. Antioxid Redox Signal. 24:329–343. 2016. View Article : Google Scholar :

116 

Meng G, Liu J, Liu S, Song Q, Liu L, Xie L, Han Y and Ji Y: Hydrogen sulfide pretreatment improves mitochondrial function in myocardial hypertrophy via a SIRT3-dependent manner. Br J Pharmacol. 175:1126–1145. 2018. View Article : Google Scholar

117 

Yang S, Xu M, Meng G and Lu Y: SIRT3 deficiency delays diabetic skin wound healing via oxidative stress and necroptosis enhancement. J Cell Mol Med. 24:4415–4427. 2020. View Article : Google Scholar : PubMed/NCBI

118 

Johan MZ, Pyne NT, Kolesnikoff N, Poltavets V, Esmaeili Z, Woodcock JM, Lopez AF, Cowin AJ, Pitson SM and Samuel MS: Accelerated closure of diabetic wounds by efficient recruitment of fibroblasts upon inhibiting a 14-3-3/ROCK regulatory axis. J Invest Dermatol. 144:2562–2573.e4. 2024. View Article : Google Scholar : PubMed/NCBI

119 

Li Z, Zhang C, Wang L, Zhang Q, Dong Y, Sha X, Wang B, Zhu Z, Wang W, Wang Y, et al: Chitooligosaccharides promote diabetic wound healing by mediating fibroblast proliferation and migration. Sci Rep. 15:5562025. View Article : Google Scholar : PubMed/NCBI

120 

Gao Q, Pan L, Li Y and Zhang X: Astragaloside IV attenuates high glucose-induced human keratinocytes injury via TGF-β/Smad signaling pathway. J Tissue Viability. 31:678–686. 2022. View Article : Google Scholar : PubMed/NCBI

121 

Dong M, Ma X and Li F: Dedifferentiated fat cells-derived exosomes (DFATs-Exos) loaded in GelMA accelerated diabetic wound healing through Wnt/β-catenin pathway. Stem Cell Res Ther. 16:1032025. View Article : Google Scholar

122 

Ridley AJ: Rho GTPase signalling in cell migration. Curr Opin Cell Biol. 36:103–112. 2015. View Article : Google Scholar : PubMed/NCBI

123 

Wu M, Zhang S, Zhang W, Zhou Y, Guo Z, Fang Y, Yang Y, Shen Z, Lian D, Shen A and Peng J: Qingda granule ameliorates vascular remodeling and phenotypic transformation of adventitial fibroblasts via suppressing the TGF-β1/Smad2/3 pathway. J Ethnopharmacol. 313:1165352023. View Article : Google Scholar

124 

Fang H, Yang S, Luo Y, Zhang C, Rao Y, Liu R, Feng Y and Yu J: Notoginsenoside R1 inhibits vascular smooth muscle cell proliferation, migration and neointimal hyperplasia through PI3K/Akt signaling. Sci Rep. 8:75952018. View Article : Google Scholar : PubMed/NCBI

125 

Carvalho JR, Fortunato IC, Fonseca CG, Pezzarossa A, Barbacena P, Dominguez-Cejudo MA, Vasconcelos FF, Santos NC, Carvalho FA and Franco CA: Non-canonical Wnt signaling regulates junctional mechanocoupling during angiogenic collective cell migration. Elife. 8:e458532019. View Article : Google Scholar : PubMed/NCBI

126 

Van den Broeke C and Favoreel HW: Actin' up: Herpesvirus Interactions with Rho GTPase signaling. Viruses. 3:278–292. 2011. View Article : Google Scholar : PubMed/NCBI

127 

Qian W, Yamaguchi N, Lis P, Cammer M and Knaut H: Pulses of RhoA signaling stimulate actin polymerization and flow in protrusions to drive collective cell migration. Curr Biol. 34:245–259.e8. 2024. View Article : Google Scholar :

128 

Takaya K, Imbe Y, Wang Q, Okabe K, Sakai S, Aramaki-Hattori N and Kishi K: Rac1 inhibition regenerates wounds in mouse fetuses via altered actin dynamics. Sci Rep. 14:272132024. View Article : Google Scholar : PubMed/NCBI

129 

Shih PC, Tzeng IS, Chen YC and Chen ML: Gastrodin mitigates ketamine-induced inhibition of F-actin remodeling and cell migration by regulating the rho signaling pathway. Biomedicines. 13:6492025. View Article : Google Scholar : PubMed/NCBI

130 

Lawson CD and Ridley AJ: Rho GTPase signaling complexes in cell migration and invasion. J Cell Biol. 217:447–457. 2018. View Article : Google Scholar :

131 

Xue C, Li G, Lu J and Li L: Crosstalk between circRNAs and the PI3K/AKT signaling pathway in cancer progression. Signal Transduct Target Ther. 6:4002021. View Article : Google Scholar : PubMed/NCBI

132 

Teng Y, Fan Y, Ma J, Lu W, Liu N, Chen Y, Pan W and Tao X: The PI3K/Akt pathway: Emerging roles in skin homeostasis and a group of non-malignant skin disorders. Cells. 10:12192021. View Article : Google Scholar : PubMed/NCBI

133 

Zhang X, Shi L, Xing M, Li C, Ma F and Ma Y and Ma Y: Interplay between lncRNAs and the PI3K/AKT signaling pathway in the progression of digestive system neoplasms (Review). Int J Mol Med. 55:152024. View Article : Google Scholar : PubMed/NCBI

134 

Karar J and Maity A: PI3K/AKT/mTOR pathway in angiogenesis. Front Mol Neurosci. 4:512011. View Article : Google Scholar : PubMed/NCBI

135 

Wu X, Sun Q, He S, Wu Y, Du S, Gong L, Yu J and Guo H: Ropivacaine inhibits wound healing by suppressing the proliferation and migration of keratinocytes via the PI3K/AKT/mTOR pathway. BMC Anesthesiol. 22:1062022. View Article : Google Scholar : PubMed/NCBI

136 

Deng S, Leong HC, Datta A, Gopal V, Kumar AP and Yap CT: PI3K/AKT signaling tips the balance of cytoskeletal forces for cancer progression. Cancers (Basel). 14:16522022. View Article : Google Scholar : PubMed/NCBI

137 

Hsu JW, Bai M, Li K, Yang JS, Chu N, Cole PA, Eck MJ, Li J and Hsu VW: The protein kinase Akt acts as a coat adaptor in endocytic recycling. Nat Cell Biol. 22:927–933. 2020. View Article : Google Scholar : PubMed/NCBI

138 

Hannigan M, Zhan L, Li Z, Ai Y, Wu D and Huang CK: Neutrophils lacking phosphoinositide 3-kinase gamma show loss of directionality during N-formyl-Met-Leu-Phe-induced chemotaxis. Proc Natl Acad Sci USA. 99:3603–3608. 2002. View Article : Google Scholar : PubMed/NCBI

139 

Najafi M, Farhood B and Mortezaee K: Extracellular matrix (ECM) stiffness and degradation as cancer drivers. J Cell Biochem. 120:2782–2790. 2019. View Article : Google Scholar

140 

Nakerakanti S and Trojanowska M: The role of TGF-β receptors in fibrosis. Open Rheumatol J. 6:156–162. 2012. View Article : Google Scholar :

141 

Tu S, Huang W, Huang C, Luo Z and Yan X: Contextual regulation of TGF-β signaling in liver cancer. Cells. 8:12352019. View Article : Google Scholar

142 

Zhang K, Zhang H, Xiang H, Liu J, Liu Y, Zhang X, Wang J and Tang Y: TGF-β1 induces the dissolution of tight junctions in human renal proximal tubular cells: role of the RhoA/ROCK signaling pathway. Int J Mol Med. 32:464–468. 2013. View Article : Google Scholar : PubMed/NCBI

143 

Gailit J, Welch MP and Clark RA: TGF-beta 1 stimulates expression of keratinocyte integrins during re-epithelialization of cutaneous wounds. J Invest Dermatol. 103:221–227. 1994. View Article : Google Scholar : PubMed/NCBI

144 

Ponugoti B, Xu F, Zhang C, Tian C, Pacios S and Graves DT: FOXO1 promotes wound healing through the up-regulation of TGF-β1 and prevention of oxidative stress. J Cell Biol. 203:327–343. 2013. View Article : Google Scholar : PubMed/NCBI

145 

Weber CE, Li NY, Wai PY and Kuo PC: Epithelial-Mesenchymal transition, TGF-β, and osteopontin in wound healing and tissue remodeling after injury. J Burn Care Res. 33:311–318. 2012. View Article : Google Scholar : PubMed/NCBI

146 

Song P, Gao Z, Bao Y, Chen L, Huang Y, Liu Y, Dong Q and Wei X: Wnt/β-catenin signaling pathway in carcinogenesis and cancer therapy. J Hematol Oncol. 17:462024. View Article : Google Scholar

147 

Moon RT, Kohn AD, De Ferrari GV and Kaykas A: WNT and beta-catenin signalling: Diseases and therapies. Nat Rev Genet. 5:691–701. 2004. View Article : Google Scholar : PubMed/NCBI

148 

Nusse R and Clevers H: Wnt/β-Catenin signaling, disease, and emerging therapeutic modalities. Cell. 169:985–999. 2017. View Article : Google Scholar : PubMed/NCBI

149 

Schaale K, Neumann J, Schneider D, Ehlers S and Reiling N: Wnt signaling in macrophages: Augmenting and inhibiting mycobacteria-induced inflammatory responses. Eur J Cell Biol. 90:553–559. 2011. View Article : Google Scholar

150 

Sedgwick AE and D'Souza-Schorey C: Wnt signaling in cell motility and invasion: Drawing parallels between development and cancer. Cancers (Basel). 8:802016. View Article : Google Scholar : PubMed/NCBI

151 

Wu B, Crampton SP and Hughes CCW: Wnt signaling induces MMP expression and regulates T cell transmigration. Immunity. 26:227–239. 2007. View Article : Google Scholar : PubMed/NCBI

152 

Xue W, Yang L, Chen C, Ashrafizadeh M, Tian Y and Sun R: Wnt/β-catenin-driven EMT regulation in human cancers. Cell Mol Life Sci. 81:792024. View Article : Google Scholar

153 

Ono M, Masaki A, Maeda A, Kilts TM, Hara ES, Komori T, Pham H, Kuboki T and Young MF: CCN4/WISP1 controls cutaneous wound healing by modulating proliferation, migration and ECM expression in dermal fibroblasts via α5β1 and TNFα. Matrix Biol. 68-69:533–546. 2018. View Article : Google Scholar : PubMed/NCBI

154 

Repertinger SK, Campagnaro E, Fuhrman J, El-Abaseri T, Yuspa SH and Hansen LA: EGFR enhances early healing after cutaneous incisional wounding. J Invest Dermatol. 123:982–989. 2004. View Article : Google Scholar : PubMed/NCBI

155 

Wang Y, Wu Z, Tian J, Mi Y, Ren X, Kang J, Zhang W, Zhou X, Wang G and Li R: Intermedin protects HUVECs from ischemia reperfusion injury via Wnt/β-catenin signaling pathway. Ren Fail. 41:159–166. 2019. View Article : Google Scholar : PubMed/NCBI

156 

Jun EK, Zhang Q, Yoon BS, Moon JH, Lee G, Park G, Kang PJ, Lee JH, Kim A and You S: 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. 15:605–628. 2014. View Article : Google Scholar : PubMed/NCBI

157 

Toraldo G, Bhasin S, Bakhit M, Guo W, Serra C, Safer JD, Bhawan J and Jasuja R: Topical androgen antagonism promotes cutaneous wound healing without systemic androgen deprivation by blocking β-catenin nuclear translocation and cross-talk with TGF-β signaling in keratinocytes. Wound Repair Regen. 20:61–73. 2012. View Article : Google Scholar : PubMed/NCBI

158 

Wang H, Wang X, Liu X, Zhou J, Yang Q, Chai B, Chai Y, Ma Z and Lu S: miR-199a-5p plays a pivotal role on wound healing via suppressing VEGFA and ROCK1 in diabetic ulcer foot. Oxid Med Cell Longev. 2022:47910592022. View Article : Google Scholar : PubMed/NCBI

159 

Yu J, Nam D and Park KS: Substance P enhances cellular migration and inhibits senescence in human dermal fibroblasts under hyperglycemic conditions. Biochem Biophys Res Commun. 522:917–923. 2020. View Article : Google Scholar

160 

Huang J, Deng Q, Tsang LL, Chang G, Guo J, Ruan YC, Wang CC, Li G, Chan HF, Zhang X and Jiang X: Mesenchymal stem cells from perinatal tissues promote diabetic wound healing via PI3K/AKT activation. Stem Cell Res Ther. 16:592025. View Article : Google Scholar : PubMed/NCBI

161 

Xu S, Jiang C, Yu T and Chen K: A multi-purpose dressing based on resveratrol-loaded ionic liquids/gelatin methacryloyl hydrogel for enhancing diabetic wound healing. Int J Biol Macromol. 283:1367732024. View Article : Google Scholar : PubMed/NCBI

162 

Peng Y, Wu S, Tang Q, Li S and Peng C: KGF-1 accelerates wound contraction through the TGF-β1/Smad signaling pathway in a double-paracrine manner. J Biol Chem. 294:8361–8370. 2019. View Article : Google Scholar : PubMed/NCBI

163 

Oyebode OA and Houreld NN: Photobiomodulation at 830 nm stimulates migration, survival and proliferation of fibroblast cells. Diabetes Metab Syndr Obes. 15:2885–2900. 2022. View Article : Google Scholar : PubMed/NCBI

164 

Wang L, Chen J, Song J, Xiang Y, Yang M, Xia L, Yang J, Hou X, Chen L and Wang L: Activation of the Wnt/β-catenin signalling pathway enhances exosome production by hucMSCs and improves their capability to promote diabetic wound healing. J Nanobiotechnology. 22:3732024. View Article : Google Scholar

165 

Liu Z, Yang S, Li X, Wang S, Zhang T, Huo N, Duan R, Shi Q, Zhang J and Xu J: Local transplantation of GMSC-derived exosomes to promote vascularized diabetic wound healing by regulating the Wnt/β-catenin pathways. Nanoscale Adv. 5:916–926. 2023. View Article : Google Scholar : PubMed/NCBI

166 

Lv Q, Deng J, Chen Y, Wang Y, Liu B and Liu J: Engineered human adipose stem-cell-derived exosomes loaded with miR-21-5p to promote diabetic cutaneous wound healing. Mol Pharm. 17:1723–1733. 2020. View Article : Google Scholar : PubMed/NCBI

167 

Yang Y, GuangXuan H, GenMeng W, MengHuan L, Bo C and XueJie Y: Idiopathic inflammatory myopathy and non-coding RNA. Front Immunol. 14:12279452023. View Article : Google Scholar : PubMed/NCBI

168 

Ballarino M, Morlando M, Fatica A and Bozzoni I: Non-coding RNAs in muscle differentiation and musculoskeletal disease. J Clin Invest. 126:2021–2030. 2016. View Article : Google Scholar : PubMed/NCBI

169 

Chen L, Shen S and Wang S: LncRNA SNHG16 knockdown promotes diabetic foot ulcer wound healing via sponging MiR-31-5p. Tohoku J Exp Med. 261:283–289. 2023. View Article : Google Scholar : PubMed/NCBI

170 

Peng J, Zhu H, Ruan B, Duan Z and Cao M: miR-155 promotes m6A modification of SOX2 mRNA through targeted regulation of HIF-1α and delays wound healing in diabetic foot ulcer in vitro models. J Diabetes Investig. 16:60–71. 2024. View Article : Google Scholar

171 

Tian M, Tang J, Huang R, Dong J and Jia H: Circ_072697 knockdown promotes advanced glycation end products-induced cell proliferation and migration in HaCaT cells via miR-3150a-3p/KDM2A axis. BMC Endocr Disord. 23:2002023. View Article : Google Scholar : PubMed/NCBI

172 

Wang F, Mei X, Yang Y, Zhang H, Li Z, Zhu L, Deng S and Wang Y: Non-coding RNA and its network in the pathogenesis of myasthenia gravis. Front Mol Biosci. 11:13884762024. View Article : Google Scholar : PubMed/NCBI

173 

Bhan A and Mandal SS: Long noncoding RNAs: Emerging stars in gene regulation, epigenetics and human disease. ChemMedChem. 9:1932–1956. 2014. View Article : Google Scholar : PubMed/NCBI

174 

Hong W, Xiong Z, Wang X, Liao X, Liu M, Jiang Z, Min D, Li J, Guo G and Fu Z: Long noncoding RNA XIST promotes cell proliferation and migration in diabetic foot ulcers through the miR-126-3p/EGFR axis. Diabetol Metab Syndr. 16:352024. View Article : Google Scholar : PubMed/NCBI

175 

He M, Tu L, Shu R, Meng Q, Du S, Xu Z and Wang S: Long noncoding RNA CASC2 facilitated wound healing through miRNA-155/HIF-1α in diabetic foot ulcers. Contrast Media Mol Imaging. 2022:62914972022. View Article : Google Scholar

176 

Li B, Zhou Y, Chen J, Wang T, Li Z, Fu Y, Zhai A and Bi C: Long noncoding RNA H19 acts as a miR-29b sponge to promote wound healing in diabetic foot ulcer. FASEB J. 35:e205262021.

177 

Hu M, Wu Y, Yang C, Wang X, Wang W, Zhou L, Zeng T, Zhou J, Wang C, Lao G, et al: Novel long noncoding RNA lnc-URIDS delays diabetic wound healing by targeting Plod1. Diabetes. 69:2144–2156. 2020. View Article : Google Scholar : PubMed/NCBI

178 

Gezer U, Özgür E, Cetinkaya M, Isin M and Dalay N: Long non-coding RNAs with low expression levels in cells are enriched in secreted exosomes. Cell Biol Int. 38:1076–1079. 2014. View Article : Google Scholar : PubMed/NCBI

179 

Li B, Luan S, Chen J, Zhou Y, Wang T, Li Z, Fu Y, Zhai A and Bi C: The MSC-derived exosomal lncRNA H19 promotes wound healing in diabetic foot ulcers by upregulating PTEN via MicroRNA-152-3p. Mol Ther Nucleic Acids. 19:814–826. 2020. View Article : Google Scholar : PubMed/NCBI

180 

Han ZF, Cao JH, Liu ZY, Yang Z, Qi RX and Xu HL: Exosomal lncRNA KLF3-AS1 derived from bone marrow mesenchymal stem cells stimulates angiogenesis to promote diabetic cutaneous wound healing. Diabetes Res Clin Pract. 183:1091262022. View Article : Google Scholar

181 

Fu W, Liang D, Wu X, Chen H, Hong X, Wang J, Zhu T, Zeng T, Lin W, Chen S, et al: Long noncoding RNA LINC01435 impedes diabetic wound healing by facilitating YY1-mediated HDAC8 expression. iScience. 25:1040062022. View Article : Google Scholar : PubMed/NCBI

182 

Roso-Mares A, Andújar I, Corpas TD and Sun BK: Non-coding RNAs as skin disease biomarkers, molecular signatures, and therapeutic targets. Hum Genet. 143:801–812. 2024. View Article : Google Scholar

183 

Shirley SN, Watson AE and Yusuf N: Pathogenesis of inflammation in skin disease: From molecular mechanisms to pathology. Int J Mol Sci. 25:101522024. View Article : Google Scholar : PubMed/NCBI

184 

Tsai HC, Chang GRL, Tung MC, Tu MY, Chen IC, Liu YH, Cidem A and Chen CM: MicroRNA signature in an in vitro keratinocyte model of diabetic wound healing. Int J Mol Sci. 25:101252024. View Article : Google Scholar : PubMed/NCBI

185 

Zhao X, Xu M, Tang Y, Xie D, Deng L, Chen M and Wang Y: Decreased expression of miR-204-3p in peripheral blood and wound margin tissue associated with the onset and poor wound healing of diabetic foot ulcers. Int Wound J. 20:413–429. 2023. View Article : Google Scholar

186 

Zhang HC, Wen T and Cai YZ: Overexpression of miR-146a promotes cell proliferation and migration in a model of diabetic foot ulcers by regulating the AKAP12 axis. Endocr J. 69:85–94. 2022. View Article : Google Scholar

187 

Wang W, Yang C, Wang XY, Zhou LY, Lao GJ, Liu D, Wang C, Hu MD, Zeng TT, Yan L and Ren M: MicroRNA-129 and -335 promote diabetic wound healing by inhibiting Sp1-mediated MMP-9 expression. Diabetes. 67:1627–1638. 2018. View Article : Google Scholar : PubMed/NCBI

188 

Li P, Hong G, Zhan W, Deng M, Tu C, Wei J and Lin H: Endothelial progenitor cell derived exosomes mediated miR-182-5p delivery accelerate diabetic wound healing via down-regulating PPARG. Int J Med Sci. 20:468–481. 2023. View Article : Google Scholar : PubMed/NCBI

189 

Wang KX, Zhao LL, Zheng LT, Meng LB, Jin L, Zhang LJ, Kong FL and Liang F: Accelerated wound healing in diabetic rat by miRNA-185-5p and its anti-inflammatory activity. Diabetes Metab Syndr Obes. 16:1657–1667. 2023. View Article : Google Scholar : PubMed/NCBI

190 

Wu M, Tu J, Huang J, Wen H, Zeng Y and Lu Y: Exosomal IRF1-loaded rat adipose-derived stem cell sheet contributes to wound healing in the diabetic foot ulcers. Mol Med. 29:602023. View Article : Google Scholar : PubMed/NCBI

191 

Wu Y, Zhang K, Liu R, Zhang H, Chen D, Yu S, Chen W, Wan S, Zhang Y, Jia Z, et al: MicroRNA-21-3p accelerates diabetic wound healing in mice by downregulating SPRY1. Aging (Albany NY). 12:15436–15445. 2020. View Article : Google Scholar : PubMed/NCBI

192 

Wang C, Huang L, Li J, Liu D and Wu B: MicroRNA miR-145-5p inhibits cutaneous wound healing by targeting PDGFD in diabetic foot ulcer. Biochem Genet. 62:2437–2454. 2024. View Article : Google Scholar

193 

Zhao X, Xu M, Tang Y, Xie D, Wang Y and Chen M: Changes in miroRNA-103 expression in wound margin tissue are related to wound healing of diabetes foot ulcers. Int Wound J. 20:467–483. 2022. View Article : Google Scholar : PubMed/NCBI

194 

Zhang P, Song X, Dong Q, Zhou L and Wang L: miR-27-3p inhibition restore fibroblasts viability in diabetic wound by targeting NOVA1. Aging (Albany NY). 12:12841–12849. 2020. View Article : Google Scholar : PubMed/NCBI

195 

Zheng L, Song H, Li Y, Li H, Lin G and Cai Z: Insulin-Induced gene 1-enhance secretion of BMSC exosome enriched in miR-132-3p promoting wound healing in diabetic mice. Mol Pharm. 21:4372–4385. 2024. View Article : Google Scholar : PubMed/NCBI

196 

Zuo C, Fan P, Yang Y and Hu C: MiR-488-3p facilitates wound healing through CYP1B1-mediated Wnt/β-catenin signaling pathway by targeting MeCP2. J Diabetes Investig. 15:145–158. 2024. View Article : Google Scholar

197 

Huang H, Zhu W, Huang Z, Zhao D, Cao L and Gao X: Adipose-derived stem cell exosome NFIC improves diabetic foot ulcers by regulating miR-204-3p/HIPK2. J Orthop Surg Res. 18:6872023. View Article : Google Scholar : PubMed/NCBI

198 

Qiu ZY, Xu WC and Liang ZH: Bone marrow mesenchymal stem cell-derived exosomal miR-221-3p promotes angiogenesis and wound healing in diabetes via the downregulation of forkhead box P1. Diabet Med. 41:e153862024. View Article : Google Scholar : PubMed/NCBI

199 

Zhou X, Ye C, Jiang L, Zhu X, Zhou F, Xia M and Chen Y: The bone mesenchymal stem cell-derived exosomal miR-146a-5p promotes diabetic wound healing in mice via macrophage M1/M2 polarization. Mol Cell Endocrinol. 579:1120892024. View Article : Google Scholar

200 

Guo E, Wang L, Wu J and Chen Q: Exosomes from MicroRNA-125b-modified adipose-derived stem cells promote wound healing of diabetic foot ulcers. Curr Stem Cell Res Ther. 20:409–420. 2024. View Article : Google Scholar : PubMed/NCBI

201 

Che D, Xiang X, Xie J, Chen Z, Bao Q and Cao D: exosomes derived from adipose stem cells enhance angiogenesis in diabetic wound via miR-146a-5p/JAZF1 axis. Stem Cell Rev Rep. 20:1026–1039. 2024. View Article : Google Scholar : PubMed/NCBI

202 

Ge L, Wang K, Lin H, Tao E, Xia W, Wang F, Mao C and Feng Y: Engineered exosomes derived from miR-132-overexpresssing adipose stem cells promoted diabetic wound healing and skin reconstruction. Front Bioeng Biotechnol. 11:11295382023. View Article : Google Scholar : PubMed/NCBI

203 

Sedykh S, Kuleshova A and Nevinsky G: Milk exosomes: Perspective agents for anticancer drug delivery. Int J Mol Sci. 21:66462020. View Article : Google Scholar : PubMed/NCBI

204 

Munagala R, Aqil F, Jeyabalan J and Gupta RC: Bovine milk-derived exosomes for drug delivery. Cancer Lett. 371:48–61. 2016. View Article : Google Scholar :

205 

Yan C, Chen J, Wang C, Yuan M, Kang Y, Wu Z, Li W, Zhang G, Machens HG and Rinkevich Y: Milk exosomes-mediated miR-31-5p delivery accelerates diabetic wound healing through promoting angiogenesis. Drug Deliv. 29:214–228. 2022. View Article : Google Scholar : PubMed/NCBI

206 

Liu R, Zhang L, Zhao X, Liu J, Chang W, Zhou L and Zhang K: circRNA: Regulatory factors and potential therapeutic targets in inflammatory dermatoses. J Cell Mol Med. 26:4389–4400. 2022. View Article : Google Scholar : PubMed/NCBI

207 

Beňačka R, Szabóová D, Guľašová Z and Hertelyová Z: Non-Coding RNAs in breast cancer: Diagnostic and therapeutic implications. Int J Mol Sci. 26:1272024. View Article : Google Scholar

208 

Zhang Y, Wu Y, Liu Z, Yang K, Lin H and Xiong K: Non-coding RNAs as potential targets in metformin therapy for cancer. Cancer Cell Int. 24:3332024. View Article : Google Scholar : PubMed/NCBI

209 

Fu Z, Jiang Z, Liao X, Liu M, Guo G, Wang X, Yang G, Zhou Z, Hu L and Xiong Z: Upregulation of circ_0080968 in diabetic foot ulcer inhibits wound healing via repressing the migration and promoting proliferation of keratinocytes. Gene. 883:1476692023. View Article : Google Scholar : PubMed/NCBI

210 

Han D, Liu W, Li G and Liu L: Circ_PRKDC knockdown promotes skin wound healing by enhancing keratinocyte migration via miR-31/FBN1 axis. J Mol Histol. 52:681–691. 2021. View Article : Google Scholar : PubMed/NCBI

211 

Wang A, Toma MA, Ma J, Li D, Vij M, Chu T, Wang J, Li X and Landén NX: Circular RNA hsa_circ_0084443 is upregulated in diabetic foot ulcer and modulates keratinocyte migration and proliferation. Adv Wound Care (New Rochelle). 9:145–160. 2020. View Article : Google Scholar : PubMed/NCBI

212 

Liang ZH, Pan NF, Lin SS, Qiu ZY, Liang P, Wang J, Zhang Z and Pan YC: Exosomes from mmu_circ_0001052-modified adipose-derived stem cells promote angiogenesis of DFU via miR-106a-5p and FGF4/p38MAPK pathway. Stem Cell Res Ther. 13:3362022. View Article : Google Scholar : PubMed/NCBI

213 

Huang Q, Chu Z, Wang Z, Li Q, Meng S, Lu Y, Ma K, Cui S, Hu W, Zhang W, et al: circCDK13-loaded small extracellular vesicles accelerate healing in preclinical diabetic wound models. Nat Commun. 15:39042024. View Article : Google Scholar : PubMed/NCBI

214 

Chen M, Przyborowski M and Berthiaume F: Stem cells for skin tissue engineering and wound healing. Crit Rev Biomed Eng. 37:399–421. 2009. View Article : Google Scholar : PubMed/NCBI

215 

Xu ZH, Ma MH, Li YQ, Li LL and Liu GH: Progress and expectation of stem cell therapy for diabetic wound healing. World J Clin Cases. 11:506–513. 2023. View Article : Google Scholar : PubMed/NCBI

216 

Deng Q, Pan S, Du F, Sang H, Cai Z, Xu X, Wei Q, Yu S, Zhang J and Li C: The Effect of conditioned medium from angiopoietin-1 gene-modified mesenchymal stem cells on wound healing in a diabetic mouse model. Bioengineering (Basel). 11:12442024. View Article : Google Scholar

217 

Liu Z, Wen X, Wang H, Zhou J, Zhao M, Lin Q, Wang Y, Li J, Li D, Du Z, et al: Molecular imaging of induced pluripotent stem cell immunogenicity with in vivo development in ischemic myocardium. PLoS One. 8:e663692013. View Article : Google Scholar : PubMed/NCBI

218 

He L, Cai Y, Du H, Shu M and Zhu C: Adipose stem cell-derived exosomes promote high glucose-induced wound healing by regulating the TRIM32/STING axis. Arch Dermatol Res. 316:3232024. View Article : Google Scholar

219 

Ding JY, Chen MJ, Wu LF, Shu GF, Fang SJ, Li ZY, Chu XR, Li XK, Wang ZG and Ji JS: Mesenchymal stem cell-derived extracellular vesicles in skin wound healing: Roles, opportunities and challenges. Mil Med Res. 10:362023.PubMed/NCBI

220 

Li Y, Zhu Z, Li S, Xie X, Qin L, Zhang Q, Yang Y, Wang T and Zhang Y: Exosomes: Compositions, biogenesis, and mechanisms in diabetic wound healing. J Nanobiotechnology. 22:3982024. View Article : Google Scholar : PubMed/NCBI

221 

Laiva AL, O'Brien FJ and Keogh MB: Innovations in gene and growth factor delivery systems for diabetic wound healing. J Tissue Eng Regen Med. 12:e296–e312. 2018. View Article : Google Scholar

222 

Li Z, Qiu X, Guan G, Shi K, Chen S, Tang J, Xiao M, Tang S, Yan Y, Zhou J and Xie H: The role of FGF-21 in promoting diabetic wound healing by modulating high glucose-induced inflammation. Heliyon. 10:e300222024. View Article : Google Scholar : PubMed/NCBI

223 

Tang L, Cai S, Lu X, Wu D, Zhang Y, Li X, Qin X, Guo J, Zhang X and Liu C: Platelet-Derived growth factor nanocapsules with tunable controlled release for chronic wound healing. Small. 20:e23107432024. View Article : Google Scholar : PubMed/NCBI

224 

Jeong S, Kim B, Park M, Ban E, Lee SH and Kim A: Improved diabetic wound healing by EGF encapsulation in gelatin-alginate coacervates. Pharmaceutics. 12:3342020. View Article : Google Scholar : PubMed/NCBI

225 

Wang H, Xu Z, Zhao M, Liu G and Wu J: Advances of hydrogel dressings in diabetic wounds. Biomater Sci. 9:1530–1546. 2021. View Article : Google Scholar : PubMed/NCBI

226 

Serpico L, Iacono SD, Cammarano A and De Stefano L: Recent advances in stimuli-responsive hydrogel-based wound dressing. Gels. 9:4512023. View Article : Google Scholar : PubMed/NCBI

227 

Liang Y, He J and Guo B: Functional hydrogels as wound dressing to enhance wound healing. ACS Nano. 15:12687–12722. 2021. View Article : Google Scholar : PubMed/NCBI

228 

Miguel SP, Ribeiro MP, Brancal H, Coutinho P and Correia IJ: Thermoresponsive chitosan-agarose hydrogel for skin regeneration. Carbohydr Polym. 111:366–373. 2014. View Article : Google Scholar : PubMed/NCBI

229 

Bei Z, Zhang L, Li J, Tong Q, Shi K, Chen W, Yu Y, Sun A, Xu Y, Liu J and Qian Z: A smart stimulation-deadhesion and antimicrobial hydrogel for repairing diabetic wounds infected with methicillin-resistant staphylococcus aureus. Adv Healthc Mater. 13:e23030422024. View Article : Google Scholar

230 

Li Y, Fu R, Duan Z, Zhu C and Fan D: Artificial nonenzymatic antioxidant MXene Nanosheet-anchored injectable hydrogel as a mild photothermal-controlled oxygen release platform for diabetic wound healing. ACS Nano. 16:7486–7502. 2022. View Article : Google Scholar : PubMed/NCBI

231 

Şeker Ş, Elçin AE and Elçin YM: Current trends in the design and fabrication of PRP-based scaffolds for tissue engineering and regenerative medicine. Biomed Mater. 20:202025. View Article : Google Scholar

232 

Xu K, Deng S, Zhu Y, Yang W, Chen W, Huang L, Zhang C, Li M, Ao L, Jiang Y, et al: Platelet rich plasma loaded multifunctional hydrogel accelerates diabetic wound healing via regulating the continuously abnormal microenvironments. Adv Healthc Mater. 12:e23013702023. View Article : Google Scholar : PubMed/NCBI

233 

Wei Q, Su J, Meng S, Wang Y, Ma K, Li B, Chu Z, Huang Q, Hu W, Wang Z, et al: MiR-17-5p-engineered sEVs encapsulated in GelMA hydrogel facilitated diabetic wound healing by targeting PTEN and p21. Adv Sci (Weinh). 11:e23077612024. View Article : Google Scholar : PubMed/NCBI

234 

Alberts A, Bratu AG, Niculescu AG and Grumezescu AM: New perspectives of hydrogels in chronic wound management. Molecules. 30:6862025. View Article : Google Scholar : PubMed/NCBI

235 

Negut I and Bita B: Exploring the potential of artificial intelligence for hydrogel development-a short review. Gels. 9:8452023. View Article : Google Scholar : PubMed/NCBI

236 

Tan CT, Liang K, Ngo ZH, Dube CT and Lim CY: Application of 3D bioprinting technologies to the management and treatment of diabetic foot ulcers. Biomedicines. 8:4412020. View Article : Google Scholar : PubMed/NCBI

237 

Wang M, Song Y, Bisoyi HK, Yang JF, Liu L, Yang H and Li Q: A liquid crystal elastomer-based unprecedented two-way shape-memory aerogel. Adv Sci (Weinh). 8:e21026742021. View Article : Google Scholar : PubMed/NCBI

238 

Jeong Y, Patel R and Patel M: Biopolymer-Based biomimetic aerogel for biomedical applications. Biomimetics (Basel). 9:3972024. View Article : Google Scholar : PubMed/NCBI

239 

Guo N, Xia Y, Zeng W, Chen J, Wu Q, Shi Y, Li G, Huang Z, Wang G and Liu Y: Alginate-based aerogels as wound dressings for efficient bacterial capture and enhanced antibacterial photodynamic therapy. Drug Deliv. 29:1086–1099. 2022. View Article : Google Scholar : PubMed/NCBI

240 

Wang X, Yuan Z, Shafiq M, Cai G, Lei Z, Lu Y, Guan X, Hashim R, El-Newehy M, Abdulhameed MM, et al: Composite aerogel scaffolds containing flexible silica nanofiber and tricalcium phosphate enable skin regeneration. ACS Appl Mater Interfaces. 16:25843–25855. 2024. View Article : Google Scholar : PubMed/NCBI

241 

Vaseghi A, Sadeghizadeh M, Herb M, Grumme D, Demidov Y, Remmler T and Maleki HH: 3D printing of biocompatible and antibacterial silica-silk-chitosan-based hybrid aerogel scaffolds loaded with propolis. ACS Appl Bio Mater. 7:7917–7935. 2024. View Article : Google Scholar : PubMed/NCBI

242 

Wu B, Pan W, Luo S, Luo X, Zhao Y, Xiu Q, Zhong M, Wang Z, Liao T, Li N, et al: Turmeric-Derived nanoparticles functionalized aerogel regulates multicellular networks to promote diabetic wound healing. Adv Sci (Weinh). 11:e23076302024. View Article : Google Scholar : PubMed/NCBI

243 

Ramos R, Silva JP, Rodrigues AC, Costa R, Guardão L, Schmitt F, Soares R, Vilanova M, Domingues L and Gama M: Wound healing activity of the human antimicrobial peptide LL37. Peptides. 32:1469–1476. 2011. View Article : Google Scholar : PubMed/NCBI

244 

Grönberg A, Mahlapuu M, Ståhle M, Whately-Smith C and Rollman O: Treatment with LL-37 is safe and effective in enhancing healing of hard-to-heal venous leg ulcers: A randomized, placebo-controlled clinical trial. Wound Repair Regen. 22:613–621. 2014. View Article : Google Scholar : PubMed/NCBI

245 

John JV, Sharma NS, Tang G, Luo Z, Su Y, Weihs S, Shahriar SMS, Wang G, McCarthy A, Dyke J, et al: Nanofiber aerogels with precision macrochannels and LL-37-mimic peptides synergistically promote diabetic wound healing. Adv Funct Mater. 33:22069362023. View Article : Google Scholar : PubMed/NCBI

246 

Zhao C, Wu Z, Pan B, Zhang R, Golestani A, Feng Z, Ge Y and Yang H: Functional biomacromolecules-based microneedle patch for the treatment of diabetic wound. Int J Biol Macromol. 267:1316502024. View Article : Google Scholar : PubMed/NCBI

247 

Hu F, Gao Q, Liu J, Chen W, Zheng C, Bai Q, Sun N, Zhang W, Zhang Y and Lu T: Smart microneedle patches for wound healing and management. J Mater Chem B. 11:2830–2851. 2023. View Article : Google Scholar : PubMed/NCBI

248 

Yin M, Wu J, Deng M, Wang P, Ji G, Wang M, Zhou C, Blum NT, Zhang W, Shi H, et al: Multifunctional magnesium organic framework-based microneedle patch for accelerating diabetic wound healing. ACS Nano. 15:17842–17853. 2021. View Article : Google Scholar : PubMed/NCBI

249 

Wang P, Wu J, Yang H, Liu H, Yao T, Liu C, Gong Y, Wang M, Ji G, Huang P and Wang X: Intelligent microneedle patch with prolonged local release of hydrogen and magnesium ions for diabetic wound healing. Bioact Mater. 24:463–476. 2023.PubMed/NCBI

250 

Avcil M and Çelik A: Microneedles in drug delivery: Progress and challenges. Micromachines (Basel). 12:13212021. View Article : Google Scholar : PubMed/NCBI

251 

Alsarayreh AZ, Oran SA, Shakhanbeh JM, Khleifat KM, Al Qaisi YT, Alfarrayeh II and Alkaramseh AM: Efficacy of methanolic extracts of some medicinal plants on wound healing in diabetic rats. Heliyon. 8:e100712022. View Article : Google Scholar : PubMed/NCBI

252 

Wu JR, Lu YC, Hung SJ, Lin JH, Chang KC, Chen JK, Tsai WT, Ho TJ and Chen HP: Antimicrobial and immunomodulatory activity of herb extracts used in burn wound healing: 'San Huang Powder'. Evid Based Complement Alternat Med. 2021:29000602021. View Article : Google Scholar

253 

Soheilifar MH, Dastan D, Masoudi-Khoram N, Neghab HN, Nobari S, Tabaie SM and Amini R: In vitro and in vivo evaluation of the diabetic wound healing properties of Saffron (Crocus Sativus L.) petals. Sci Rep. 14:193732024. View Article : Google Scholar : PubMed/NCBI

254 

Huang L, Cai HA, Zhang MS, Liao RY, Huang X and Hu FD: Ginsenoside Rg1 promoted the wound healing in diabetic foot ulcers via miR-489-3p/Sirt1 axis. J Pharmacol Sci. 147:271–283. 2021. View Article : Google Scholar : PubMed/NCBI

255 

Sun X, Wang X, Zhao Z, Chen J, Li C and Zhao G: Paeoniflorin accelerates foot wound healing in diabetic rats though activating the Nrf2 pathway. Acta Histochem. 122:1516492020. View Article : Google Scholar : PubMed/NCBI

256 

Zhang S, Xu Y, Junior CZ, Chen X and Zhu J: Dang-Gui-Si-Ni decoction facilitates wound healing in diabetic foot ulcers by regulating expression of AGEs/RAGE/TGF-β/Smad2/3. Arch Dermatol Res. 316:3382024. View Article : Google Scholar

257 

Gong Y, Jiang Y, Huang J, He Z and Tang Q: Moist exposed burn ointment accelerates diabetes-related wound healing by promoting re-epithelialization. Front Med (Lausanne). 9:10420152022. View Article : Google Scholar

258 

Ding R, Wang Y, Zhu JP, Lu WG, Wei GL, Gu ZC, An ZT and Huo JG: Danggui Sini decoction protects against oxaliplatin-induced peripheral neuropathy in rats. J Integr Neurosci. 19:663–671. 2020. View Article : Google Scholar : PubMed/NCBI

259 

Mabvuure NT, Brewer CF, Gervin K and Duffy S: The use of moist exposed burn ointment (MEBO) for the treatment of burn wounds: A systematic review. J Plast Surg Hand Surg. 54:337–343. 2020. View Article : Google Scholar : PubMed/NCBI

260 

Zhao D, Luo S, Xu W, Hu J, Lin S and Wang N: Efficacy and safety of hyperbaric oxygen therapy used in patients with diabetic foot: A meta-analysis of randomized clinical trials. Clin Ther. 39:2088–2094.e2. 2017. View Article : Google Scholar : PubMed/NCBI

261 

Damineni U, Divity S, Gundapaneni SRC, Burri RG and Vadde T: Clinical outcomes of hyperbaric oxygen therapy for diabetic foot ulcers: A systematic review. Cureus. 17:e786552025.PubMed/NCBI

262 

Huang X, Liang P, Jiang B, Zhang P, Yu W, Duan M, Guo L, Cui X, Huang M and Huang X: Hyperbaric oxygen potentiates diabetic wound healing by promoting fibroblast cell proliferation and endothelial cell angiogenesis. Life Sci. 259:1182462020. View Article : Google Scholar : PubMed/NCBI

263 

Borys S, Hohendorff J, Koblik T, Witek P, Ludwig-Slomczynska AH, Frankfurter C, Kiec-Wilk B and Malecki MT: Negative-pressure wound therapy for management of chronic neuropathic noninfected diabetic foot ulcerations-short-term efficacy and long-term outcomes. Endocrine. 62:611–616. 2018. View Article : Google Scholar : PubMed/NCBI

264 

Chen L, Zhang S, Da J, Wu W, Ma F, Tang C, Li G, Zhong D and Liao B: A systematic review and meta-analysis of efficacy and safety of negative pressure wound therapy in the treatment of diabetic foot ulcer. Ann Palliat Med. 10:10830–10839. 2021. View Article : Google Scholar : PubMed/NCBI

265 

Huang Y, Yu Z, Xu M, Zhao X, Tang Y, Luo L, Deng D and Chen M: Negative pressure wound therapy promotes wound healing by down-regulating miR-155 expression in granulation tissue of diabetic foot ulcers. Sci Rep. 15:67332025. View Article : Google Scholar : PubMed/NCBI

266 

Liu L, Chen R, Jia Z, Li X, Tang Y, Zhao X, Zhang S, Luo L, Fang Z, Zhang Y and Chen M: Downregulation of hsa-miR-203 in peripheral blood and wound margin tissue by negative pressure wound therapy contributes to wound healing of diabetic foot ulcers. Microvasc Res. 139:1042752022. View Article : Google Scholar

267 

Houreld NN: Shedding light on a new treatment for diabetic wound healing: A review on phototherapy. ScientificWorldJournal. 2014:3984122014. View Article : Google Scholar : PubMed/NCBI

268 

Houreld N and Abrahamse H: Low-intensity laser irradiation stimulates wound healing in diabetic wounded fibroblast cells (WS1). Diabetes Technol Ther. 12:971–978. 2010. View Article : Google Scholar : PubMed/NCBI

269 

Gupta A, Dai T and Hamblin MR: Effect of red and near-infrared wavelengths on low-level laser (light) therapy-induced healing of partial-thickness dermal abrasion in mice. Lasers Med Sci. 29:257–265. 2014. View Article : Google Scholar

270 

Cai W, Hamushan M, Zhang Y, Xu Z, Ren Z, Du J, Ju J, Cheng P, Tan M and Han P: Synergistic effects of photobiomodulation therapy with combined wavelength on diabetic wound healing in vitro and in vivo. Photobiomodul Photomed Laser Surg. 40:13–24. 2022.

271 

Frampton JE, Lee CR and Faulds D: Filgrastim. A review of its pharmacological properties and therapeutic efficacy in neutropenia. Drugs. 48:731–760. 1994. View Article : Google Scholar : PubMed/NCBI

272 

Edmonds M, Gough A, Solovera J and Standaert B: Filgrastim in the treatment of infected diabetic foot ulcers. Clin Drug Investig. 17:275–286. 1999. View Article : Google Scholar

273 

Orgill DP and Bayer LR: Negative pressure wound therapy: Past, present and future. Int Wound J. 10:15–19. 2013. View Article : Google Scholar : PubMed/NCBI

274 

Yoon D, Byun HJ, Oh SJ, Park JH and Lee DY: A case of cutaneous leukocytoclastic vasculitis associated with granulocyte colony-stimulating factor: An unusual presentation. Ann Dermatol. 32:164–167. 2020. View Article : Google Scholar

275 

Song P, Liang Q, Ge X, Zhou D, Yuan M, Chu W and Xu J: Adipose-derived stem cell exosomes promote scar-free healing of diabetic wounds via miR-204-5p/TGF-β1/Smad pathway. Stem Cells Int. 2025:63448442025. View Article : Google Scholar

276 

Van Netten JJ, Woodburn J and Bus SA: The future for diabetic foot ulcer prevention: A paradigm shift from stratified healthcare towards personalized medicine. Diabetes Metab Res Rev. 36(Suppl 1): e32342020. View Article : Google Scholar : PubMed/NCBI

277 

Ng GW, Gan KF, Liew H, Ge L, Ang G, Molina J, Sun Y, Prakash PS, Harish KB and Lo ZJ: A Systematic review and classification of factors influencing diabetic foot ulcer treatment adherence, in accordance with the WHO dimensions of adherence to long-term therapies. Int J Low Extrem Wounds. 20:153473462412339622024. View Article : Google Scholar

278 

Zhang YW, Sun L, Wang YN and Zhan SY: Role of macrophage polarization in diabetic foot ulcer healing: A bibliometric study. World J Diabetes. 16:997552025. View Article : Google Scholar : PubMed/NCBI

279 

Choudhury S, Dhoke NR, Chawla S and Das A: Bioengineered MSCCxcr2 transdifferentiated keratinocyte-like cell-derived organoid potentiates skin regeneration through ERK1/2 and STAT3 signaling in diabetic wound. Cell Mol Life Sci. 81:1722024. View Article : Google Scholar :

280 

de Souza A, Martignago CCS, Santo GDE, Sousa KDSJ, Cruz MA, Amaral GO, Parisi JR, Estadella D, Ribeiro DA, Granito RN, et al: 3D printed wound constructs for skin tissue engineering: A systematic review in experimental animal models. J Biomed Mater Res B Appl Biomater. 111:1419–1433. 2023. View Article : Google Scholar : PubMed/NCBI

281 

Yuan T, Tan M, Xu Y, Xiao Q, Wang H, Wu C, Li F and Peng L: All-in-one smart dressing for simultaneous angiogenesis and neural regeneration. J Nanobiotechnology. 21:382023. View Article : Google Scholar : PubMed/NCBI

282 

Mirani B, Hadisi Z, Pagan E, Dabiri SMH, van Rijt A, Almutairi L, Noshadi I, Armstrong DG and Akbari M: Smart dual-sensor wound dressing for monitoring cutaneous wounds. Adv Healthc Mater. 12:e22032332023. View Article : Google Scholar : PubMed/NCBI

283 

Su R, Wang L, Han F, Bian S, Meng F, Qi W, Zhai X, Li H, Wu J, Pan X, et al: A highly stretchable smart dressing for wound infection monitoring and treatment. Mater Today Bio. 26:1011072024. View Article : Google Scholar : PubMed/NCBI

284 

Lin B, Ma Y and Wu S: Multi-Omics and artificial intelligence-guided data integration in chronic liver disease: Prospects and challenges for precision medicine. OMICS. 26:415–421. 2022. View Article : Google Scholar : PubMed/NCBI

285 

Wang Z, Xu H, Xue B, Liu L, Tang Y, Wang Z and Yao K: MSC-derived exosomal circMYO9B accelerates diabetic wound healing by promoting angiogenesis through the hnRNPU/CBL/KDM1A/VEGFA axis. Commun Biol. 7:17002024. View Article : Google Scholar : PubMed/NCBI

286 

Liu J, Qu M, Wang C, Xue Y, Huang H, Chen Q, Sun W, Zhou X, Xu G and Jiang X: A dual-cross-linked hydrogel patch for promoting diabetic wound healing. Small. 18:e21061722022. View Article : Google Scholar : PubMed/NCBI

287 

Wu C, Long L, Zhang Y, Xu Y, Lu Y, Yang Z, Guo Y, Zhang J, Hu X and Wang Y: Injectable conductive and angiogenic hydrogels for chronic diabetic wound treatment. J Control Release. 344:249–260. 2022. View Article : Google Scholar : PubMed/NCBI

288 

Liu W, Zhai X, Zhao X, Cai Y, Zhang X, Xu K, Weng J, Li J and Chen X: Multifunctional double-layer and dual drug-loaded microneedle patch promotes diabetic wound healing. Adv Healthc Mater. 12:e23002972023. View Article : Google Scholar : PubMed/NCBI

289 

Xiong W, Zhang X, Zhou J, Chen J, Liu Y, Yan Y, Tan M, Huang H, Si Y and Wei Y: Astragaloside IV promotes exosome secretion of endothelial progenitor cells to regulate PI3KR2/SPRED1 signaling and inhibit pyroptosis of diabetic endothelial cells. Cytotherapy. 26:36–50. 2024. View Article : Google Scholar

290 

Lei T, Gao Y, Duan Y, Cui C, Zhang L and Si M: Panax notoginseng saponins improves healing of high glucose-induced wound through the GSK-3β/β-catenin pathway. Environ Toxicol. 37:1867–1877. 2022. View Article : Google Scholar : PubMed/NCBI

291 

Lu Y, Ding X, Qi F, Ru Y, Kuai L, Chen S, Yang Y, Li X, Li F, Li B, et al: Quyu Shengji formula facilitates diabetic wound healing via inhibiting the expression of prostaglandin transporter. Evid Based Complement Alternat Med. 2021:88499352021. View Article : Google Scholar : PubMed/NCBI

292 

Liu Y, Mo J, Liang F, Jiang S, Xiong J, Meng X and Mo Z: Pien-tze-huang promotes wound healing in streptozotocin-induced diabetes models associated with improving oxidative stress via the Nrf2/ARE pathway. Front Pharmacol. 14:10626642023. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

August-2025
Volume 56 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Song J, Zhao T, Wang C, Sun X, Sun J and Zhang Z: Cell migration in diabetic wound healing: Molecular mechanisms and therapeutic strategies (Review). Int J Mol Med 56: 126, 2025.
APA
Song, J., Zhao, T., Wang, C., Sun, X., Sun, J., & Zhang, Z. (2025). Cell migration in diabetic wound healing: Molecular mechanisms and therapeutic strategies (Review). International Journal of Molecular Medicine, 56, 126. https://doi.org/10.3892/ijmm.2025.5567
MLA
Song, J., Zhao, T., Wang, C., Sun, X., Sun, J., Zhang, Z."Cell migration in diabetic wound healing: Molecular mechanisms and therapeutic strategies (Review)". International Journal of Molecular Medicine 56.2 (2025): 126.
Chicago
Song, J., Zhao, T., Wang, C., Sun, X., Sun, J., Zhang, Z."Cell migration in diabetic wound healing: Molecular mechanisms and therapeutic strategies (Review)". International Journal of Molecular Medicine 56, no. 2 (2025): 126. https://doi.org/10.3892/ijmm.2025.5567