Skip to main content
International Journal of Nanomedicine logoLink to International Journal of Nanomedicine
. 2025 Sep 13;20:11211–11233. doi: 10.2147/IJN.S548986

Extracellular Vesicles in Skin: Biological Function and Therapeutic Potential

Siqi Yuan 1,2,3, Mengyu Jin 1,2,3, Yanyan Zhang 1,2,3, Mengting Zhang 4, Minjia Yuan 5, Xiaolei Ding 1,2,3,, Juan Wang 1,2,3
PMCID: PMC12442902  PMID: 40969665

Abstract

Extracellular vesicles (EVs) are nanoscale, lipid bilayer-enclosed particles containing bioactive molecules that play crucial roles in tissue homeostasis. These vesicles regulate cellular functions via delivery of protein, lipid, and nucleic acid cargos to target cells, thereby orchestrating essential biological processes, including cellular proliferation, activation, angiogenesis, and immune responses. The past decade has witnessed unprecedented growth in research examining EV functions in cutaneous physiology and pathogenesis. Here, we describe the biogenesis, composition and cellular uptake of EVs, critically highlighting the therapeutic effects and potential mechanisms of EVs from mammalian, plant and bacterial cells in skin diseases, including skin wound healing, hair growth, aging, inflammatory diseases and cancers. In addition, we discuss clinical translation challenges and outline future research directions for advancing EV-based therapeutics in clinical dermatology.

Keywords: extracellular vesicles, wound healing, hair growth, aging, inflammatory skin diseases, skin cancer

Graphical Abstract

graphic file with name IJN-20-11211-g0001.jpg

Introduction

The skin serves as a multifunctional barrier organ, performing critical protective roles including prevention of transepidermal water loss, defense against mechanical and environmental insults, thermoregulation, and immune surveillance.1 This complex organ comprises three distinct anatomical layers: the epidermis, dermis, and hypodermis, each containing specialized cellular populations. The epidermis contains proliferative and differentiated keratinocytes that form the outermost barrier, protecting against external insults and preventing water loss. The dermis primarily houses fibroblasts that provide structural support and confer skin elasticity. The deepest hypodermis predominantly consists of adipocyte-rich tissue. These structures collectively engage in sophisticated intercellular communication networks to preserve tissue homeostasis, the ability to maintain its stable structure and function by resisting stress and remodeling over various scales.2,3 Perturbations in these regulated homeostatic networks frequently lead to pathological conditions, ranging from impaired wound healing, aging, to inflammatory skin diseases, and even tumors, collectively posing substantial socioeconomic burdens.4–6 Currently available therapies mainly contain skincare, medicines, surgeries, and cell therapies. However, these strategies often yield only modest, transient benefits and carry side effects,4 necessitating deeper mechanistic insights and innovative therapeutic approaches.

Extracellular vesicles (EVs) are lipid bilayer-enclosed nanoparticles generated by diverse cell types, including mammalian, plant, and bacteria.7,8 EVs contain diverse bioactive molecules,9–11 and may transport these cargos to local tissue microenvironments or distant organs, where they are absorbed by recipient cells to modulate physiological and pathological processes, such as cell proliferation, migration, angiogenesis, and inflammatory responses.12–15 This highlights their dual roles as both pathogenic mediators and potential therapeutic agents. Due to their nanoscale size and intrinsic targeting capabilities enabled by specific surface markers, EVs have additionally been investigated as carriers for delivery of therapeutic compounds.16 Given their ubiquitous presence in biofluids, such as blood and urine, EVs also serve as diagnostic and prognostic markers.17–20

Accumulating evidence has established EVs as pivotal mediators of intercellular communication and promising therapeutic agents in maintaining skin homeostasis and treating cutaneous disorders.21–23 For instance, we recently demonstrated that EVs derived from Lactobacillus rhamnosus GG promoted angiogenesis and re-epithelialization, leading to accelerated skin wound healing.24 Compared to conventional dermatological therapy, EVs show multiple superior advantages, such as low immunogenicity, multifunctionality, biosafety, and targeted delivery,25 that confer them as a promising platform for skin therapy.

In this review, we summarize current knowledge on biogenesis, secretion, and uptake of EVs, as well as their crucial roles in skin physiology. We present the latest progress in EV-based therapeutic strategies for skin repair, aging, and disease treatment.

The Biology of EVs

Classification of EVs

EVs are heterogeneous particles, typically cup-shaped or spherical, with diameters ranging from 40 to 1000 nm.8 According to their dimensions and origins, EVs can be grouped into three types: exosomes, ectosomes, and apoptotic vesicles.10 Ectosomes, ranging from 100 to 1000 nm in diameter, are formed via plasma membrane outward budding driven by cytoskeletal contraction.26 Apoptotic vesicles, with diameters ranging from 100 to 2000 nm, are shed by cells during late apoptosis and encompass cytoplasmic components. Their release is often triggered by cellular stress and activation signals.27 The exosomes are the smallest EV subtypes, with diameters ranging from 30 to 150 nm, which are mainly derived from endosomal compartments. Unlike mammalian and plant extracellular vesicles (PEVs), bacterial EVs (BEVs) originate through distinct biogenesis pathways. Gram-negative bacteria primarily generate outer membrane vesicles (OMVs) via membrane blebbing or explosive cell lysis, whereas Gram-positive bacteria produce cytoplasmic membrane-derived vesicles (CMVs) primarily via blebbing.28 Notably, most studies on EVs do not strictly distinguish these subtypes.7,29 Hence, this review employs the unified term “EVs” to refer to all types of vesicles unless specific subtype identification is warranted.

The Composition and Functional Significance of EVs

EVs house a variety of bioactive compounds such as proteins, lipids, and nucleic acids, which participate in multiple cellular processes.30 For example, growth factors, such as VEGF, TGF-β and EGF in EVs, directly promote cell proliferation and migration.31 Meanwhile, tetraspanins-including CD9, CD63, CD81, and TSG101 not only mediate cell adhesion but also function as well-established EV biomarkers.32–34 The lipid composition of EVs primarily includes ceramide, sphingomyelin, phosphatidylserine, and cholesterol. A distinguishing feature of EVs is their elevated membrane lipid contents compared to their cells of origin, which likely contribute to their structural stability and membrane integrity.34,35 The nucleic acid components of EVs encompass various molecular species, including DNA, RNA, microRNA (miRNA), circular RNA (circRNA), and long non-coding RNA (lncRNA), which also mediate critical biological functions.34,36,37 For instance, pluripotent stem cell-derived epidermal organoids were enriched with miRNAs associated with cellular proliferation, migration, and angiogenesis, such as miR-31-5p, miR-146a-5p and miR-191-5p.38

The molecular composition of EVs varies significantly based on their cellular origin and the surrounding extracellular environment. This compositional variability is exemplified by serum EVs derived from psoriasis patients, which have elevated miR-199a-3p compared to those from healthy individuals. Notably, miR-199a-3p expression was positively correlated with psoriasis area and severity index score,39 underscoring the diagnostic value of circulating EVs. Cells or tissues under different culture conditions also generate EVs with distinct cargo profiles. EVs from three-dimensional dermal papilla (DP) spheroids contained elevated levels of pro-hair growth miRNAs compared to those from 2D monolayer cultures, thereby mediating superior hair growth-promoting activity.40 Collectively, EVs are promising diagnostic biomarkers due to their potential to mirror the physiological or disease conditions of their parent cells.

The Biogenesis of EVs

The generation of EVs primarily involves the formation of intracellular multivesicular bodies (MVBs) and EV secretion29 (Figure 1). Initially, the plasma membrane undergoes endocytosis to generate cup-shaped early-sorting endosomes (ESEs), which encapsulate cell surface proteins, extracellular constituents, such as lipids and proteins. Following this, the ESEs can merge with existing ESEs or exchange materials with the trans-Golgi network and endoplasmic reticulum, thereby expanding into the late endosomes, where a double invagination occurs and eventually generate MVBs containing intraluminal vesicles (ILVs), which are precursors to EVs.41 The MVBs are either broken down by lysosomes for recycling or merge with the plasma membrane, secreting ILVs as EVs.42 This process is controlled by various proteins, notably the endosomal sorting complex required for transport (ESCRT) machinery.43 The apoptotic cells protrude outward to form apoptotic bodies.

Figure 1.

Figure 1

Biogenesis pathways of main EVs. Exosomes initially form through plasma membrane invagination to generate early endosomes, which subsequently mature into late endosomes and ultimately form MVBs. MVBs either fuse with lysosomes to degrade their contents or merge with the plasma membrane to secrete exosomes into the extracellular space. Apoptotic bodies and ectosomes are generated through outward buds.

Abbreviations: BEVs, bacterial extracellular vesicles; MEVs, mammalian extracellular vesicles; MVBs, multivesicular bodies; PEVs, plant extracellular vesicles.

The Uptake of EVs

The mechanisms underlying EV uptake remain incompletely understood.44 Up to now, three primary pathways have been identified, including direct interaction, membrane fusion, and internalization.45,46 EVs can directly interact with recipient cells via ligand-receptor binding, triggering downstream signaling cascades and regulating recipient cells. In membrane fusion, the lipid bilayers of EVs and the target cell merge, facilitating direct luminal component release into the cytoplasm. This process initiates with a hemi-fusion stalk formation via the hydrophobic EVs-plasma membrane interactions, which then expands to create a consistent structure. In the case of internalization, EVs are engulfed by the recipient cells, merging with intracellular spaces or endosomal routes to discharge their contents. This intricate process leverages standard endocytic methods, including clathrin-dependent endocytosis, lipid raft-driven endocytosis, caveolin-aided endocytosis, phagocytosis, and macropinocytosis. Notably, these pathways are not mutually exclusive and may coexist with the same set of EVs, which potentially utilize multiple routes for uptake.

Following uptake, EVs deliver biological information to local or distant target cells via body fluids, influencing gene expression and cellular functions, consequently modulating diverse physiological and pathophysiological processes, including tissue repair, metabolic functions, aging and immune responses.

Biological Functions and Therapeutic Activities of EVs in Skin

EVs are enriched with a variety of bioactive molecules that modulate the functions of recipient cells. This intercellular communication contributes critically to skin homeostasis maintenance. For example, EVs-derived from erythrocytes, a cell type in circulation, can be converted to hair components and promote lipid formation of the skin surface, thereby maintaining cutaneous structural integrity.47 Consequently, their potential applications in dermatology have attracted considerable attention.21,48,49 Here, we summarize the latest advances in the role of EVs for skin-related conditions, highlighting their regenerative, hair growth-promoting, anti-aging, anti-inflammatory, and anticancer properties (Table 1).

Table 1.

The Effects of EVs on Skin Physiology and Pathology

Application Source Targets or Pathways Bioactive Cargos Processes and Effects References
Wound healing HUMSCs ITCH/JUNB/IRE1α miR-27b Activate keratinocytes and fibroblasts in vitro, accelerate wound healing in vivo [50]
RETNLG, SLC2A3, BCL2A1B ND Promote M2 macrophage polarization [51]
TLR4-NF-κB miR-181c Reduce inflammatory cytokine production [52]
Wnt4/β-catenin ND Promote angiogenesis [53]
PI3K/Akt; ERK1/2 miR-21-3p Promote angiogenesis and fibroblast function [54]
BMSCs ND miR-223 Promote M2 polarization of macrophages [55]
ADMSCs ND ND Promote M2 polarization of macrophages, activate endothelial cells and fibroblasts, facilitate wound healing in vivo [56]
miR-146a/Src ND Mitigate endothelial cell senescence, promote angiogenesis, promote wound healing in natural aging and type-2 diabetes mouse wound-healing models [57]
White adipose tissues ND ND Promote diabetic wound healing [58]
Endothelial cells YAP, PI3K/Akt/mTOR ND Promote fibroblast proliferation, increase microvascular density, collagen deposition, macrophage infiltration [59]
Epidermal stem cells PI3K/ protein kinase B, TGFβ, MAPK ND Reduce inflammation, promote M2 polarization of macrophages, activate wound cell proliferation [60]
Young fibroblasts Sirt7 miR-125b Promote fibroblast transdifferentiation into myofibroblasts [61]
Lactobacillus rhamnosus GG PI3K/AKT miR-21-5p Activate keratinocytes and endothelial cells in vitro, accelerate wound healing in excisional wound healing mice model [24]
Lactobacillus druckerii ND ND Promote the proliferation of skin cells, new blood vessel formation and wound healing in excisional wound healing mice model [62]
Dandelion S.aureus exotoxins ND Neutralize S. aureus exotoxins, accelerate S. aureus-associated wound healing [63]
Hair growth DP cells (human and mouse) Wnt/β/Shh/ TGF-β miR-140-5p Accelerate the onset of hair follicle anagen and delay catagen, enhance the proliferation and migration of ORS cells, promote HFSC proliferation and differentiation [64–66]
3D-cultured mouse DP cells Wnt/β-catenin miR-218-5p Promote the development of hair follicles [40]
Human fibroblasts Wnt/β-catenin; Axin2, Lef1 ND Promote the migration, proliferation, and differentiation of ORS cells and elongation of the hair shaft [67]
Murine macrophages (RAW264.7) Wnt/β-catenin, P-AKT Wnt3a and Wnt7b Promote the proliferation, migration, and levels of hair-inductive markers of DP cells, increase the hair shaft size in vivo [68]
Human ADMSCs Wnt/β-catenin Wnt3a Promote the proliferation of DP and ORS cells, increase hair shaft elongation [69]
Skin or hair follicle MSCs-originated apo-EVs Wnt/β-catenin ND Improve skin and hair follicle MSC functions, promote hair growth [70]
Anti-aging ADMSCs Src kinase signaling miR-146a Alleviate oxidative stress-induced endothelial cell senescence, restore endothelial cell functions, improve photoaging rat phenotypes [57, 71]
BMSCs MMP1 miR-29b-3p Reverse UVB-induced inhibition on HDF migration and oxidative stress [72]
Human dermal fibroblasts TNF-α/P-AKT/NF-κB, TGF-β miR-223, miR-133a, miR-196a, TIMP-1 Promote the proliferation and migration of fibroblasts, improve UVB-induced aging phenotype [73]
Atopic dermatitis ADMSCs ND ND Reduce pathological symptoms, inhibit the infiltration of inflammatory cells, restore epidermal barrier functions in AD mouse model [74, 75]
IFN-γ-primed MSCs JAK1, STAT6, IL-4/13 ND Attenuate skin inflammation, alleviate pruritus, and improve barrier function [76, 77]
Lactobacillus plantarum ND ND Inhibit the secretion of IL-6, restore the cell viability of keratinocytes, reduce epidermal thickening and the IL-4 level in S. aureus-induced mouse AD models [78]
Grapefruit JAK-STAT3/MAPK/PI3K-AKT Hexosylceramides and ceramides, cis-miR159a-3p Display anti‐inflammatory and antioxidant effects in keratinocytes and macrophages, mitigate DNCB-induced atopic dermatitis [79]
Vitiligo Keratinocytes SOX1, β-catenin miR-200c Enhance melanogenesis [80]
Psoriasis HUMSCs or IFN-γ-primed HUMSCs ND ND Inhibit the proliferation and activation of keratinocytes, peripheral blood mononuclear cells and T cells in vitro, reduce psoriasis symptoms including thickness, erythema, and scales of skin lesions, exhaust Th17 cells, increase Th2 cells, reduce the enrichment of inflammatory cytokines [81, 82]
ESCs IL-17, C5b-9 complex formation ND Inhibit the infiltration of neutrophils and cytokine production in vivo [83]
Human gingiva-derived MSCs ND ND Ameliorate murine psoriasis-like skin inflammation, reduce the levels of Th1- and Th17-related cytokines [84]
Cutibacterium acnes ND ND Inhibit the proliferation and migration of keratinocytes, inhibit inflammatory factors in vitro, restore microbiota diversity, and alleviate psoriasis symptoms in vivo [85]
Staphylococcus Epidermidis ATCC12228 strain IL-36R ND Reduce the characteristic psoriatic skin features, including acanthosis, cellular infiltrate, and IL-6, IL-23, IL-17F and IL-36R expression [86]
Parabacteroides goldsteinii ND ND Reduce epidermal hyperplasia, suppress skin inflammation and ameliorate symptoms of psoriasis [87]
MSCs-EVs with high PD-L1 expression PD-L1 ND Inhibit the activation of various immune cells, reshape the inflammatory ecosystem of psoriasis mouse models [88]
Melanoma Human melanoma cells (A375) CREB and STAT signaling PD-L1 Promote lipid metabolism and facilitate senescence in T cells [89]
Mouse melanoma cells (B16) VCAM-1, MHC-I Integrins (α4, α9 and β1), tumor antigens Induce transcriptional changes and proliferation of lymphatic endothelial cells, promote the apoptosis of CD8+ T cells [90]
Ginseng TLR-4, MyD88 Ceramide lipids and proteins Promote the M1 polarization of macrophages, increase the apoptosis of melanoma cells, suppress melanoma growth in vivo [91]
iPSCs ND ND Enhance dendritic cells-mediated antitumor immunity
Natural Killer Cells PARP Perforin and FasL Exert cytotoxic effects on melanoma cells in vitro and in vivo [92]
Engineered EVs from HUMSCs loading triptolide Caspase cascade ND Inhibit proliferation, invasion, and apoptosis of melanoma cells in vitro, inhibit tumor growth and extended the survival time in vivo [93]
Basal cell carcinoma The blood of metastasis BCC ND ND Promote fibroblast proliferation, migration, and invasion. [94]
Cutaneous squamous cell carcinoma M2 macrophage STEAP3 Circ_0088494 Inhibit Ferroptosis [95]
ALA-PDT-treated SCC ND ND Promote dendritic cell maturation [95]

Abbreviations: ADMSCs, adipose derived mesenchymal stem cells; apo-EVs, apoptotic EVs; BMSCs, bone marrow derived mesenchymal stem cells; DP, dermal papilla; ESCs, embryonic stem cells; HDF, human dermal fibroblast; HFSC, hair follicle stem cell; HUMSCs, human umbilical cord derived mesenchymal stem cells; iPSCs, induced pluripotent stem cells; MSCs, mesenchymal stem cells; ND, not determined; ORS, outer root sheath.

EVs and Wound Healing

Cutaneous damage and wounds represent major clinical challenges, imposing substantial social and medical burdens.96 Chronic conditions such as diabetes, ageing or infection, frequently lead to impaired wound healing.97,98 The wound healing process involves three precisely coordinated stages, mainly inflammation, proliferation, and remodeling.99 After injury, the vessels contract and initiate blood coagulation program. The immune cells, like neutrophils and macrophages, are quickly drawn to the wound sites to kill pathogens and release cytokines after injury. During the proliferative stage, keratinocytes, endothelial cells, and fibroblasts activate to restore the wound epithelial layer, form new blood vessels (neovascularization), and secrete extracellular matrix (ECM) to seal the wound. Subsequently, the ECM is degraded and remodeled. Dysregulation of cellular activity within the wound microenvironment is the primary factor contributing to delayed healing processes.

Emerging research underscores the promising role of EVs in wound healing through the regulation of cellular behaviors to promote re-epithelialization, angiogenesis, and the restoration of inflammatory responses (Figure 2). Among the most extensively studied EVs in regenerative medicine are those derived from mesenchymal stem cells (MSCs).100 EVs from human MSCs of the umbilical cord (HUMSCs) activated human keratinocytes and fibroblasts, thus enhanced re-epithelialization and collagen formation, and accelerated cutaneous wound healing in vivo.50 Since excessive inflammation compromises wound healing processes, a well-regulated inflammatory response is required. EVs from HUMSCs,52,101 bone marrow MSCs (BMSCs),55 and adipose MSCs (ADMSCs)56 have been shown to suppress excessive inflammation by reprogramming pro-inflammatory M1 macrophages into the anti-inflammatory M2 phenotype, thereby accelerating chronic wound healing. Pseudotime analysis of single-cell RNA sequencing further confirmed macrophage differentiation following the application of EVs from HUMSCs, leading to a shift from M1 macrophages to M2a and M2c phenotypes with enhanced anti-inflammatory capacities.51 Xuan et al identified miR-181c within these EVs as a key mediator of the anti-inflammatory effect, achieved by inhibiting Toll-like receptor 4 (TLR4) signaling in macrophages.52 Additionally, these EVs activated endothelial cells and alleviated oxidative stress-driven senescence, ultimately enhanced angiogenesis in chronic wounds.53,54,57,102,103 Notably, EVs derived from white adipose tissue exhibited functions similar to those of ADMSCs-EVs.58 Apart from MSCs-EVs, EVs produced by other mammalian sources, such as EVs from endothelial cells,59 epidermal stem cells (ESCs)60 and young fibroblasts61 have also been demonstrated to promote the healing of wounds. However, research on the application of PEVs and BEVs in cutaneous wound healing is still scarce. We have revealed the beneficial effects of gut probiotic Lactobacillus rhamnosus GG derived EVs in accelerating reepithelization and angiogenesis via activating PI3K/AKT signaling pathway.24 Especially, probiotic Lactobacillus druckerii-derived EVs seemed to have the ability to promote wound regeneration and reduce skin scar formation.62 However, the potential mechanisms are not clear. The PEVs from Dandelion showed anti-virulence activity by binding to Staphylococcus aureus (S. aureus) exotoxins, thus facilitating S. aureus infected wound healing.63 Due to their promising effects, the therapeutic capabilities of BEVs and PEVs in wound healing merit further investigation in future studies.

Figure 2.

Figure 2

The therapeutic application of EVs in cutaneous medicine and aesthetics. EVs from diverse sources, including MSCs, endothelial cells, fibroblasts, immune cells, tissue extracts, plants, and bacterial cultures (A), modulate key regenerative processes in dermatological applications including wound healing (B), hair follicle growth (C), and skin aging (D). Upward red arrows represent promotion; downward green arrows represent repression. Created with Biorender.com.

Abbreviations: ADMSCs, adipose mesenchymal stem cells; BMSCs, bone marrow mesenchymal stem cells; DAMPs, damage associated molecular patterns; DP, dermal papilla; ESCs, epidermal stem cells; HUMSCs, human umbilical cord mesenchymal stem cells; MSCs, mesenchymal stem cells; ORS, outer root sheath. TLR, Toll-like receptor.

Of note, nearly 250 EVs-related clinical trials are documented on ClinicalTrials.gov, spanning both therapeutic and diagnostic applications for multiple disease states, including 9 dedicated to wound healing (ClinicalTrials.gov). A recent Phase I clinical trial (Plexoval II, ACTRN12620000944932) assessed the safety of platelet-derived EVs in wound healing using healthy adult volunteers. Results confirmed a favorable safety profile, however, no significant difference in wound closure time was observed between treated and untreated wounds, which possibly attributed to inherently rapid healing capacity of healthy volunteers, thus the therapeutic efficacy in patients with impaired healing remains to be established.104 Another clinical pilot study to investigate the therapeutic efficacy of human adipose tissue derived EVs in wound healing has been completed, but the results have not been posted (NCT05475418). Other clinical trials remain primarily in recruitment phases, and no EV-based products have received regulatory approval for wound management.

EVs and Hair Growth

Hair growth is vital for health maintenance, physical appearance, and personal identity.12,105 Hair growth and regeneration are fueled by hair follicle stem cells (HFSCs) situated at the outer layer of the follicle called bulge, cycling between activation and dormancy.106–109 Correspondingly, the hair follicles undergo three consecutive phases of rest (telogen), growth (anagen), and regression (catagen).110 During telogen, HFSCs receive inhibitory signals, such as bone morphogenetic proteins (BMPs) produced by DP cells and remain quiescent. Upon entry into anagen, activation signals such as Wnt and Shh overwhelm inhibitory signals, leading dormant HFSCs to exit quiescence. Then, the HFSCs divide asymmetrically to produce transient amplifying cells, which generate hair matrix and sheath of the hair shaft at the follicle bulb. Simultaneously, the HFSCs also generate outer root sheath (ORS) cells. This process is governed by an intricate interaction among diverse cellular components and signaling pathways. Especially, the Wnt signaling pathway is prerequisite for the telogen-to-anagen transition.111 Disruptions in these regulatory mechanisms can lead to hair loss. Currently, the primary approaches to treating hair loss include pharmacological interventions and surgical procedures.112,113 However, conventional therapies are limited by potential side effects, suboptimal results and high costs, putting forward a critical need for innovative strategies.

Recent years have seen growing interest in EVs for their ability to influence hair growth cycles and enhance hair growth114 (Figure 2). DP cells are critical to inducing the hair cycle and regeneration via interacting with surrounding environment. For this reason, it is not surprising that the DP-derived EVs (DP-EVs) have a role in promoting hair growth. Human DP-EVs induced the proliferation of ORS cells and HFSCs through Wnt pathway, ultimately triggering hair cycle progression in mice.64–66 miRNAs within EVs are pivotal for cell-to-cell communication, as evidenced by the fact that miRNAs can directly control gene expression via either translational repression or mRNA degradation.115,116 miR-140-5p in DP-EVs directly targeted and suppressed BMP2, an established inhibitor of HFSCs, therefore promoting hair growth.66 Notably, EVs derived from spheroid-cultured DP cells exhibited significantly greater efficacy in accelerating hair regrowth and angiogenesis compared to those from 2D-cultured DP cells, underscoring the enhanced biological activity of EVs produced in 3D culture systems.40,117 Topical injection of 3D DP-EVs surpassed the pro-hair growth efficacy of 5% minoxidil, the first-line therapy for hair loss, while 2D DP-EVs showed comparable efficacy to minoxidil. No obvious toxicity was reported. Mechanistically, EVs derived from spheroid-cultured DP cells exhibited higher levels of miR-218-5p, which inhibited Wnt antagonist secreted frizzled-related protein 2 (SFRP2), thereby activating Wnt/β-catenin pathway and promoting hair follicle regeneration.40 Critically, 3D DP-EVs showed higher yields and enhanced therapeutic efficacy than that of 2D,117,118 supporting their scalability for clinical translation. Advancements in EV engineering address key clinical barriers, including rapid clearance and nontargeted distribution. Chen et al developed an innovative approach using degradable and injectable microgels to encapsulate DP-derived EVs (OSA-EVs). Compared to free DP-EVs, OSA-EVs demonstrated sustained release and improved retention in the skin, significantly enhancing therapeutic efficacy.119

Apart from DP-EVs, EVs derived from various cell types also demonstrated potential for hair regeneration. For instance, EVs from human fibroblasts enhanced ORS and DP cell proliferation in vitro and stimulated elongation of human hair shafts in cultured hair follicles.67 These effects are mediated through Wnt3a-induced stimulation of the Wnt signaling pathway. Similarly, macrophage-derived EVs are also enriched with Wnt3a, thus harboring hair-inductive capabilities.68 Furthermore, MSCs-EVs have been demonstrated to significantly stimulate hair growth.69 Interestingly, MSC transplantation, a widely recognized strategy for tissue regeneration, is accompanied by the production of a substantial number of apoptotic EVs (apo-EVs). Latest research showed that MSC-originated apo-EVs are essential in boosting hair development via activating Wnt/β-catenin in hair follicle MSCs.70 These results underscore the profound hair promoting capabilities of EVs, not only as carriers of regenerative molecules but also as key mediators of cellular signaling pathways essential for hair follicle regeneration.

EVs and Skin Aging

Aging involves a series of gradual transformations across cellular, tissue, organ, and systemic levels.120 With increasing global prevalence, aging has emerged as a significant public health challenge.121–123 Among its many manifestations, cutaneous aging has garnered particular attention from researchers due to its visible and impactful nature. Clinically, skin aging is characterized by wrinkles formation, a rough and uneven texture, impaired barrier function, and pigmentation. The skin aging process results from intrinsic factors like genetics and hormones, as well as extrinsic influences such as ultraviolet (UV) exposure (also known as photoaging), oxidative stress, smoking, dietary habits and the use of cosmetics.124,125 Since skin is frequently exposed to environmental insults, preventive measures against extrinsic aging factors, particularly UV radiation and pollution, are an essential part of routine skin care to reduce skin ageing.126

During skin aging, there are fewer key cell populations including fibroblasts and keratinocytes.127 Meanwhile, these senescent cells display irreversible growth arrest and develop senescence-associated secretory phenotypes, such as the production of inflammatory mediators that compromise tissue function. Recent research has shown that aged cells tend to produce more EVs, which disturbed the processes of cell growth and differentiation, ultimately skin homeostasis.128–131 Keratinocyte proliferation and differentiation constitute the key elements of epidermal homeostasis. Choi EJ et al suggested that, relative to those from youthful fibroblasts, EVs from senescent fibroblasts induced proinflammatory cytokines and exerted a reduced support on keratinocyte differentiation and wound closure.128–131 This effect seems at least in part associated with miR-23a-3p/ E-cadherin axis.130 The cargos of EVs originating from senescent cells vary significantly from those of non-senescent cells.132,133 For instance, senescent fibroblasts exhibited altered levels of proteins associated with wound healing and cell adhesion, highlighting the functional impacts.131

Extensive studies have underscored the anti-aging benefits of EVs134 (Figure 2). In vitro research indicated that ADSCs-EVs can reduce oxidative stress-triggered endothelial cell aging and recover endothelial cell functions, as evidenced by enhanced angiogenesis and migration capabilities. This protective effect appeared to be mediated through miR-146a-induced inhibition of Src kinase signaling.57 Furthermore, topical application of these EVs in a photoaging rat model significantly improved skin conditions by reducing epidermal thickness and increasing dermal thickness, emphasizing their therapeutic potential for treating photoaged skin.71 Similarly, Yang et al reported that BMSCs-EVs effectively suppressed UVB-induced senescent signals, such as matrix metalloproteinase-1 (MMP1), while restoring levels of type I procollagen and mitigating skin senescence.72 Further comparative studies have shown that EVs originating from 3D-cultured dermal fibroblast spheroids possess enhanced anti-aging effects on the skin relative to BMSCs-EVs, likely due to their enhanced bioactive cargo and functional potency.73 Herein, we propose that combination of EVs-based therapies with preventive measures, which provide additional anti-aging protection, would yield significant clinical benefits. Although limited evidence exists for this synergistic strategy, therapeutic potential of combinational therapy warrants further investigation.

EVs and Inflammatory Skin Diseases

Diverse inflammatory dermatopathies are marked by pathological deterioration of skin tissue driven by immune imbalance. Among the most prevalent chronic inflammatory skin diseases are atopic dermatitis (AD), vitiligo, and psoriasis (Figure 3). Key cytokines implicated in these inflammatory responses include interleukin-4 (IL-4), IL-13, IL-17, and tumor necrosis factor-alpha (TNF-α), which collectively shape the intricate immunological landscape of these diseases.135,136

Figure 3.

Figure 3

The role of EVs in inflammatory skin diseases. (A) EVs can promote de novo ceramide synthesis, restore epidermal barrier function and downregulate inflammatory responses, thereby alleviating AD. In addition, the EVs released by S. aureus upregulate inflammatory factor levels in keratinocytes. (B) EVs from keratinocytes stimulate the proliferation and melanin synthesis of melanocytes, thereby alleviating vitiligo. (C) EVs exert beneficial effects on psoriasis via regulating skin barrier and inflammatory responses. Tregs, regulatory T cells. Upward red arrows represent promotion; downward green arrows represent repression. Created with Biorender.com.

Abbreviations: AD, atopic dermatitis; ROS, reactive oxygen species; S. aureus, Staphylococcus aureus.

AD

AD is a chronic inflammatory skin disorder marked by dry, scaly skin, intense itching and recurrent eczematous lesions.137–139 The pathogenesis of AD mainly involves epidermal barrier dysfunction, Th2-type immune responses, microbial dysregulation, and neuroimmune interactions.139 The beneficial effects of MSCs from ADMSCs, BMSCs and HUMSCs have been reported.140 Therefore, the application of MSC-EVs for AD treatment also gained significant attention as a novel cell-free therapeutic approach25,79 (Figure 3). In two independent studies, EVs derived from ADMSCs were shown to alleviate AD symptoms in mouse models, as evidenced by reduced clinical scores, downregulated levels of inflammatory cytokines and reduced mast cells.74,75 These therapeutic effects were associated with the restoration of epidermal barrier function through the promotion of de novo ceramide synthesis, a critical component of the skin protective barrier.75 Approaches to pre-treatment have been applied to enhance the therapeutic efficiency and reliability of EVs. For instance, EVs derived from interferon-gamma (IFN-γ)-primed MSCs were found to downregulate the expression of Th2 cytokine receptors, including IL-4Rα, IL-13Rα1, and IL-31Rα, as well as their downstream signaling mediators JAK1 and STAT, thereby mitigating AD symptoms in mice.76,77 Furthermore, these EVs appeared to recover epidermal barrier function through increasing lipid production, further highlighting their therapeutic potential.76,77 Both S. aureus and Lactobacillus plantarum are naturally present on the skin, but their roles in skin health and disease are markedly different. EVs released by S. aureus have been shown to induce and exacerbate AD-like skin inflammation, whereas EVs derived from Lactobacillus plantarum have demonstrated protective effects, such as reducing epidermal thickening and lowering IL-4 levels in S. aureus-induced mouse models of AD.78 The study underscored the intricate function of EVs from skin microbiome bacteria in modulating skin inflammation and suggested that focusing on certain bacterial EVs might offer an innovative therapeutic approach to AD. While the impact of BEVs on skin health has been increasingly studied, research on PEVs remains limited. Recently, an interesting study investigated the healing possibilities of EVs isolated from grapefruit, revealing the significant anti-inflammatory and antioxidant properties in vitro. These grapefruit-EVs were shown to reduce reactive oxygen species (ROS) levels and suppress the proliferation of CD4+ T cells, leading to alleviated AD symptoms in vivo.79 Notably, lipidomic analysis revealed that the lipid composition of grapefruit-EVs closely resembled that of the parent plant tissue, suggesting that EVs retain key bioactive components of their sources. This finding supports the view that EVs can serve as a natural and effective treatment option for inflammatory skin diseases.

Vitiligo

Vitiligo is an autoimmune disorder characterized by the destruction of pigment-producing melanocytes in the skin, resulting in depigmented patches.141–143 It affects approximately 1% of the global population, posing significant psychological and social challenges for patients. The pathogenesis of vitiligo is primarily driven by autoimmune dysfunction, particularly an imbalance in the adaptive immune system involving CD8+ T cells, regulatory T cells (Tregs), and dendritic cells, which attack and destroy melanocytes.144–146 In addition, oxidative stress is believed to be an initiating factor in melanocyte loss. Excessive ROS induces DNA damage and protein misfolding, ultimately leading to melanocyte deficiency. Current therapies, including topical corticosteroids, phototherapy and immunosuppressants, often fail to meet clinical requirements.142 Recently, EVs have attracted considerable interest within vitiligo research studies, offering new insights into disease progression and therapy147,148 (Figure 3). For example, EVs isolated from vitiligo patients exhibited high levels of miR-493-3p and miR-21-5p, which increased ROS, promoted melanocyte apoptosis, and inhibited the melanogenesis of melanocytes, underscoring their detrimental effects.149,150 Conversely, miR-200c was downregulated in EVs keratinocytes in vitiligo lesions. This downregulation promotes the expression of melanogenesis-related genes by suppressing SOX1, which leads to β-catenin activation.80 In addition, EVs from keratinocytes of healthy individuals can be taken up by melanocytes and stimulate their proliferation and melanin synthesis.151 These results emphasize the promise of EVs and their molecular contents as therapeutic targets and biomarkers for vitiligo.

Psoriasis

Psoriasis is a widespread, chronic skin condition marked by papulosquamous features, exhibiting notable genetic susceptibility and autoimmune mechanisms.152 Immunological and genetic research has pinpointed IL-17 and IL-23 as crucial triggers in the pathogenesis of psoriasis.152,153 In recent years, EVs have gained attention as a potential treatment option for psoriasis (Figure 3). Among the most studied are MSCs-derived EVs, particularly those sourced from HUMSCs, IFN-γ primed HUMSCs, embryonic stem cells, and gingival, which have demonstrated significant anti-inflammatory and immunomodulatory effects. Notably, these EVs significantly reduced key inflammatory cytokines, such as IL-17, IL-23 and TNF-α in psoriasis-like mouse models.81–84 It was also reported that EVs from skin commensal bacteria (Cutibacterium acnes, Staphylococcus Epidermidis ATCC12228 strain) and gut commensal bacteria (Parabacteroides goldsteinii) restored skin barrier function and attenuated the infiltration of pro-inflammatory cells, thereby downregulating the level of inflammatory cytokines, consequently alleviating psoriasis progression.85–87 Of note, Cutibacterium acnes-derived EVs also restored the microbiota diversity on the skin of mice, reduced the colonization of S. aureus.85 EVs from Parabacteroides goldsteinii have shown good stability, and upon oral administration, they can travel to the colon, cross the intestinal barrier, and reach inflamed skin,87 indicating a novel therapeutic option based on the EV-mediated gut-skin axis.

Beyond naturally occurring EVs, engineered EVs loaded with specific therapeutic cargos via gene transfection have also shown promising results in psoriasis treatment.88,154,155 Programmed cell death-ligand 1 (PD-L1) is one of the key components to inhibit immune responses and prevent immune cells from indiscriminately attacking self-tissue.156 MSCs-EVs with PD-L1 over-expression effectively remodeled the inflammatory ecosystem in the affected skin tissues, suppressing key inflammatory cytokines in both skin and system, including IL-17A, TNF-α, IL-4 and IFN-γ via PD-1/PD-L1 axis. Notably, these EVs displayed enhanced efficacy than conventional MSCs-EVs in imiquimod-induced psoriasis.88

In summary, existing research on EVs in inflammatory skin conditions represents just a small fraction of the potential knowledge, and there is still a huge potential to be explored.

EVs and Skin Cancers

By facilitating cell-to-cell communication, EVs critically influence tumor development and progression, positioning them as promising therapeutic agents. Additionally, their inherent stability, easy accessibility and dynamic cargo alterations across distinct pathologic states render them as attractive diagnostic tools via comprehensive omics techniques (Figure 4). While EV research is well-established in many cancers, investigations into their functions in skin malignancies remain at an early stage. In this part, we discuss the role of EVs in skin cancers, including melanoma and non-melanoma skin cancers (NMSCs), focusing on their diagnostic and therapeutic potential.

Figure 4.

Figure 4

The role of EVs in cancers. (A) EVs function as diagnostic tools in tumor research fields. (B) EVs modulate multiple biological processes, including apoptosis, angiogenesis, metastasis, EMT, immunity, and ECM remodeling. (C) EVs function as therapeutic agents. Created by biorender.

Abbreviations: ECM, extracellular matrix; EMT, epithelial-mesenchymal transition.

Melanoma

Melanoma is one of the most dangerous and aggressive cutaneous malignancies, causing 75% of skin cancer-related deaths, even though it constitutes only 4% of all skin cancers.157–159 Even with modern therapies, overall survival rates remain suboptimal, partly due to delayed diagnosis, metastasis and therapeutic resistance. Consequently, there is an urgent need to identify novel biomarkers and develop effective therapeutic strategies.

To date, several studies have revealed the pro-tumorigenic properties of melanoma EVs, including their role in immunosuppression, tumor metastasis, angiogenesis, and drug resistance.160,161 Ma et al reported that tumor EVs were enriched in PD-L1, which facilitated T cell senescence and conferred immunotherapy resistance in melanoma models. Conversely, inhibiting EV synthesis reversed these effects.89 In addition, melanoma cell B16F10-derived EVs induced the proliferation of lymphatic endothelial cells in draining lymph nodes, a process associated with lymphatic metastasis, while simultaneously induced apoptosis of CD8⁺ T cells to facilitate tumor immune evasion and therapy resistance.90 These findings highlight the potential of EVs as therapeutic targets.

EVs have gained significant attention as novel diagnostic biomarkers in melanoma (Table 2). Patients with melanoma showed upregulated EV concentrations compared to healthy individuals.169 Through omics profiling, several bioactive molecules in EVs have been identified as valuable biomarkers, enabling both early diagnosis and treatment in melanoma. Among those, miRNAs represent the most extensively investigated diagnostic biomarkers for melanoma.170,171 Xiong et al revealed that the elevated miR-550a-3p and reduced miR-150-5p levels in plasma-derived EVs from melanoma patients were significantly associated with decreased overall survival.162 Additionally, high enrichment of miR-92b-3p, miR-182-5p, miR-183-5p, miRNA-532-5p and miRNA-106b in serum-derived EVs from melanoma patients was also identified, but not the healthy ones.163,164 Bioinformatics analyses confirmed the strong association between miR-92b-3p, miR-182-5p, miR-183-5p, and oncogenic pathways and tumor progression.163 Meanwhile, miR-532-5p and miR-106b panel also differentiated metastatic melanoma patients from non-metastatic subjects, and stratified disease stages.164 In patients with metastatic sporadic melanoma, EV miR-17, miR-19a, miR-21, miR-126, and miR-149 in plasma had higher levels than in unaffected individuals and familial melanoma patients, supporting their utility as metastasis-predictive biomarkers.165 EV-enriched proteins have also emerged as promising melanoma biomarkers. For example, García-Silva et al showed that nerve growth factor receptor (NGFR) was enriched in EVs from metastatic melanoma cell lines.166 This increase was associated with lymphangiogenesis and tumor adhesion, indicative of the diagnostic metastasis. Additionally, several independent studies revealed that PD-L1 in circulating EVs can serve as a diagnostic biomarker for immunotherapy outcome and stratification of responders versus non-responders.167,172,173 This finding is significant, since the direct detection of PD-L1 is difficult due to the unstable nature of proteins.

Table 2.

EVs as Diagnostic Biomarkers in Skin Cancers

Caner Type Biomarker Sources Clinical Value References
Melanoma miR-550a-3p Plasma Expression levels of miR-550a-3p are negatively correlated with overall survival. [162]
miR-150-5p Plasma Expression levels of miR-150-5p are positively correlated with overall survival. [162]
miR-92b-3p, miR-182-5p, miR-183-5p Serum Diagnostic biomarkers [163]
miR-532-5p, miR-106b Serum Diagnosis and stage stratification [164]
miR-17, miR-19a, miR-21, miR-126,
and miR-149
Plasma Diagnostic biomarker of metastasis [165]
NGFR Metastatic melanoma cells Diagnostic biomarker of metastasis [166]
PD-L1 Blood Prognostic biomarker for anti-PD-1 therapies in metastatic melanoma, stratification of responders from non-responders [167]
BCC miR-197-5p, miR-365b-5p, miR-6503-3p,
miR-5190, miR-3940-3p, miR-3909, miR-615-3p, miR-548am-5p, miR-548ah-3p
Blood Diagnostic biomarker of metastasis [94]
SCC Circ-CYP24A1 Plasma Diagnostic biomarker [168]

Abbreviations: BCC, basal cell carcinoma; SCC, squamous cell carcinoma.

The therapeutic applications of EVs on melanoma have also been reported. Tumor-associated macrophages (TAMs) exhibited a biphasic nature, characterized by tumor-suppressing M1 phenotype or tumor-promoting M2 phenotype.174 Cao et al found that ginseng-derived EVs reprogrammed M2 macrophages to M1 polarization, and therefore inhibited melanoma growth.91 Dendritic cells are critical antigen presenting cells to boost tumor immunity.175 Induced pluripotent stem cells (iPSCs) pulsed dendritic cells displayed enhanced immune response.176 Natural killer cells-derived EVs remarkably inhibited the growth of melanoma by inducing apoptosis in vitro and in vivo.92 Alongside natural EVs, engineered EVs with enhanced therapeutic efficacy have also been developed for melanoma. Multiple EV modification strategies have been developed, such as chemical modification and drug loading.177 TLR3/7/8/9 participate in regulating endocytosis of EVs by dendritic cells via endosomes.178 Using click chemistry, Tang et al developed engineered tumor EVs with the display of TLR9 agonists on the surface, which stimulated TLR signals on intracellular endosomes, consequently enhancing dendritic cell activation, improving the processing and presentation of antigens encapsulated in EVs and priming T cells.179 This cascade led to improved antitumor efficacy against melanoma. Additionally, to overcome the aqueous insolubility and short half-life of triptolide, an anticancer agent, Gu et al developed an EV platform to load triptolide using EVs from HUMSCs, referred to as cRGD-Exo/TP. These engineered EVs displayed enhanced tumor targeting and prolonged triptolide pharmacokinetics, ultimately inhibiting tumor growth with significant survival benefit in malignant melanoma models.93

NMSCs

NMSCs are far more common than melanoma and primarily comprise basal cell carcinoma (BCC) and squamous cell carcinoma (SCC).180,181 BCC is the most common skin cancer, with around 3.6 million new annual diagnoses globally.182 Although early-stage BCC is highly treatable, managing locally advanced or metastatic BCC continues to pose significant clinical challenges.183 A study comparing EVs from the peripheral blood of metastatic BCC and non-metastatic BCC (NMBCC) patients, revealed that NMBCC-derived EVs exhibit greater potential to promote fibroblast proliferation, migration, and invasion.94 miRNA sequencing identified nine upregulated miRNA in metastatic BCC-derived EVs, suggesting their potential utility as prognostic biomarkers for metastatic progression (Table 2). However, the specific miRNAs or other key bioactive components responsible for mediating these distinct functions remain determined.

SCC is more invasive and prone to spreading than BCC, especially in immunocompromised patients or those with prolonged UV exposure.184 The pathogenic functions of EVs have been studied. It has been reported that pro-oncogenic Dsg2 enhanced EV secretion from SCC cells, leading to increased IL-8 packaging in EVs and promoting SCC progression.185 These EVs contained a high level of miRNA-31, which directly targeted RhoBTB1 and resulted in SCC cell proliferation, migration, and invasion.186 In addition, M2 macrophage-derived EVs promote SCC progression by inhibiting ferroptosis, a form of cell death.95 5-Aminolevulinic acid photodynamic therapy (ALA-PDT) is an effective therapeutic strategy for SCC. Interestingly, EVs from ALA-PDT-treated SCC cells induced dendritic cell maturation.187 These studies reveal the complicated role of EVs in tumor microenvironment.

Compared with melanoma, the diagnostic utility of EVs in SCC is less explored. Zhang et al reported that, compared with healthy individuals, cutaneous SCC patients displayed elevated expression of circ-CYP24A1 in EVs from plasma.168 Critically, circ-CYP24A1 promoted the proliferation, migration, and invasion of SCC cells, supporting its dual utility as both an early diagnostic biomarker and therapeutic target.

Collectively, research on EVs in skin cancers remains in its infancy. Investigating how EVs influence the tumor microenvironment and engage with immune cells could provide valuable insights into skin cancer pathogenesis and uncover novel therapeutic targets. Furthermore, the integration of multi-omics approaches, such as proteomics and transcriptomics, could deepen our understanding of EV biology and identify novel biomarkers for early detection and treatment monitoring. By leveraging these advancements, researchers and clinicians can move closer to achieving precision medicine in melanoma.

Conclusion and Perspectives

The application of EVs in dermatology is a rapidly evolving field with transformative potential for treating diverse cutaneous disorders. EVs exhibit remarkable capabilities in immune modulation and cellular activation through delivering bioactive molecules, making them a promising tool for treating diseases such as chronic wound healing, psoriasis, AD, vitiligo, and even skin cancers. However, despite their therapeutic promise, several significant challenges must be addressed to facilitate the widespread clinical adoption of EVs.

A significant obstacle is the absence of standardized procedures for EV extraction, preservation, and analysis. The present techniques for isolating particles, like ultracentrifugation, size-exclusion chromatography, and chemical precipitation, often yield a mixture of vesicles with varied sizes, which can mess with the consistency of results and complicate the follow-up analysis. Additionally, the complex and diverse composition of EVs, which includes proteins, lipids, nucleic acids, and other biomolecules, makes it difficult to pinpoint the specific mechanisms by which they exert their therapeutic effects. This heterogeneity also poses challenges for quality control and batch-to-batch consistency, which are critical for clinical translation. Moreover, most current studies are performed on cells or animal models. Despite significant preclinical efficacy, biological differences between species, such as immune responses, skin structure, and disease progression, limit the direct applicability of EV findings to humans, necessitating rigorous validation in clinical trials. However, early-phase trials with EVs concentrate predominantly on safety and effectiveness evaluations. Although EVs are already marketed in beauty/skincare and undergoing Phase III trials for pharmaceutical applications, significant uncertainties regarding their commercial viability persist. Critical considerations, including scalability, economic viability, regulatory clearance, and enduring safety concerns, must be resolved before EVs are widely adopted as a standard treatment method.

To overcome these challenges, future research should prioritize the development of standardized protocols for EV production and characterization, as well as the elucidation of their precise mechanisms of action. Advanced technologies, such as single-vesicle analysis and multi-omics approaches, could provide deeper insights into EV composition and function, enabling the design of more targeted and effective therapies. Additionally, robust preclinical models that better mimic human skin biology and disease pathology will be essential for bridging the gap between animal studies and human trials. Joint efforts by scientists, healthcare professionals, corporate partners, and governing entities are essential in progressing EV-derived treatments from research settings to clinical use. Through a structured approach to tackling these technical and translational obstacles, the sector can maximize the therapeutic utility of EVs, facilitating the development of innovative, minimally invasive, and highly effective treatments for complex dermatological conditions. Integration of cutting-edge analytical tools with enhanced mechanistic understanding will accelerate the emergence of precision-based, personalized EV therapeutics tailored to diverse patient needs.

Funding Statement

This work was supported by National Natural Science Foundation of China (82204450 and 82272278), the National Key Research and Development Program of China (2023YFC2509000), and Shanghai Qiran Biotechnology Co., Ltd.

Data Sharing Statement

Data sharing not applicable - no new data generated.

Author Contributions

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

Disclosure

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

References

  • 1.Fuchs E. Scratching the surface of skin development. Nature. 2007;445(7130):834–842. doi: 10.1038/nature05659 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Rognoni E, watt FM. Skin Cell Heterogeneity in Development, Wound Healing, and Cancer. Trends Cell Biol. 2018;28(9):709–722. doi: 10.1016/j.tcb.2018.05.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Meizlish ML, Franklin RA, Zhou X, Medzhitov R. Tissue Homeostasis and Inflammation. Annu Rev Immunol. 2021;39:557–581. doi: 10.1146/annurev-immunol-061020-053734 [DOI] [PubMed] [Google Scholar]
  • 4.Urban K, Chu S, Giesey RL, et al. Burden of skin disease and associated socioeconomic status in Asia: a cross-sectional analysis from the Global Burden of Disease Study 1990-2017. JAAD Int. 2021;2:40–50. doi: 10.1016/j.jdin.2020.10.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kong Y, Xing P, Huai P, Zhang F. The burden of skin diseases in China: global burden of Disease Study 2019. Arch Dermatol Res. 2024;316(6):277. doi: 10.1007/s00403-024-03046-5 [DOI] [PubMed] [Google Scholar]
  • 6.Flohr C, Hay R. Putting the burden of skin diseases on the global map. Br J Dermatol. 2021;184(2):189–190. doi: 10.1111/bjd.19704 [DOI] [PubMed] [Google Scholar]
  • 7.Welsh JA, Goberdhan DCI, O’Driscoll L, et al. Minimal information for studies of extracellular vesicles (MISEV2023): from basic to advanced approaches. J Extracell Vesicles. 2024;13(2):e12404. doi: 10.1002/jev2.12404 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Li Q, Li B, Ye T, et al. Requirements for human mesenchymal stem cell-derived small extracellular vesicles. Interdisciplinary Medicine. 2023;1(1):e20220015. DOI: 10.1002/INMD.20220015 [DOI] [Google Scholar]
  • 9.Raposo G, Stahl PD. Extracellular vesicles: a new communication paradigm? Nat Rev Mol Cell Biol. 2019;20(9):509–510. doi: 10.1038/s41580-019-0158-7 [DOI] [PubMed] [Google Scholar]
  • 10.Jeppesen DK, Zhang Q, Franklin JL, Coffey RJ. Extracellular vesicles and nanoparticles: emerging complexities. Trends Cell Biol. 2023;33(8):667–681. doi: 10.1016/j.tcb.2023.01.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Jeppesen DK, Fenix AM, Franklin JL, et al. Reassessment of Exosome Composition. Cell. 2019;177(2):428–445.e418. doi: 10.1016/j.cell.2019.02.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.A SEL, Mäger I, Breakefield XO, Wood MJ. Extracellular vesicles: biology and emerging therapeutic opportunities. Nat Rev Drug Discov. 2013;12(5):347–357. doi: 10.1038/nrd3978 [DOI] [PubMed] [Google Scholar]
  • 13.Tkach M, Théry C. Communication by Extracellular Vesicles: where We Are and Where We Need to Go. Cell. 2016;164(6):1226–1232. doi: 10.1016/j.cell.2016.01.043 [DOI] [PubMed] [Google Scholar]
  • 14.Yin Y, Chen H, Wang Y, Zhang L, Wang X. Roles of extracellular vesicles in the aging microenvironment and age-related diseases. J Extracell Vesicles. 2021;10(12):e12154. doi: 10.1002/jev2.12154 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Kalluri R. The biology and function of extracellular vesicles in immune response and immunity. Immunity. 2024;57(8):1752–1768. doi: 10.1016/j.immuni.2024.07.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Herrmann IK, Wood MJA, Fuhrmann G. Extracellular vesicles as a next-generation drug delivery platform. Nat Nanotechnol. 2021;16(7):748–759. doi: 10.1038/s41565-021-00931-2 [DOI] [PubMed] [Google Scholar]
  • 17.Das S, Lyon CJ, Hu T. A Panorama of Extracellular Vesicle Applications: from Biomarker Detection to Therapeutics. ACS Nano. 2024;18(14):9784–9797. doi: 10.1021/acsnano.4c00666 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Merchant ML, Rood IM, Deegens JKJ, Klein JB. Isolation and characterization of urinary extracellular vesicles: implications for biomarker discovery. Nat Rev Nephrol. 2017;13(12):731–749. doi: 10.1038/nrneph.2017.148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Medhin LB, Beasley AB, Warburton L, Amanuel B, Gray ES. Extracellular vesicles as a liquid biopsy for melanoma: are we there yet? Semin Cancer Biol. 2023;89:92–98. doi: 10.1016/j.semcancer.2023.01.008 [DOI] [PubMed] [Google Scholar]
  • 20.Park YJ, Kim DC, Lee SJ, et al. Keratinocyte-derived circulating microRNAs in extracellular vesicles: a novel biomarker of psoriasis severity and potential therapeutic target. J Transl Med. 2024;22(1):235. doi: 10.1186/s12967-024-05030-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wang J, Yuan S, Tu Y, et al. Extracellular vesicles in skin health, diseases, and aging. Interdisciplinary Medicine. 2024;2(3):e20240011. DOI: 10.1002/INMD.20240011 [DOI] [Google Scholar]
  • 22.Fusco C, De Rosa G, Spatocco I, et al. Extracellular vesicles as human therapeutics: a scoping review of the literature. J Extracell Vesicles. 2024;13(5):e12433. doi: 10.1002/jev2.12433 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Yuan Y, Cao K, Gao P, et al. Extracellular vesicles and bioactive peptides for regenerative medicine in cosmetology. Ageing Res Rev. 2025;107:102712. doi: 10.1016/j.arr.2025.102712 [DOI] [PubMed] [Google Scholar]
  • 24.Wang J, Li X, Zhao X, et al. Lactobacillus rhamnosus GG-derived extracellular vesicles promote wound healing via miR-21-5p-mediated re-epithelization and angiogenesis. J Nanobiotechnology. 2024;22(1):644. doi: 10.1186/s12951-024-02893-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Yu H, Feng H, Zeng H, et al. Exosomes: the emerging mechanisms and potential clinical applications in dermatology. Int J Biol Sci. 2024;20(5):1778–1795. doi: 10.7150/ijbs.92897 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Hu Y, Sun Y, Wan C, et al. Microparticles: biogenesis, characteristics and intervention therapy for cancers in preclinical and clinical research. J Nanobiotechnology. 2022;20(1):189. doi: 10.1186/s12951-022-01358-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Zhou M, Li YJ, Tang YC, et al. Apoptotic bodies for advanced drug delivery and therapy. J Control Release. 2022;351:394–406. doi: 10.1016/j.jconrel.2022.09.045 [DOI] [PubMed] [Google Scholar]
  • 28.Xie J, Haesebrouck F, Van Hoecke L, Vandenbroucke RE. Bacterial extracellular vesicles: an emerging avenue to tackle diseases. Trends Microbiol. 2023;31(12):1206–1224. doi: 10.1016/j.tim.2023.05.010 [DOI] [PubMed] [Google Scholar]
  • 29.Dixson AC, Dawson TR, Di Vizio D, Weaver AM. Context-specific regulation of extracellular vesicle biogenesis and cargo selection. Nat Rev Mol Cell Biol. 2023;24(7):454–476. doi: 10.1038/s41580-023-00576-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Kumar MA, Baba SK, Sadida HQ, et al. Extracellular vesicles as tools and targets in therapy for diseases. Signal Transduct Target Ther. 2024;9(1):27. doi: 10.1038/s41392-024-01735-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Huang J, Wang X, Wang Z, et al. Extracellular vesicles as a novel mediator of interkingdom communication. Cytokine Growth Factor Rev. 2023;73:173–184. doi: 10.1016/j.cytogfr.2023.08.005 [DOI] [PubMed] [Google Scholar]
  • 32.Keulers TG, Libregts SF, Beaumont JEJ, et al. Secretion of pro-angiogenic extracellular vesicles during hypoxia is dependent on the autophagy-related protein GABARAPL1. J Extracell Vesicles. 2021;10(14):e12166. doi: 10.1002/jev2.12166 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Wu J, Huang QM, Liu Y, et al. Long-term hypoxic hUCMSCs-derived extracellular vesicles alleviates allergic rhinitis through triggering immunotolerance of their VEGF-mediated inhibition of dendritic cells maturation. Int Immunopharmacol. 2023;124(Pt B):110875. doi: 10.1016/j.intimp.2023.110875 [DOI] [PubMed] [Google Scholar]
  • 34.van Niel G, D’Angelo G, Raposo G. Shedding light on the cell biology of extracellular vesicles. Nat Rev Mol Cell Biol. 2018;19(4):213–228. doi: 10.1038/nrm.2017.125 [DOI] [PubMed] [Google Scholar]
  • 35.Crivelli SM, Giovagnoni C, Zhu Z, et al. Function of ceramide transfer protein for biogenesis and sphingolipid composition of extracellular vesicles. J Extracell Vesicles. 2022;11(6):e12233. doi: 10.1002/jev2.12233 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Fabbiano F, Corsi J, Gurrieri E, et al. RNA packaging into extracellular vesicles: an orchestra of RNA-binding proteins? J Extracell Vesicles. 2020;10(2):e12043. doi: 10.1002/jev2.12043 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Zhao J, Li Q, Hu J, et al. Circular RNA landscape in extracellular vesicles from human biofluids. Genome Med. 2024;16(1):126. doi: 10.1186/s13073-024-01400-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Kwak S, Song CL, Lee J, et al. Development of pluripotent stem cell-derived epidermal organoids that generate effective extracellular vesicles in skin regeneration. Biomaterials. 2024;307:122522. doi: 10.1016/j.biomaterials.2024.122522 [DOI] [PubMed] [Google Scholar]
  • 39.Wang ZY, Yan BX, Zhou Y, et al. miRNA Profiling of Extracellular Vesicles Reveals Biomarkers for Psoriasis. J Invest Dermatol. 2021;141(1):185–189.e184. doi: 10.1016/j.jid.2020.04.021 [DOI] [PubMed] [Google Scholar]
  • 40.Hu S, Li Z, Lutz H, et al. Dermal exosomes containing miR-218-5p promote hair regeneration by regulating β-catenin signaling. Sci Adv. 2020;6(30):eaba1685. doi: 10.1126/sciadv.aba1685 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Momen-Heravi F, Getting SJ, Moschos SA. Extracellular vesicles and their nucleic acids for biomarker discovery. Pharmacol Ther. 2018;192:170–187. doi: 10.1016/j.pharmthera.2018.08.002 [DOI] [PubMed] [Google Scholar]
  • 42.Koynova R, Tenchov B, MacDonald RC. Nonlamellar Phases in Cationic Phospholipids, Relevance to Drug and Gene Delivery. ACS Biomater Sci Eng. 2015;1(3):130–138. doi: 10.1021/ab500142w [DOI] [PubMed] [Google Scholar]
  • 43.Keshavarz Azizi Raftar S, Ashrafian F, Yadegar A, et al. The protective effects of live and pasteurized Akkermansia muciniphila and its extracellular vesicles against HFD/CCl4-induced liver injury. Microbiol Spectr. 2021;9(2):e0048421. doi: 10.1128/Spectrum.00484-21 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Mathieu M, Martin-Jaular L, Lavieu G, Théry C. Specificities of secretion and uptake of exosomes and other extracellular vesicles for cell-to-cell communication. Nat Cell Biol. 2019;21(1):9–17. doi: 10.1038/s41556-018-0250-9 [DOI] [PubMed] [Google Scholar]
  • 45.Kimiz-Gebologlu I, Oncel SS. Exosomes: large-scale production, isolation, drug loading efficiency, and biodistribution and uptake. J Control Release. 2022;347:533–543. doi: 10.1016/j.jconrel.2022.05.027 [DOI] [PubMed] [Google Scholar]
  • 46.Gurung S, Perocheau D, Touramanidou L, Baruteau J. The exosome journey: from biogenesis to uptake and intracellular signalling. Cell Commun Signal. 2021;19(1):47. doi: 10.1186/s12964-021-00730-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Cao Z, Li P, Zhang M, et al. Erythrocyte Extracellular Vesicles Amalgamate into the Hair and Skin to Maintain Homeostasis. J Extracell Vesicles. 2025;14(5):e70080. doi: 10.1002/jev2.70080 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lou P, Liu S, Wang Y, et al. Neonatal-Tissue-Derived Extracellular Vesicle Therapy (NEXT): a Potent Strategy for Precision Regenerative Medicine. Adv Mater. 2023;35(33):e2300602. doi: 10.1002/adma.202300602 [DOI] [PubMed] [Google Scholar]
  • 49.Xiong M, Zhang Q, Hu W, et al. The novel mechanisms and applications of exosomes in dermatology and cutaneous medical aesthetics. Pharmacol Res. 2021;166:105490. doi: 10.1016/j.phrs.2021.105490 [DOI] [PubMed] [Google Scholar]
  • 50.Cheng S, Xi Z, Chen G, et al. Extracellular vesicle-carried microRNA-27b derived from mesenchymal stem cells accelerates cutaneous wound healing via E3 ubiquitin ligase ITCH. J Cell Mol Med. 2020;24(19):11254–11271. doi: 10.1111/jcmm.15692 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Liu Y, Zhang M, Liao Y, et al. Human umbilical cord mesenchymal stem cell-derived exosomes promote murine skin wound healing by neutrophil and macrophage modulations revealed by single-cell RNA sequencing. Front Immunol. 2023;14:1142088. doi: 10.3389/fimmu.2023.1142088 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Li X, Liu L, Yang J, et al. Exosome Derived From Human Umbilical Cord Mesenchymal Stem Cell Mediates MiR-181c Attenuating Burn-induced Excessive Inflammation. EBioMedicine. 2016;8:72–82. doi: 10.1016/j.ebiom.2016.04.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Zhang B, Wu X, Zhang X, et al. Human umbilical cord mesenchymal stem cell exosomes enhance angiogenesis through the Wnt4/β-catenin pathway. Stem Cells Transl Med. 2015;4(5):513–522. doi: 10.5966/sctm.2014-0267 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Hu Y, Rao SS, Wang ZX, et al. Exosomes from human umbilical cord blood accelerate cutaneous wound healing through miR-21-3p-mediated promotion of angiogenesis and fibroblast function. Theranostics. 2018;8(1):169–184. doi: 10.7150/thno.21234 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.He X, Dong Z, Cao Y, et al. MSC-Derived Exosome Promotes M2 Polarization and Enhances Cutaneous Wound Healing. Stem Cells Int. 2019;2019:7132708. doi: 10.1155/2019/7132708 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Yang J, Zhang X, Wang G, et al. ApoSEVs-Mediated Modulation of Versatile Target Cells Promotes Diabetic Wound Healing: unveiling a Promising Strategy. Int J Nanomed. 2023;18:6955–6977. doi: 10.2147/ijn.S436350 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Xiao X, Xu M, Yu H, et al. Mesenchymal stem cell-derived small extracellular vesicles mitigate oxidative stress-induced senescence in endothelial cells via regulation of miR-146a/Src. Signal Transduct Target Ther. 2021;6(1):354. doi: 10.1038/s41392-021-00765-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Zhang C, Jiang T, Jiang G, et al. White adipose tissue-derived small extracellular vesicles: a new potential therapeutic reagent for accelerating diabetic wound healing. FASEB j. 2023;37(12):e23314. doi: 10.1096/fj.202301549R [DOI] [PubMed] [Google Scholar]
  • 59.Wei F, Wang A, Wang Q, et al. Plasma endothelial cells-derived extracellular vesicles promote wound healing in diabetes through YAP and the PI3K/Akt/mTOR pathway. Aging. 2020;12(12):12002–12018. doi: 10.18632/aging.103366 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wang P, Theocharidis G, Vlachos IS, et al. Exosomes Derived from Epidermal Stem Cells Improve Diabetic Wound Healing. J Invest Dermatol. 2022;142(9):2508–2517.e2513. doi: 10.1016/j.jid.2022.01.030 [DOI] [PubMed] [Google Scholar]
  • 61.Xia W, Li M, Jiang X, et al. Young fibroblast-derived exosomal microRNA-125b transfers beneficial effects on aged cutaneous wound healing. J Nanobiotechnology. 2022;20(1):144. doi: 10.1186/s12951-022-01348-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Han F, Wang K, Shen K, et al. Extracellular vesicles from Lactobacillus druckerii inhibit hypertrophic scar fibrosis. J Nanobiotechnology. 2023;21(1):113. doi: 10.1186/s12951-023-01861-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Tan S, Liu Z, Cong M, et al. Dandelion-derived vesicles-laden hydrogel dressings capable of neutralizing Staphylococcus aureus exotoxins for the care of invasive wounds. J Control Release. 2024;368:355–371. doi: 10.1016/j.jconrel.2024.02.045 [DOI] [PubMed] [Google Scholar]
  • 64.Zhou L, Wang H, Jing J, et al. Regulation of hair follicle development by exosomes derived from dermal papilla cells. Biochem Biophys Res Commun. 2018;500(2):325–332. doi: 10.1016/j.bbrc.2018.04.067 [DOI] [PubMed] [Google Scholar]
  • 65.Li J, Zhao B, Yao S, et al. Dermal PapillaCell-Derived Exosomes Regulate Hair Follicle Stem Cell Proliferation via LEF1. Int J Mol Sci. 2023;24(4). doi: 10.3390/ijms24043961 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Chen Y, Huang J, Liu Z, et al. miR-140-5p in Small Extracellular Vesicles From Human Papilla Cells Stimulates Hair Growth by Promoting Proliferation of Outer Root Sheath and Hair Matrix Cells. Front Cell Dev Biol. 2020;8:593638. doi: 10.3389/fcell.2020.593638 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Rajendran RL, Gangadaran P, Kwack MH, et al. Human fibroblast-derived extracellular vesicles promote hair growth in cultured human hair follicles. FEBS Lett. 2021;595(7):942–953. doi: 10.1002/1873-3468.14050 [DOI] [PubMed] [Google Scholar]
  • 68.Rajendran RL, Gangadaran P, Seo CH, et al. Macrophage-Derived Extracellular Vesicle Promotes Hair Growth. Cells. 2020;9(4). doi: 10.3390/cells9040856 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Rajendran RL, Gangadaran P, Kwack MH, et al. Application of extracellular vesicles from mesenchymal stem cells promotes hair growth by regulating human dermal cells and follicles. World J Stem Cells. 2022;14(7):527–538. doi: 10.4252/wjsc.v14.i7.527 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Ma L, Chen C, Liu D, et al. Apoptotic extracellular vesicles are metabolized regulators nurturing the skin and hair. Bioact Mater. 2023;19:626–641. doi: 10.1016/j.bioactmat.2022.04.022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Liang JX, Liao X, Li SH, et al. Antiaging Properties of Exosomes from Adipose-Derived Mesenchymal Stem Cells in Photoaged Rat Skin. Biomed Res Int. 2020;2020:6406395. doi: 10.1155/2020/6406395 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Yan T, Huang L, Yan Y, et al. Bone marrow mesenchymal stem cell-derived exosome miR-29b-3p alleviates UV irradiation-induced photoaging in skin fibroblast. Photodermatol Photoimmunol Photomed. 2023;39(3):235–245. doi: 10.1111/phpp.12827 [DOI] [PubMed] [Google Scholar]
  • 73.Hu S, Li Z, Cores J, et al. Needle-Free Injection of Exosomes Derived from Human Dermal Fibroblast Spheroids Ameliorates Skin Photoaging. ACS Nano. 2019;13(10):11273–11282. doi: 10.1021/acsnano.9b04384 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Cho BS, Kim JO, Ha DH, Yi YW. Exosomes derived from human adipose tissue-derived mesenchymal stem cells alleviate atopic dermatitis. Stem Cell Res Ther. 2018;9(1):187. doi: 10.1186/s13287-018-0939-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Shin KO, Ha DH, Kim JO, et al. Exosomes from human adipose tissue-derived mesenchymal stem cells promote epidermal barrier repair by inducing de novo synthesis of ceramides in atopic dermatitis. Cells. 2020;9(3). doi: 10.3390/cells9030680 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Kim J, Lee SK, Jung M, et al. Extracellular vesicles from IFN-γ-primed mesenchymal stem cells repress atopic dermatitis in mice. J Nanobiotechnology. 2022;20(1):526. doi: 10.1186/s12951-022-01728-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Yoon J, Lee SK, Park A, et al. Exosome from IFN-γ-Primed Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells Improved Skin Inflammation and Barrier Function. Int J Mol Sci. 2023;24(14). doi: 10.3390/ijms241411635 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Kim MH, Choi SJ, Choi HI, et al. Lactobacillus plantarum-derived Extracellular Vesicles Protect Atopic Dermatitis Induced by Staphylococcus aureus-derived Extracellular Vesicles. Allergy Asthma Immunol Res. 2018;10(5):516–532. doi: 10.4168/aair.2018.10.5.516 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Huang R, Jia B, Su D, et al. Plant exosomes fused with engineered mesenchymal stem cell-derived nanovesicles for synergistic therapy of autoimmune skin disorders. J Extracell Vesicles. 2023;12(10):e12361. doi: 10.1002/jev2.12361 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Zhao C, Wang D, Wang X, et al. Down-regulation of exosomal miR-200c derived from keratinocytes in vitiligo lesions suppresses melanogenesis. J Cell Mol Med. 2020;24(20):12164–12175. doi: 10.1111/jcmm.15864 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Zhang Y, Yan J, Li Z, Zheng J, Sun Q. Exosomes Derived from Human Umbilical Cord Mesenchymal Stem Cells Alleviate Psoriasis-like Skin Inflammation. J Interferon Cytokine Res. 2022;42(1):8–18. doi: 10.1089/jir.2021.0146 [DOI] [PubMed] [Google Scholar]
  • 82.Zhang W, Lin J, Shi P, et al. Small Extracellular Vesicles Derived From MSCs Have Immunomodulatory Effects to Enhance Delivery of ASO-210 for Psoriasis Treatment. Front Cell Dev Biol. 2022;10:842813. doi: 10.3389/fcell.2022.842813 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Zhang B, Lai RC, Sim WK, et al. Topical Application of Mesenchymal Stem Cell Exosomes Alleviates the Imiquimod Induced Psoriasis-Like Inflammation. Int J Mol Sci. 2021;22(2). doi: 10.3390/ijms22020720 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ye Z, Liang Y, Lin B, et al. Gingiva-Derived Mesenchymal Stem Cells Attenuate Imiquimod- (IMQ-) Induced Murine Psoriasis-Like Skin Inflammation. Stem Cells Int. 2022;2022:6544514. doi: 10.1155/2022/6544514 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Xu Y, Gan Y, Qi F, et al. Innate lymphoid cell-based immunomodulatory hydrogel microspheres containing Cutibacterium acnes extracellular vesicles for the treatment of psoriasis. Acta Biomater. 2024;184:296–312. doi: 10.1016/j.actbio.2024.06.006 [DOI] [PubMed] [Google Scholar]
  • 86.Gómez-Chávez F, Cedillo-Peláez C, Zapi-Colín LA, et al. The Extracellular Vesicles from the Commensal Staphylococcus Epidermidis ATCC12228 Strain Regulate Skin Inflammation in the Imiquimod-Induced Psoriasis Murine Model. Int J Mol Sci. 2021;22(23). doi: 10.3390/ijms222313029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Su D, Li M, Xie Y, et al. Gut commensal bacteria Parabacteroides goldsteinii-derived outer membrane vesicles suppress skin inflammation in psoriasis. J Control Release. 2024;377:127–145. doi: 10.1016/j.jconrel.2024.11.014 [DOI] [PubMed] [Google Scholar]
  • 88.Xu F, Fei Z, Dai H, et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles with High PD-L1 Expression for Autoimmune Diseases Treatment. Adv Mater. 2022;34(1):e2106265. doi: 10.1002/adma.202106265 [DOI] [PubMed] [Google Scholar]
  • 89.Ma F, Liu X, Zhang Y, et al. Tumor extracellular vesicle-derived PD-L1 promotes T cell senescence through lipid metabolism reprogramming. Sci Transl Med. 2025;17(785):eadm7269. doi: 10.1126/scitranslmed.adm7269 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Leary N, Walser S, He Y, et al. Melanoma-derived extracellular vesicles mediate lymphatic remodelling and impair tumour immunity in draining lymph nodes. J Extracell Vesicles. 2022;11(2):e12197. doi: 10.1002/jev2.12197 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Cao M, Yan H, Han X, et al. Ginseng-derived nanoparticles alter macrophage polarization to inhibit melanoma growth. J Immunother Cancer. 2019;7(1):326. doi: 10.1186/s40425-019-0817-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Zhu L, Kalimuthu S, Gangadaran P, et al. Exosomes Derived From Natural Killer Cells Exert Therapeutic Effect in Melanoma. Theranostics. 2017;7(10):2732–2745. doi: 10.7150/thno.18752 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Gu Y, Du Y, Jiang L, et al. αvβ3 integrin-specific exosomes engineered with cyclopeptide for targeted delivery of triptolide against malignant melanoma. J Nanobiotechnology. 2022;20(1):384. doi: 10.1186/s12951-022-01597-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Chang J, Tran DC, Zhu GA, et al. Initial in vitro functional characterization of serum exosomal microRNAs from patients with metastatic basal cell carcinoma. Br J Dermatol. 2017;177(5):e187–e190. doi: 10.1111/bjd.15508 [DOI] [PubMed] [Google Scholar]
  • 95.Yin J, Pei Z, Wu C, et al. M2 Macrophage-Derived Exosomal circ_0088494 Inhibits Ferroptosis via Promoting H3K4me1 Modification of STEAP3 in Cutaneous Squamous Cell Carcinoma. Mol, Carcinog. 2025;64(3):513–525. doi: 10.1002/mc.23862 [DOI] [PubMed] [Google Scholar]
  • 96.Falanga V, Isseroff RR, Soulika AM, et al. Chronic wounds. Nat Rev Dis Primers. 2022;8(1):50. doi: 10.1038/s41572-022-00377-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Martin P, Nunan R. Cellular and molecular mechanisms of repair in acute and chronic wound healing. Br J Dermatol. 2015;173(2):370–378. doi: 10.1111/bjd.13954 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Freedman BR, Hwang C, Talbot S, et al. Breakthrough treatments for accelerated wound healing. Sci Adv. 2023;9(20):eade7007. doi: 10.1126/sciadv.ade7007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Peña OA, Martin P. Cellular and molecular mechanisms of skin wound healing. Nat Rev Mol Cell Biol. 2024;25(8):599–616. doi: 10.1038/s41580-024-00715-1 [DOI] [PubMed] [Google Scholar]
  • 100.Wu T, Liu Y, Wang S, Shi C. MSC-Derived Extracellular Vesicles: roles and Molecular Mechanisms for Tissue Repair. Int J Nanomed. 2025;20:7953–7974. doi: 10.2147/ijn.S525394 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Al-Rashed F, Sindhu S, Arefanian H, et al. Repetitive Intermittent Hyperglycemia Drives the M1 Polarization and Inflammatory Responses in THP-1 Macrophages Through the Mechanism Involving the TLR4-IRF5 Pathway. Cells. 2020;9(8). doi: 10.3390/cells9081892 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Pomatto M, Gai C, Negro F, et al. Differential Therapeutic Effect of Extracellular Vesicles Derived by Bone Marrow and Adipose Mesenchymal Stem Cells on Wound Healing of Diabetic Ulcers and Correlation to Their Cargoes. Int J Mol Sci. 2021;22(8). doi: 10.3390/ijms22083851 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Song Y, You Y, Xu X, et al. Adipose-derived mesenchymal stem cell-derived exosomes biopotentiated extracellular matrix hydrogels accelerate diabetic wound healing and skin regeneration. Adv Sci. 2023;10(30):e2304023. doi: 10.1002/advs.202304023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Johnson J, Law SQK, Shojaee M, et al. First-in-human clinical trial of allogeneic, platelet-derived extracellular vesicles as a potential therapeutic for delayed wound healing. J Extracell Vesicles. 2023;12(7):e12332. doi: 10.1002/jev2.12332 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Gentile P, Garcovich S. Advances in Regenerative Stem Cell Therapy in Androgenic Alopecia and Hair Loss: wnt pathway, Growth-Factor, and Mesenchymal Stem Cell Signaling Impact Analysis on Cell Growth and Hair Follicle Development. Cells. 2019;8(5). doi: 10.3390/cells8050466 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Choi BY. Hair-Growth Potential of Ginseng and Its Major Metabolites: a Review on Its Molecular Mechanisms. Int J Mol Sci. 2018;19(9). doi: 10.3390/ijms19092703 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Stenn KS, Paus R. Controls of hair follicle cycling. Physiol Rev. 2001;81(1):449–494. doi: 10.1152/physrev.2001.81.1.449 [DOI] [PubMed] [Google Scholar]
  • 108.Ji S, Zhu Z, Sun X, Fu X. Functional hair follicle regeneration: an updated review. Signal Transduct Target Ther. 2021;6(1):66. doi: 10.1038/s41392-020-00441-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Müller-Röver S, Handjiski B, van der Veen C, et al. A comprehensive guide for the accurate classification of murine hair follicles in distinct hair cycle stages. J Invest Dermatol. 2001;117(1):3–15. doi: 10.1046/j.0022-202x.2001.01377.x [DOI] [PubMed] [Google Scholar]
  • 110.Zhang B, Chen T. Local and systemic mechanisms that control the hair follicle stem cell niche. Nat Rev Mol Cell Biol. 2024;25(2):87–100. doi: 10.1038/s41580-023-00662-3 [DOI] [PubMed] [Google Scholar]
  • 111.Plikus MV, Baker RE, Chen CC, et al. Self-organizing and stochastic behaviors during the regeneration of hair stem cells. Science. 2011;332(6029):586–589. doi: 10.1126/science.1201647 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Wolff H, Fischer TW, Blume-Peytavi U. The Diagnosis and Treatment of Hair and Scalp Diseases. Dtsch Arztebl Int. 2016;113(21):377–386. doi: 10.3238/arztebl.2016.0377 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Adil A, Godwin M. The effectiveness of treatments for androgenetic alopecia: a systematic review and meta-analysis. J Am Acad Dermatol. 2017;77(1):136–141.e135. doi: 10.1016/j.jaad.2017.02.054 [DOI] [PubMed] [Google Scholar]
  • 114.Gangadaran P, Rajendran RL, Kwack MH, et al. Application of Cell-Derived Extracellular Vesicles and Engineered Nanovesicles for Hair Growth: from Mechanisms to Therapeutics. Front Cell Dev Biol. 2022;10:963278. doi: 10.3389/fcell.2022.963278 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Diener C, Keller A, Meese E. Emerging concepts of miRNA therapeutics: from cells to clinic. Trends Genet. 2022;38(6):613–626. doi: 10.1016/j.tig.2022.02.006 [DOI] [PubMed] [Google Scholar]
  • 116.Sun Z, Shi K, Yang S, et al. Effect of exosomal miRNA on cancer biology and clinical applications. Mol Cancer. 2018;17(1):147. doi: 10.1186/s12943-018-0897-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Wang Y, Shen K, Sun Y, et al. Extracellular vesicles from 3D cultured dermal papilla cells improve wound healing via Krüppel-like factor 4/vascular endothelial growth factor A -driven angiogenesis. Burns Trauma. 2023;11:tkad034. doi: 10.1093/burnst/tkad034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Zhou G, Li R, Sheng S, et al. Organoids and organoid extracellular vesicles-based disease treatment strategies. J Nanobiotechnology. 2024;22(1):679. doi: 10.1186/s12951-024-02917-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Chen Y, Huang J, Chen R, et al. Sustained release of dermal papilla-derived extracellular vesicles from injectable microgel promotes hair growth. Theranostics. 2020;10(3):1454–1478. doi: 10.7150/thno.39566 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Kroemer G, Maier AB, Cuervo AM, et al. From geroscience to precision geromedicine: understanding and managing aging. Cell. 2025;188(8):2043–2062. doi: 10.1016/j.cell.2025.03.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Campisi J, Kapahi P, Lithgow GJ, et al. From discoveries in ageing research to therapeutics for healthy ageing. Nature. 2019;571(7764):183–192. doi: 10.1038/s41586-019-1365-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.de Magalhães JP. Distinguishing between driver and passenger mechanisms of aging. Nat Genet. 2024;56(2):204–211. doi: 10.1038/s41588-023-01627-0 [DOI] [PubMed] [Google Scholar]
  • 123.de Magalhães JP. Cellular senescence in normal physiology. Science. 2024;384(6702):1300–1301. doi: 10.1126/science.adj7050 [DOI] [PubMed] [Google Scholar]
  • 124.Böhm M, Stegemann A, Paus R, et al. Endocrine Controls of Skin Aging. Endocr Rev. 2025;46(3):349–375. doi: 10.1210/endrev/bnae034 [DOI] [PubMed] [Google Scholar]
  • 125.Draelos Z, Bogdanowicz P, Saurat J-H. Top weapons in skin aging and actives to target the consequences of skin cell senescence. J Eur Acad Dermatol Venereol. 2024;38(S4):15–22. doi: 10.1111/jdv.19648 [DOI] [PubMed] [Google Scholar]
  • 126.Griffiths TW, Watson REB, Langton AK. Skin ageing and topical rejuvenation strategies. Br J Dermatol. 2023;189(Suppl Supplement_1):i17–i23. doi: 10.1093/bjd/ljad282 [DOI] [PubMed] [Google Scholar]
  • 127.Jin S, Li K, Zong X, et al. Hallmarks of Skin Aging: update. Aging Dis. 2023;14(6):2167–2176. doi: 10.14336/ad.2023.0321 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Choi EJ, Kil IS, Cho EG. Extracellular Vesicles Derived from Senescent Fibroblasts Attenuate the Dermal Effect on Keratinocyte Differentiation. Int J Mol Sci. 2020;21(3). doi: 10.3390/ijms21031022 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Terlecki-Zaniewicz L, Lämmermann I, Latreille J, et al. Small extracellular vesicles and their miRNA cargo are anti-apoptotic members of the senescence-associated secretory phenotype. Aging. 2018;10(5):1103–1132. doi: 10.18632/aging.101452 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Terlecki-Zaniewicz L, Pils V, Bobbili MR, et al. Extracellular Vesicles in Human Skin: cross-Talk from Senescent Fibroblasts to Keratinocytes by miRNAs. J Invest Dermatol. 2019;139(12):2425–2436.e2425. doi: 10.1016/j.jid.2019.05.015 [DOI] [PubMed] [Google Scholar]
  • 131.Borghesan M, Fafián-Labora J, Eleftheriadou O, et al. Small Extracellular Vesicles Are Key Regulators of Non-cell Autonomous Intercellular Communication in Senescence via the Interferon Protein IFITM3. Cell Rep. 2019;27(13):3956–3971.e3956. doi: 10.1016/j.celrep.2019.05.095 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Nedachi T, Bonod C, Rorteau J, et al. Chronological aging impacts abundance, function and microRNA content of extracellular vesicles produced by human epidermal keratinocytes. Aging. 2023;15(22):12702–12722. doi: 10.18632/aging.205245 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Chevalier FP, Rorteau J, Ferraro S, et al. MiR-30a-5p Alters Epidermal Terminal Differentiation during Aging by Regulating BNIP3L/NIX-Dependent Mitophagy. Cells. 2022;11(5). doi: 10.3390/cells11050836 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Yoon H, Jo J, Hyun H, et al. Extracellular vesicle as therapeutic agents in anti-aging: mechanistic insights and future potential. J Control Release. 2025;383:113796. doi: 10.1016/j.jconrel.2025.113796 [DOI] [PubMed] [Google Scholar]
  • 135.Ni X, Xu Y, Wang W, et al. IL-17D-induced inhibition of DDX5 expression in keratinocytes amplifies IL-36R-mediated skin inflammation. Nat Immunol. 2022;23(11):1577–1587. doi: 10.1038/s41590-022-01339-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Liu T, Li S, Ying S, et al. The IL-23/IL-17 Pathway in Inflammatory Skin Diseases: from Bench to Bedside. Front Immunol. 2020;11:594735. doi: 10.3389/fimmu.2020.594735 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Langan SM, Irvine AD, Weidinger S. Atopic dermatitis. Lancet. 2020;396(10247):345–360. doi: 10.1016/s0140-6736(20)31286-1 [DOI] [PubMed] [Google Scholar]
  • 138.Ständer S. Atopic Dermatitis. N Engl J Med. 2021;384(12):1136–1143. doi: 10.1056/NEJMra2023911 [DOI] [PubMed] [Google Scholar]
  • 139.Schuler CFT, Billi AC, Maverakis E, Tsoi LC, Gudjonsson JE. Novel insights into atopic dermatitis. J Allergy Clin Immunol. 2023;151(5):1145–1154. doi: 10.1016/j.jaci.2022.10.023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Ha DH, Kim HK, Lee J, et al. Mesenchymal Stem/Stromal Cell-Derived Exosomes for Immunomodulatory Therapeutics and Skin Regeneration. Cells. 2020;9(5). doi: 10.3390/cells9051157 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Frisoli ML, Essien K, Harris JE. Vitiligo: mechanisms of Pathogenesis and Treatment. Annu Rev Immunol. 2020;38:621–648. doi: 10.1146/annurev-immunol-100919-023531 [DOI] [PubMed] [Google Scholar]
  • 142.Mukhatayev Z, Le Poole IC. Vitiligo: advances in pathophysiology research and treatment development. Trends Mol Med. 2024;30(9):844–862. doi: 10.1016/j.molmed.2024.04.009 [DOI] [PubMed] [Google Scholar]
  • 143.Xu Z, Chen D, Hu Y, et al. Anatomically distinct fibroblast subsets determine skin autoimmune patterns. Nature. 2022;601(7891):118–124. doi: 10.1038/s41586-021-04221-8 [DOI] [PubMed] [Google Scholar]
  • 144.Kaur M, Bagga PK, Kaur T, Kataria AS. Evaluation of Histologically and Histochemically Proven Cases of Vitiligo and its Correlation with CD4+ and CD8+ Lymphocyte Counts using Flow Cytometry. J Clin Diagn Res. 2017;11(5):Ec09–ec12. doi: 10.7860/jcdr/2017/25665.9821 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Rodrigues M, Ezzedine K, Hamzavi I, Pandya AG, Harris JE. New discoveries in the pathogenesis and classification of vitiligo. J Am Acad Dermatol. 2017;77(1):1–13. doi: 10.1016/j.jaad.2016.10.048 [DOI] [PubMed] [Google Scholar]
  • 146.Zhang Q, Cui T, Chang Y, et al. HO-1 regulates the function of Treg: association with the immune intolerance in vitiligo. J Cell Mol Med. 2018;22(9):4335–4343. doi: 10.1111/jcmm.13723 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Wong PM, Yang L, Yang L, et al. New insight into the role of exosomes in vitiligo. Autoimmun Rev. 2020;19(11):102664. doi: 10.1016/j.autrev.2020.102664 [DOI] [PubMed] [Google Scholar]
  • 148.Li W, Pang Y, He Q, et al. Exosome-derived microRNAs: emerging players in vitiligo. Front Immunol. 2024;15:1419660. doi: 10.3389/fimmu.2024.1419660 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149.Li D, Zhou T, She Q, et al. Circulating Exosomal miR-493-3p Affects Melanocyte Survival and Function by Regulating Epidermal Dopamine Concentration in Segmental Vitiligo. J Invest Dermatol. 2022;142(12):3262–3273.e3211. doi: 10.1016/j.jid.2022.05.1086 [DOI] [PubMed] [Google Scholar]
  • 150.Zhang C, Guo W, Wang S, Di Y, Wu D. Peripheral Blood of Vitiligo Patients-Derived Exosomal MiR-21-5p Inhibits Melanocytes Melanogenesis via Targeting SATB1. Iran J Public Health. 2022;51(12):2706–2716. doi: 10.18502/ijph.v51i12.11461 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Shi HX, Zhang RZ, Xiao L, Wang L. Effects of keratinocyte-derived and fibroblast-derived exosomes on human epidermal melanocytes. Indian J Dermatol Venereol Leprol. 2022;88(3):322–331. doi: 10.25259/ijdvl_1087_19 [DOI] [PubMed] [Google Scholar]
  • 152.Boehncke WH, Schön MP. Psoriasis. Lancet. 2015;386(9997):983–994. doi: 10.1016/s0140-6736(14)61909-7 [DOI] [PubMed] [Google Scholar]
  • 153.Rendon A, Schäkel K. Psoriasis Pathogenesis and Treatment. Int J Mol Sci. 2019;20(6). doi: 10.3390/ijms20061475 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Jiang X, Jiang Z, Huang S, et al. Ultraviolet B radiation-induced JPH203-loaded keratinocyte extracellular vesicles exert etiological interventions for psoriasis therapy. J Control Release. 2023;362:468–478. doi: 10.1016/j.jconrel.2023.08.059 [DOI] [PubMed] [Google Scholar]
  • 155.Zhou X, Tang B, Huang Q, et al. Engineered Mesenchymal Stem Cell-Derived Extracellular Vesicles Scavenge Self-Antigens for Psoriasis Therapy via Modulating Metabolic and Immunological Disorders. Adv Sci. 2024:e2410067. doi: 10.1002/advs.202410067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Sun C, Mezzadra R, Schumacher TN. Regulation and Function of the PD-L1 Checkpoint. Immunity. 2018;48(3):434–452. doi: 10.1016/j.immuni.2018.03.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Long GV, Swetter SM, Menzies AM, Gershenwald JE, Scolyer RA. Cutaneous melanoma. Lancet. 2023;402(10400):485–502. doi: 10.1016/s0140-6736(23)00821-8 [DOI] [PubMed] [Google Scholar]
  • 158.Merkel EA, Mohan LS, Shi K, et al. Paediatric melanoma: clinical update, genetic basis, and advances in diagnosis. Lancet Child Adolesc Health. 2019;3(9):646–654. doi: 10.1016/s2352-4642(19)30116-6 [DOI] [PubMed] [Google Scholar]
  • 159.Shain AH, Bastian BC. From melanocytes to melanomas. Nat Rev Cancer. 2016;16(6):345–358. doi: 10.1038/nrc.2016.37 [DOI] [PubMed] [Google Scholar]
  • 160.Marar C, Starich B, Wirtz D. Extracellular vesicles in immunomodulation and tumor progression. Nat Immunol. 2021;22(5):560–570. doi: 10.1038/s41590-021-00899-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Dou X, Feng C, Li J, Jiang E, Shang Z. Extracellular vesicle-mediated crosstalk in tumor microenvironment dominates tumor fate. Trends Cell Biol. 2025;35(3):230–247. doi: 10.1016/j.tcb.2024.08.008 [DOI] [PubMed] [Google Scholar]
  • 162.Xiong J, Xue Y, Xia Y, Zhao J, Wang Y. Identification of key microRNAs of plasma extracellular vesicles and their diagnostic and prognostic significance in melanoma. Open Med. 2020;15(1):464–482. doi: 10.1515/med-2020-0111 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Gerloff D, Kewitz-Hempel S, Hause G, et al. Comprehensive Analyses of miRNAs Revealed miR-92b-3p, miR-182-5p and miR-183-5p as Potential Novel Biomarkers in Melanoma-Derived Extracellular Vesicles. Front Oncol. 2022;12:935816. doi: 10.3389/fonc.2022.935816 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Tengda L, Shuping L, Mingli G, et al. Serum exosomal microRNAs as potent circulating biomarkers for melanoma. Melanoma Res. 2018;28(4):295–303. doi: 10.1097/cmr.0000000000000450 [DOI] [PubMed] [Google Scholar]
  • 165.Pfeffer SR, Grossmann KF, Cassidy PB, et al. Detection of Exosomal miRNAs in the Plasma of Melanoma Patients. J Clin Med. 2015;4(12):2012–2027. doi: 10.3390/jcm4121957 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.García-Silva S, Benito-Martín A, Nogués L, et al. Melanoma-derived small extracellular vesicles induce lymphangiogenesis and metastasis through an NGFR-dependent mechanism. Nat Cancer. 2021;2(12):1387–1405. doi: 10.1038/s43018-021-00272-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Chen G, Huang AC, Zhang W, et al. Exosomal PD-L1 contributes to immunosuppression and is associated with anti-PD-1 response. Nature. 2018;560(7718):382–386. doi: 10.1038/s41586-018-0392-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Zhang Z, Guo H, Yang W, Li J. Exosomal Circular RNA RNA-seq Profiling and the Carcinogenic Role of Exosomal circ-CYP24A1 in Cutaneous Squamous Cell Carcinoma. Front Med Lausanne. 2021;8:675842. doi: 10.3389/fmed.2021.675842 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Sharma P, Diergaarde B, Ferrone S, Kirkwood JM, Whiteside TL. Melanoma cell-derived exosomes in plasma of melanoma patients suppress functions of immune effector cells. Sci Rep. 2020;10(1):92. doi: 10.1038/s41598-019-56542-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 170.Carpi S, Polini B, Fogli S, et al. Circulating microRNAs as biomarkers for early diagnosis of cutaneous melanoma. Expert Rev Mol Diagn. 2020;20(1):19–30. doi: 10.1080/14737159.2020.1696194 [DOI] [PubMed] [Google Scholar]
  • 171.Mirzaei H, Gholamin S, Shahidsales S, et al. MicroRNAs as potential diagnostic and prognostic biomarkers in melanoma. Eur J Cancer. 2016;53:25–32. doi: 10.1016/j.ejca.2015.10.009 [DOI] [PubMed] [Google Scholar]
  • 172.Serratì S, Guida M, Di Fonte R, et al. Circulating extracellular vesicles expressing PD1 and PD-L1 predict response and mediate resistance to checkpoint inhibitors immunotherapy in metastatic melanoma. Mol Cancer. 2022;21(1):20. doi: 10.1186/s12943-021-01490-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Cordonnier M, Nardin C, Chanteloup G, et al. Tracking the evolution of circulating exosomal-PD-L1 to monitor melanoma patients. J Extracell Vesicles. 2020;9(1):1710899. doi: 10.1080/20013078.2019.1710899 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 174.Xiang X, Wang J, Lu D, Xu X. Targeting tumor-associated macrophages to synergize tumor immunotherapy. Signal Transduct Target Ther. 2021;6(1):75. doi: 10.1038/s41392-021-00484-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Wang Y, Xiang Y, Xin VW, et al. Dendritic cell biology and its role in tumor immunotherapy. J Hematol Oncol. 2020;13(1):107. doi: 10.1186/s13045-020-00939-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 176.Wang R, Zhu T, Hou B, Huang X. An iPSC-derived exosome-pulsed dendritic cell vaccine boosts antitumor immunity in melanoma. Mol Ther. 2023;31(8):2376–2390. doi: 10.1016/j.ymthe.2023.06.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 177.Babaei S, Fadaee M, Abbasi-Kenarsari H, Shanehbandi D, Kazemi T. Exosome-based immunotherapy as an innovative therapeutic approach in melanoma. Cell Commun Signal. 2024;22(1):527. doi: 10.1186/s12964-024-01906-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 178.Heil F, Hemmi H, Hochrein H, et al. Species-specific recognition of single-stranded RNA via toll-like receptor 7 and 8. Science. 2004;303(5663):1526–1529. doi: 10.1126/science.1093620 [DOI] [PubMed] [Google Scholar]
  • 179.Bhatta R, Han J, Liu Y, et al. Metabolic tagging of extracellular vesicles and development of enhanced extracellular vesicle based cancer vaccines. Nat Commun. 2023;14(1):8047. doi: 10.1038/s41467-023-43914-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Perez M, Abisaad JA, Rojas KD, Marchetti MA, Jaimes N. Skin cancer: primary, secondary, and tertiary prevention. Part I. J Am Acad Dermatol. 2022;87(2):255–268. doi: 10.1016/j.jaad.2021.12.066 [DOI] [PubMed] [Google Scholar]
  • 181.Reddy P, Yao M, Patel M. Investigative Landscape in Advanced Non-Melanoma Skin Cancers. Curr Treat Options Oncol. 2021;22(7):56. doi: 10.1007/s11864-021-00853-0 [DOI] [PubMed] [Google Scholar]
  • 182.Krakowski AC, Hafeez F, Westheim A, Pan EY, Wilson M. Advanced basal cell carcinoma: what dermatologists need to know about diagnosis. J Am Acad Dermatol. 2022;86(6s):S1–s13. doi: 10.1016/j.jaad.2022.03.023 [DOI] [PubMed] [Google Scholar]
  • 183.Wilson M, Johnson RP, Senft SC, Pan EY, Krakowski AC. Advanced basal cell carcinoma: what dermatologists need to know about treatment. J Am Acad Dermatol. 2022;86(6s):S14–s24. doi: 10.1016/j.jaad.2022.03.022 [DOI] [PubMed] [Google Scholar]
  • 184.Chang MS, Azin M, Demehri S. Cutaneous Squamous Cell Carcinoma: the Frontier of Cancer Immunoprevention. Annu Rev Pathol. 2022;17:101–119. doi: 10.1146/annurev-pathol-042320-120056 [DOI] [PubMed] [Google Scholar]
  • 185.Flemming JP, Hill BL, Haque MW, et al. miRNA- and cytokine-associated extracellular vesicles mediate squamous cell carcinomas. J Extracell Vesicles. 2020;9(1):1790159. doi: 10.1080/20013078.2020.1790159 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 186.Mu Y, Lian C, Chen X, et al. Cutaneous squamous cell carcinoma-derived exosomal MicroRNA-31 acts as an oncogene by targeting the tumor suppressor RhoBTB1. Arch Dermatol Res. 2024;317(1):114. doi: 10.1007/s00403-024-03558-0 [DOI] [PubMed] [Google Scholar]
  • 187.Zhao Z, Zhang H, Zeng Q, et al. Exosomes from 5-aminolevulinic acid photodynamic therapy-treated squamous carcinoma cells promote dendritic cell maturation. Photodiagnosis Photodyn Ther. 2020;30:101746. doi: 10.1016/j.pdpdt.2020.101746 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing not applicable - no new data generated.


Articles from International Journal of Nanomedicine are provided here courtesy of Dove Press

RESOURCES