Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (771)

Search Parameters:
Keywords = mesenchymal extracellular vesicles

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 4023 KB  
Article
Hacking Extracellular Vesicles: Using Vesicle-Related Tags to Engineer Mesenchymal Stromal Cell-Derived Extracellular Vesicles
by Gabriele Scattini, Giulia Pianigiani, Stefano Capomaccio, Maria Rachele Ceccarini, Samanta Mecocci, Laura Musa, Luca Avellini, Olimpia Barbato, Antonello Bufalari, Patrizia Casagrande Proietti, Rodolfo Gialletti, Alessia Sulla, Tommaso Beccari and Luisa Pascucci
Pharmaceutics 2025, 17(11), 1435; https://doi.org/10.3390/pharmaceutics17111435 - 6 Nov 2025
Viewed by 342
Abstract
Background/Objectives: Extracellular Vesicles (EVs) have shown great promise as diagnostic and therapeutic tools, as well as pharmacological nanocarriers. Various strategies are being explored to develop EVs for monitoring, imaging, loading with pharmacological agents, and surface decoration with tissue-specific ligands. EVs derived from [...] Read more.
Background/Objectives: Extracellular Vesicles (EVs) have shown great promise as diagnostic and therapeutic tools, as well as pharmacological nanocarriers. Various strategies are being explored to develop EVs for monitoring, imaging, loading with pharmacological agents, and surface decoration with tissue-specific ligands. EVs derived from Mesenchymal Stromal Cells (MSC-EVs) are of particular interest both as therapeutics per se and as natural nanocarriers for the targeted delivery of biotherapeutics. Methods: In this study, we investigated the ability of different tags to deliver a reporter protein into canine MSC-EVs with the aim of identifying the most effective endogenous loading mechanism. To this aim, canine MSCs were engineered to express the Green Fluorescent Protein (GFP) fused to CD63, Syntenin-1, TSG101, and the palmitoylation signal of Lck, which were expected to promote GFP incorporation into EVs. Overexpression of tagged GFP in canine MSCs was confirmed by Western blotting and examined by confocal microscopy and transmission electron microscopy to map intracellular localization. Results: All tags were able to deliver GFP into EVs. Syntenin-1 showed relatively high loading efficiency and secretion index but exhibited a diffuse localization pattern in the transfected cells. The palmitoylation signal showed low loading efficiency and localization specificity. TSG101 displayed a morphological pattern consistent with specific localization in endosomal structures, but its low expression level prevented further evaluations. Finally, CD63 showed the highest expression efficiency, as GFP-CD63 levels were approximately 5-fold higher than untagged GFP. Conclusions: In conclusion, CD63 emerged as the most suitable tag for canine MSC-EV engineering. Indeed, even if the secretion index favours Syntenin-1, CD63’s higher abundance in the lysate suggests its substantial post-secretion uptake. Further studies aimed at elucidating CD63’s specific contribution and identifying the domains involved in vesicle trafficking could provide valuable insights into EV bioengineering. Full article
(This article belongs to the Special Issue Extracellular Vesicles for Targeted Delivery)
Show Figures

Graphical abstract

20 pages, 1368 KB  
Review
Targeting Mitochondrial Dynamics via EV Delivery in Regenerative Cardiology: Mechanistic and Therapeutic Perspectives
by Dhienda C. Shahannaz, Tadahisa Sugiura, Brandon E. Ferrell and Taizo Yoshida
Cells 2025, 14(21), 1738; https://doi.org/10.3390/cells14211738 - 5 Nov 2025
Viewed by 455
Abstract
Mitochondrial dysfunction is a key contributor to cardiac injury and heart failure, and extracellular vesicles (EVs) have emerged as promising therapeutic agents due to their ability to deliver mitochondrial-targeted cargo. This review systematically maps the evidence on how EVs modulate mitochondrial dynamics—including fusion, [...] Read more.
Mitochondrial dysfunction is a key contributor to cardiac injury and heart failure, and extracellular vesicles (EVs) have emerged as promising therapeutic agents due to their ability to deliver mitochondrial-targeted cargo. This review systematically maps the evidence on how EVs modulate mitochondrial dynamics—including fusion, fission, mitophagy, and biogenesis—in regenerative cardiology. We comprehensively searched PubMed, Scopus, and Web of Science up to September 2025 for original studies. A total of 48 studies were included, with most utilizing EVs from mesenchymal stem cells, induced pluripotent stem cells, or cardiac progenitors. The review found that EV cargo influences key pathways such as DRP1 and MFN2, restores mitochondrial membrane potential, reduces ROS accumulation, and improves cardiomyocyte survival. While engineered EVs showed enhanced specificity, a lack of standardized preparation and quantitative assessment methods remains a significant challenge. We conclude that EV-mediated mitochondrial modulation is a promising strategy for cardiac repair, but the field needs harmonized protocols, deeper mechanistic understanding, and improved translational readiness to advance beyond preclinical research. The future of this research lies in integrating systems biology and precision targeting. Full article
(This article belongs to the Special Issue Advances in Cardiomyocyte and Stem Cell Biology in Heart Disease)
Show Figures

Figure 1

31 pages, 1502 KB  
Review
Non-Coding RNAs (microRNAs, lncRNAs, circRNAs) in Adenomyosis: A Systematic Review of Mechanistic and Translational Evidence
by Rafał Watrowski, Stoyan Kostov, Mario Palumbo, Andrea Rosati, Radmila Sparić, Ibrahim Alkatout, Ingolf Juhasz-Böss, Salvatore Giovanni Vitale and Liliana Mereu
Int. J. Mol. Sci. 2025, 26(21), 10713; https://doi.org/10.3390/ijms262110713 - 4 Nov 2025
Viewed by 453
Abstract
Adenomyosis (AM) is a hormonally responsive uterine disorder defined by ectopic endometrial tissue within the myometrium, causing pain, abnormal bleeding, and subfertility. Non-coding RNAs (ncRNAs)—including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs)—are post-transcriptional regulators implicated also in uterine remodeling. We [...] Read more.
Adenomyosis (AM) is a hormonally responsive uterine disorder defined by ectopic endometrial tissue within the myometrium, causing pain, abnormal bleeding, and subfertility. Non-coding RNAs (ncRNAs)—including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs)—are post-transcriptional regulators implicated also in uterine remodeling. We systematically reviewed original studies evaluating ncRNAs in AM using human samples, in vitro and animal models, or bioinformatic approaches. Data sources included PubMed and Google Scholar (inception up to 10 August 2025). Forty-one studies were included and synthesized across mechanistic, diagnostic, and translational domains. miRNAs (n = 31) were the most studied subclass, followed by lncRNAs (n = 10) and circRNAs (n = 5). Recurrent miRNAs such as miR-10b and miR-30c-5p (downregulated, inhibitory) and miR-145 (upregulated, promotive) regulate epithelial invasion, epithelial–mesenchymal transition, and cytoskeletal remodeling via PI3K–AKT/MAPK and Talin1 signaling. The let-7a/LIN28B axis governed estrogen-sensitive proliferation in the junctional zone, while miR-21 exhibited compartment-specific roles in decidualization and ectopic cell survival. Extracellular-vesicle (EV)-bornemiRNAs (e.g., miR-92a-3p, miR-25-3p, miR-4669) contributed to immune polarization and show early diagnostic potential. lncRNAs and circRNAs acted via chromatin modifiers and ceRNA networks. Most findings remain at the discovery stage. Convergent dysregulation was observed in key signaling pathways, including JAK–STAT, Wnt/β-catenin, and Hippo–YAP. ncRNAs regulate critical axes of invasion, proliferation, immune modulation, and hormonal response in AM. Targets with preliminary causal support—miR-10b/ZEB1, let-7a/LIN28B, and miR-145/Talin1—warrant further validation. Circulating miRNAs—especially in EVs—offer promise for non-invasive diagnosis. Full article
(This article belongs to the Special Issue MicroRNAs as Biomarkers and Therapeutic Targets in Human Diseases)
Show Figures

Figure 1

22 pages, 3393 KB  
Article
Comprehensive Characterization and In Vitro Functionality Study of Small Extracellular Vesicles Isolated by Different Purification Methods from Mesenchymal Stem Cell Cultures
by Marta Venturella, Ali Navaei and Davide Zocco
Int. J. Mol. Sci. 2025, 26(21), 10602; https://doi.org/10.3390/ijms262110602 - 30 Oct 2025
Viewed by 398
Abstract
Mesenchymal stem cells (MSCs) exhibit therapeutic properties, which have been attributed to their secretome, the set of secreted factors comprising cytokines, growth factors, and extracellular vesicles (EVs). In particular, small extracellular vesicles (sEVs) or exosomes, ranging between 30 nm and 120 nm in [...] Read more.
Mesenchymal stem cells (MSCs) exhibit therapeutic properties, which have been attributed to their secretome, the set of secreted factors comprising cytokines, growth factors, and extracellular vesicles (EVs). In particular, small extracellular vesicles (sEVs) or exosomes, ranging between 30 nm and 120 nm in diameter, can target specific tissues to deliver molecular payloads, thus lending themselves as promising platform for cell-free therapies. In this study, sEVs were purified from the conditioned medium (CM) harvested from human bone marrow-derived MSC culture and purified using size-exclusion chromatography (SEC) or density gradient ultracentrifugation (DG-UC). Then sEVs were fully characterized for identity and integrity using multiple analytical methods, including single-particle, transcriptomic and proteomic analyses. Different in vitro cell-based assays were established to evaluate the biological effects of the purified sEVs. Specifically, scratch wound healing and tube formation assays using human umbilical vein endothelial cells (HUVECs) were used to evaluate the regenerative properties of MSC-sEVs. Our findings demonstrated that the in vitro functional properties of MSC-sEVs are correlated with sEVs’ purity levels obtained by different purification methods. Full article
(This article belongs to the Special Issue Exosomes—3rd Edition)
Show Figures

Graphical abstract

23 pages, 4846 KB  
Article
Characterizing the Long Non-Coding RNA Profile of Endometrial Mesenchymal Stem/Stromal Cell-Derived Extracellular Vesicles and Their Anti-Inflammatory Role in Osteoarthritis
by Cole Conforti, Darden Wood Kimbrough, Neep Patel, Michelle B. R. G. Ley, Jose Medina Flores, Diego Correa, Lee D. Kaplan, Thomas M. Best and Dimitrios Kouroupis
Int. J. Mol. Sci. 2025, 26(21), 10567; https://doi.org/10.3390/ijms262110567 - 30 Oct 2025
Viewed by 784
Abstract
Endometrial tissue-derived mesenchymal stem/stromal cells (eMSCs) have potential therapeutic properties partially exerted via their secreted extracellular vesicles (EVs). eMSC-EVs contain cargos with regenerative and immunomodulatory properties. Specifically, the miRNA profile of CD146High eMSC-EVs has been shown to promote anti-inflammatory M2 macrophage polarization in [...] Read more.
Endometrial tissue-derived mesenchymal stem/stromal cells (eMSCs) have potential therapeutic properties partially exerted via their secreted extracellular vesicles (EVs). eMSC-EVs contain cargos with regenerative and immunomodulatory properties. Specifically, the miRNA profile of CD146High eMSC-EVs has been shown to promote anti-inflammatory M2 macrophage polarization in vitro. Herein, we aimed to characterize the lncRNA profile of CD146High and CD146Low eMSC-EVs and further assess their immunomodulatory and anabolic therapeutic function in osteoarthritis (OA). We hypothesized that the CD146High eMSC-EVs lncRNA profile is enriched with potent anti-inflammatory and pro-anabolic cartilage effects when compared to the CD146Low eMSC-EVs lncRNA profile. Human endometrial tissue was collected, and the eMSCs were magnetically sorted to yield the CD146High and CD146Low eMSC subpopulations. The eMSC-EVs were isolated via ultracentrifugation and CD63 magnetic immunoselection methods and characterized by nanosight and flow cytometry analyses. Our results showed that CD146High eMSC-EVs display an lncRNA profile with both anabolic and catabolic features, exerting a more dynamic effect on chondrocyte gene expression than CD146Low eMSC-EVs, suggesting a potential benefit of using CD146High eMSC-EVs to attenuate the negative effects of inflammation in OA. CD146High eMSC-EVs also demonstrated greater endothelial repair capacity under inflammatory stress. In conclusion, cell-free CD146High eMSC-EV has therapeutic potential through its protective anti-inflammatory effects, warranting further pre-clinical investigation. Full article
Show Figures

Figure 1

19 pages, 3678 KB  
Article
miR-7704-Enriched Stem Cell-Derived Extracellular Vesicles Attenuate Hyperoxia-Induced Apoptosis and Oxidation in Lung Epithelial Cells
by Yu-Hsun Chang, Kun-Chi Wu and Dah-Ching Ding
Curr. Issues Mol. Biol. 2025, 47(11), 893; https://doi.org/10.3390/cimb47110893 - 28 Oct 2025
Viewed by 239
Abstract
Bronchopulmonary dysplasia (BPD) is a significant complication of hyperoxia in preterm neonates. Extracellular vesicle (EV)-based therapies derived from mesenchymal stem cells (MSCs) show regenerative potential. We investigated the therapeutic efficacy of EVs derived from human umbilical cord mesenchymal stem cells (HUCMSCs), particularly those [...] Read more.
Bronchopulmonary dysplasia (BPD) is a significant complication of hyperoxia in preterm neonates. Extracellular vesicle (EV)-based therapies derived from mesenchymal stem cells (MSCs) show regenerative potential. We investigated the therapeutic efficacy of EVs derived from human umbilical cord mesenchymal stem cells (HUCMSCs), particularly those engineered to overexpress miR-7704 in a hyperoxia-induced BPD cell model. EVs were isolated from GFP- and miR-7704-transfected HUCMSCs. A549 alveolar epithelial cells were exposed to normoxic or hyperoxic conditions and treated with HUCMSC-EV or miR-7704-HUCMSC-EV. EV uptake was confirmed using fluorescence microscopy. Cell proliferation was evaluated, and apoptosis was assessed by means of Western blot analysis of caspase family proteins and apoptosis-related markers. Both HUCMSC-EV and miR-7704-HUCMSC-EV enhanced A549 cell proliferation under hyperoxic stress, with miR-7704-HUCMSC-EV showing greater efficacy. Protein-level analyses revealed hyperoxia-induced increases in cleaved caspase-3, caspase-7, and FasL, along with decreased Bcl-2. Treatment with miR-7704-HUCMSC-EV significantly reversed these effects, whereas HUCMSC-EVs minimally impacted apoptotic protein expression. Bioinformatic analysis predicted that hsa-miR-7704 targeted the 3′ UTR of APOPT1. miR-7704-HUCMSC EVs also enhanced the expression of key antioxidant enzymes, including SOD1, SOD2, and HO-1. miR-7704-enriched HUCMSC-derived EV significantly promoted cell survival and mitigated hyperoxia-induced apoptosis and oxidation in a BPD cell model, suggesting their potential therapeutic role in neonatal lung injury. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

21 pages, 10601 KB  
Article
Hypoxia-Induced Extracellular Vesicles Derived from Human Umbilical Cord Mesenchymal Stem Cells Regulate Macrophage Polarization and Enhance Angiogenesis to Promote Diabetic Wound Healing
by Yongfeng Su, Junda Lu, Feiyuan Liang and Jianwen Cheng
Biomolecules 2025, 15(11), 1504; https://doi.org/10.3390/biom15111504 - 24 Oct 2025
Viewed by 548
Abstract
Background: Diabetic wound healing has always been a clinical challenge with minimal response or efficacy to standard treatment. This study aims to assess the therapeutic potential of hypoxia-induced extracellular vesicles (hy-EVs) produced by human umbilical cord mesenchymal stem cells (HUCMSCs) to treat [...] Read more.
Background: Diabetic wound healing has always been a clinical challenge with minimal response or efficacy to standard treatment. This study aims to assess the therapeutic potential of hypoxia-induced extracellular vesicles (hy-EVs) produced by human umbilical cord mesenchymal stem cells (HUCMSCs) to treat diabetic wounds. Methods: HUCMSCs were isolated from umbilical cord tissue, cultured under hypoxic conditions to induce the release of extracellular vesicles (EVs) and compared with normoxia-induced extracellular vesicles (n-EVs). We assessed the functions of hy-EVs on human skin fibroblasts (HSFs) and human umbilical vein endothelial cells (HUVECs) in vitro. Simultaneously, we analyzed the pro-angiogenic effects of hy-EVs, their effects on macrophage polarization, and their ability to scavenge endogenous reactive oxygen species (ROS). In addition, a diabetic wound model was established to assess the curative effect of hy-EVs in diabetic wound healing. Results: We found by in vitro study that hy-EVs markedly improved the functional activities of HSFs, thus significantly promoting wound repair. Remarkably, it was determined that hy-EVs greatly enhanced the proliferation and migration ability as well as the angiogenic ability of HUVECs, while promoting the expression of hypoxia-inducible factor-1α (HIF-1α), vascular endothelial-generation-associated factor A (VEGFA), and platelet endothelial adhesion molecule (CD31), which suggested that hy-EVs can effectively activate the HIF-1α pathway to promote angiogenesis. Above all, we found that hy-EVs promoted the expression of CD206 while decreasing the expression of CD86, suggesting that hy-EVs could induce macrophages to shift from M1-type (pro-inflammatory) to M2-type (anti-inflammatory), thereby modulating the inflammatory response. Additionally, hy-EVs inhibited ROS production in both HSFs and HUVECs to reduce oxidative stress. In vivo results showed that hy-EVs enhanced collagen deposition and angiogenesis, modulated macrophage polarization, and inhibited immune response at the wound spot, which significantly enhanced diabetic wound healing. Conclusions: Our study shows that hy-EVs significantly promote angiogenesis through activation of the HIF-1α pathway, modulate macrophage polarization and attenuate cellular oxidative stress, possibly through delivery of specific miRNAs and proteins. Our discoveries offer a key theoretical basis and potential application to develop novel therapeutic strategies against diabetes-related tissue injury. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

24 pages, 4267 KB  
Article
“Attractive” Treatment for Abdominal Aortic Aneurysm Repair: Magnetic Localization of Silk-Iron Packaged Extracellular Vesicles
by Ande X. Marini, Kiran J. McLoughlin, Amanda R. Pellegrino, Golnaz N. Tomaraei, Bo Li, John A. Curci, Mostafa Bedewy, Justin S. Weinbaum and David A. Vorp
J. Funct. Biomater. 2025, 16(11), 395; https://doi.org/10.3390/jfb16110395 - 22 Oct 2025
Viewed by 998
Abstract
Abdominal aortic aneurysm (AAA) is a dilatation of the distal aorta to a diameter of 50% or more of its normal size of about 2 cm. Risk of aortic rupture can be nearly eliminated with either open surgery or endovascular repair. Procedural risks [...] Read more.
Abdominal aortic aneurysm (AAA) is a dilatation of the distal aorta to a diameter of 50% or more of its normal size of about 2 cm. Risk of aortic rupture can be nearly eliminated with either open surgery or endovascular repair. Procedural risks limit the value of these interventions unless the diameter of the aneurysm has reached a critical threshold (established as 5.5 cm in men or 5.0 cm in women). Thus, patients are monitored until this threshold is reached. Approximately 80% of small AAA will grow and exceed the threshold, providing a therapeutic window for altering this natural history and reducing the risk of rupture. Previous work in our lab has utilized adipose-derived mesenchymal stem cells (ASCs) to treat AAA in vivo, preserving elastic fibers and slowing aneurysm expansion. This work sought to create a delivery system for therapeutic extracellular vesicles (ASC-EVs) secreted by ASCs. Our delivery system incorporated the biocompatibility of regenerated silk fibroin (RSF), the magnetic moveability of iron oxide nanoparticles (IONPs), and the regenerative nature of ASC-EVs to create silk-iron packaged extracellular vesicles (SIPEs). Using this system, we tested the ability to magnetically localize the SIPEs and release their encapsulated ASC-EVs to exert their regenerative effects in vitro. We were successful in magnetically localizing the SIPEs in vitro and silk-iron microparticles (SIMPs) in vivo and in detecting their releasates via flow cytometry and cellular uptake assays. However, while their releasates were detected, their biological effects were diminished compared to unencapsulated controls. Thus, additional optimization related to loading efficiency is needed. Full article
(This article belongs to the Special Issue Cardiovascular Tissue Engineering: Current Status and Advances)
Show Figures

Graphical abstract

20 pages, 2411 KB  
Article
Selective Paracrine Modulation of Stromal Cells: Wharton’s Jelly MSC Secretome Enhances Adipose-Derived MSC Functionality While Maintaining Dermal Fibroblast Quiescence
by Tanya Stoyanova, Lora Topalova, Stanimir Kyurkchiev, Regina Komsa-Penkova, Svetla Todinova and George Altankov
Int. J. Mol. Sci. 2025, 26(20), 10095; https://doi.org/10.3390/ijms262010095 - 16 Oct 2025
Viewed by 496
Abstract
Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) secrete a rich array of paracrine factors including growth factors, cytokines, and extracellular vesicles that hold promises for regenerative medicine. This study evaluated the effects of WJ-MSC-derived secretome on adipose-derived mesenchymal stem cells (AD-MSCs) and human dermal [...] Read more.
Wharton’s jelly-derived mesenchymal stem cells (WJ-MSCs) secrete a rich array of paracrine factors including growth factors, cytokines, and extracellular vesicles that hold promises for regenerative medicine. This study evaluated the effects of WJ-MSC-derived secretome on adipose-derived mesenchymal stem cells (AD-MSCs) and human dermal fibroblasts (HDFs), focusing on their adhesion, spreading, proliferation, endogenous collagen secretion, and migration. Morphometric analysis revealed that the secretome enhanced cell adhesion and spreading on rat tail collagen (RTC) substrates after 24 h. AD-MSCs showed a ~30% increase in the cell spreading area (from 4007 μm2 to 5081 μm2p < 0.05), though without notable shape changes. In contrast, fetal bovine serum (FBS) promoted cell elongation with a reduced aspect ratio. Proliferation assays demonstrated a selective stimulatory effect of the secretome on AD-MSCs with a significant increase at day 3, while HDFs’ proliferation remained unchanged. Cell cycle profiling showed transient S-phase accumulation in AD-MSCs (24–48 h), followed by G0/G1 arrest (72 h), while HDFs remained in G0/G1. Immunofluorescence analysis confirmed the enhanced extracellular deposition of endogenously synthesized collagen in AD-MSCs, while no comparable response was observed in HDFs. Scratch assays showed increased migration in both cell types upon secretome exposure compared to collagen-only controls, suggesting a paracrine-mediated pro-migratory effect. These results demonstrate that WJ-MSC secretome boosts the regenerative capacity in AD-MSCs while keeping fibroblasts quiescent, highlighting its strong potential for cell-free therapies in tissue engineering, wound repair, and regenerative medicine. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Graphical abstract

52 pages, 3943 KB  
Review
Applications of Modern Cell Therapies: The Latest Data in Ophthalmology
by Ioannis Iliadis, Nadezhda A. Pechnikova, Malamati Poimenidou, Diamantis D. Almaliotis, Ioannis Tsinopoulos, Tamara V. Yaremenko and Alexey V. Yaremenko
Life 2025, 15(10), 1610; https://doi.org/10.3390/life15101610 - 16 Oct 2025
Viewed by 1523
Abstract
Cell-based therapeutics are redefining interventions for vision loss by enabling tissue replacement, regeneration, and neuroprotection. This review surveys contemporary cellular strategies in ophthalmology through the lenses of therapeutic effectiveness, translational readiness, and governance. We profile principal sources—embryonic and induced pluripotent stem cells, mesenchymal [...] Read more.
Cell-based therapeutics are redefining interventions for vision loss by enabling tissue replacement, regeneration, and neuroprotection. This review surveys contemporary cellular strategies in ophthalmology through the lenses of therapeutic effectiveness, translational readiness, and governance. We profile principal sources—embryonic and induced pluripotent stem cells, mesenchymal stromal cells, retinal pigment epithelium, retinal progenitor and limbal stem cells—and enabling platforms including extracellular vesicles, encapsulated cell technology and biomaterial scaffolds. We synthesize clinical evidence across age-related macular degeneration, inherited retinal dystrophies, and corneal injury/limbal stem-cell deficiency, and highlight emerging applications for glaucoma and diabetic retinopathy. Delivery routes (subretinal, intravitreal, anterior segment) and graft formats (single cells, sheets/patches, organoids) are compared using standardized structural and functional endpoints. Persistent barriers include GMP-compliant derivation and release testing; differentiation fidelity, maturation, and potency; genomic stability and tumorigenicity risk; graft survival, synaptic integration, and immune rejection despite ocular immune privilege; the scarcity of validated biomarkers and harmonized outcome measures and ethical, regulatory, and health-economic constraints. Promising trajectories span off-the-shelf allogeneic products, patient-specific iPSC-derived grafts, organoid and 3D-bioprinted tissues, gene-plus-cell combinations, and cell-free extracellular-vesicle therapeutics. Overall, cell-based therapies remain investigational. With adequately powered trials, methodological harmonization, long-term surveillance, scalable xeno-free manufacturing, and equitable access frameworks, they may eventually become standards of care; at present, approvals are limited to specific products/indications and regions, and no cell therapy is the standard of care for retinal disease. Full article
(This article belongs to the Special Issue Advances in Biomedical Frontier Technologies and Disease Diagnosis)
Show Figures

Figure 1

29 pages, 2004 KB  
Review
Emerging Roles of Extracellular Vesicles in the Pathogenesis, Diagnosis, and Therapy of Periodontitis
by Yiru Fu, Mengmeng Wang, Rui Teng and Ang Li
Biomedicines 2025, 13(10), 2521; https://doi.org/10.3390/biomedicines13102521 - 16 Oct 2025
Viewed by 997
Abstract
Periodontitis is a globally prevalent oral disease and is closely associated with various systemic diseases. Periodontitis arises from dynamic and complex interactions between polymicrobial communities and host immune responses. Extracellular vesicles (EVs) are circulating subcellular particles carrying multiple signaling molecules. EVs play a [...] Read more.
Periodontitis is a globally prevalent oral disease and is closely associated with various systemic diseases. Periodontitis arises from dynamic and complex interactions between polymicrobial communities and host immune responses. Extracellular vesicles (EVs) are circulating subcellular particles carrying multiple signaling molecules. EVs play a key role in intercellular communication, and hold promise for diagnostic and therapeutic purposes. Bacterial extracellular vesicles (BEVs), released from oral pathogens, have been implicated in delivering virulence factors to host cells. In contrast, host cell-derived EVs (CEVs), secreted by periodontal cells, contain molecular cargo that reflect disease status. Both BEVs and CEVs contribute to periodontitis progression by exacerbating inflammation and tissue destruction, and they may also influence related systemic diseases. Moreover, the molecular components of EVs derived from saliva and gingival crevicular fluid (GCF) show potential as diagnostic biomarkers for periodontitis. In addition, mesenchymal stem cell-derived EVs (MSC-EVs) exhibit therapeutic potential in periodontitis, and engineering approaches have been developed to enhance their therapeutic efficacy and accelerate clinical translation. This review summarizes recent advances in understanding the pathogenic, diagnostic, and therapeutic roles of EVs in periodontitis and discusses current challenges and future directions toward their clinical application. Full article
(This article belongs to the Special Issue Biomedicine in Dental and Oral Rehabilitation)
Show Figures

Figure 1

18 pages, 4921 KB  
Article
Nano-Encapsulated Spicule System Enhances Delivery of Wharton’s Jelly MSC Secretome and Promotes Skin Rejuvenation: Preclinical and Clinical Evaluation
by Na Eun Lee, Ji Eun Kim, Chi Young Bang and Oh Young Bang
Int. J. Mol. Sci. 2025, 26(20), 10024; https://doi.org/10.3390/ijms262010024 - 15 Oct 2025
Viewed by 1280
Abstract
Wharton’s Jelly-derived mesenchymal stem cell (WJ-MSC) secretome contains diverse bioactive factors with potential for skin regeneration, but its clinical efficacy is limited by poor transdermal delivery. In this study, we developed a dual-delivery system by nanoencapsulating WJ-MSC secretome and coating it onto marine [...] Read more.
Wharton’s Jelly-derived mesenchymal stem cell (WJ-MSC) secretome contains diverse bioactive factors with potential for skin regeneration, but its clinical efficacy is limited by poor transdermal delivery. In this study, we developed a dual-delivery system by nanoencapsulating WJ-MSC secretome and coating it onto marine sponge-derived spicules. Physicochemical characterization, in vitro assays (fibroblast and keratinocyte proliferation, keratinocyte migration, type I procollagen secretion, and antioxidant activity), and in vivo penetration studies were conducted. A single-arm clinical trial evaluated dermal absorption, pore characteristics, skin texture, wrinkles, and pigmentation following topical application. Transdermal penetration efficiency was significantly higher in the nano-coated spicule group than in the uncoated secretome control. In vitro, secretome treatment promoted fibroblast and keratinocyte activity, accelerated wound closure, and increased collagen synthesis. Clinically, a single application enhanced dermal absorption and significantly reduced pore number, while two weeks of treatment decreased wrinkles and pigmentation. Spicule-based nanoencapsulation effectively overcomes the skin barrier, enhances the regenerative activity of WJ-MSC secretome, and induces measurable clinical improvements in skin rejuvenation. This platform represents a promising cosmetic and therapeutic strategy in dermatology. Full article
(This article belongs to the Special Issue Roles and Function of Extracellular Vesicles in Diseases: 3rd Edition)
Show Figures

Graphical abstract

28 pages, 1878 KB  
Review
Exosomes in Osteoarthritis: Breakthrough Innovations and Advanced Tissue Engineering for Cartilage Regeneration Since 2020
by Xiao-He Yang, Shu-Yin Chen, Quan-Fa Zhou and You-Zhi Cai
Biomedicines 2025, 13(10), 2486; https://doi.org/10.3390/biomedicines13102486 - 13 Oct 2025
Viewed by 1400
Abstract
Background/Objectives: Osteoarthritis (OA) is a prevalent age-related degenerative joint disease causing cartilage damage, leading to a debilitating lifestyle. However, there are currently no drugs on the market that promote cartilage repair, and advanced cases often require arthroplasty. Increasing evidence suggests that exosomes, the [...] Read more.
Background/Objectives: Osteoarthritis (OA) is a prevalent age-related degenerative joint disease causing cartilage damage, leading to a debilitating lifestyle. However, there are currently no drugs on the market that promote cartilage repair, and advanced cases often require arthroplasty. Increasing evidence suggests that exosomes, the smallest extracellular vesicles (30–150 nm) secreted by all cell types, are involved in the pathological process of OA and play a crucial and complex role in its progression. This review aims to provide in-depth insights into exosome biology, isolation techniques, their role in OA pathophysiology, and their clinical therapeutic potential. Methods: We systematically reviewed studies published since 2020 on exosomes in OA, focusing on their biological properties, isolation techniques, pathological roles, and therapeutic applications. Results: Exosomes derived from synovial fluid, chondrocytes, synoviocytes, and mesenchymal stem cells regulate key processes in OA progression, including inflammation, apoptosis, extracellular matrix degradation, and regeneration. Various cell-derived exosomes show therapeutic potential for cartilage damage/OA. However, their mechanisms of action have not been fully investigated. Moreover, emerging methodologies, such as utilizing novel materials for exosome delivery, potentially facilitate the development of more effective and personalized therapeutic interventions. Conclusions: Exosomes exert dual roles in OA pathogenesis and therapy. Although challenges remain regarding their sources, dosage, delivery, and standardization, exosome-based strategies represent a promising cell-free therapeutic approach with potential applications in personalized and precision medicine. Full article
(This article belongs to the Special Issue Advances in Extracellular Vesicles in Therapeutics)
Show Figures

Figure 1

20 pages, 3797 KB  
Article
Induced Mammary Epithelial Cell-Derived Extracellular Vesicles Promote the Repair of Skin Trauma
by Siyao Pan, Dandan Zhang, Guodong Wang, Longfei Sun, Mengzhen Wei, Shan Deng, Jianwei Chen, Prasanna Kallingappa, Xiang Yuan and Ben Huang
Int. J. Mol. Sci. 2025, 26(20), 9929; https://doi.org/10.3390/ijms26209929 - 12 Oct 2025
Viewed by 531
Abstract
Although extracellular vesicles (EVs) from mesenchymal stem cells have shown potential in skin wound repair, the diversity of EV sources and the optimization of delivery systems still need further exploration. This study is the first to demonstrate that extracellular vesicles from chemically induced [...] Read more.
Although extracellular vesicles (EVs) from mesenchymal stem cells have shown potential in skin wound repair, the diversity of EV sources and the optimization of delivery systems still need further exploration. This study is the first to demonstrate that extracellular vesicles from chemically induced mammary epithelial cells (CiMECs-EVs) possess distinct skin wound repair activity. To enhance the therapeutic efficacy of CiMECs-EVs and optimize their delivery efficiency, we innovatively combined them with a chitosan hydrogel to construct a composite repair system (CiMECs-EVs-chitosan hydrogel, CMECG). This system was then applied to a rat skin wound model. The results showed that CMECG significantly promoted the proliferation and migration of fibroblasts and mammary epithelial cells (MECs). In animal experiments, the relative wound closure efficiency of the control group was approximately 70% on day 14, while that of the CMECG group (loaded with 200 μg CiMECs-Exo) was enhanced to 90%, markedly accelerating the wound healing process. Histological analysis indicated that this system could effectively restore the structural continuity of various skin layers and significantly promote the synthesis and remodeling of collagen at the wound site. Mechanistically, the wound healing effect of CiMECs-EVs is closely associated with the endogenous miRNAs they encapsulate. These miRNAs can coordinately regulate cell proliferation, migration, and angiogenesis, modulate the inflammatory microenvironment, and inhibit excessive scar formation—thus regulating the entire repair process. This process involves multiple wound healing-related signaling pathways, including MAPK, PI3K-Akt, FoxO, TGF-β, and JAK-STAT. In summary, this study successfully constructed a novel EV-chitosan hydrogel repair system. This system is expected to provide an effective and innovative EV-based therapeutic strategy for the clinical treatment of skin wound repair. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

17 pages, 3452 KB  
Article
CAP-LAMP2b–Modified Stem Cells’ Extracellular Vesicles Hybrid with CRISPR-Cas9 Targeting ADAMTS4 to Reverse IL-1β–Induced Aggrecan Loss in Chondrocytes
by Kun-Chi Wu, Yu-Hsun Chang, Raymond Yuh-Shyan Chiang and Dah-Ching Ding
Int. J. Mol. Sci. 2025, 26(19), 9812; https://doi.org/10.3390/ijms26199812 - 9 Oct 2025
Viewed by 760
Abstract
Extracellular vesicles (EVs) from mesenchymal stem cells hold therapeutic promise for inflammatory and degenerative diseases; however, limited delivery and targeting capabilities hinder their clinical use. In this study, we sought to enhance the anti-inflammatory and chondroprotective effects of EVs through CAP-LAMP2b (chondrocyte affinity [...] Read more.
Extracellular vesicles (EVs) from mesenchymal stem cells hold therapeutic promise for inflammatory and degenerative diseases; however, limited delivery and targeting capabilities hinder their clinical use. In this study, we sought to enhance the anti-inflammatory and chondroprotective effects of EVs through CAP-LAMP2b (chondrocyte affinity peptide fused to an EV membrane protein) engineering and ADAMTS4 gene editing hybrid vesicle formation. Human umbilical cord MSCs (hUCMSCs) were characterized via morphology, immunophenotyping, and trilineage differentiation. EVs from control and CAP-LAMP2b-transfected hUCMSCs were fused with liposomes carrying CRISPR-Cas9 ADAMTS4 gRNA. DiI-labeled EV uptake was assessed via fluorescence imaging. CAP-LAMP2b was expressed in hUCMSCs and their EVs. EVs exhibited the expected size (~120 nm), morphology, and exosomal markers (CD9, CD63, CD81, HSP70). CAP-modified hybrid EVs significantly enhanced chondrocyte uptake compared to control EVs and liposomes. IL-1β increased ADAMTS4 expression, whereas CAP-LAMP2b-ADAMTS4 EVs, particularly clone SG3, reversed these effects by reducing ADAMTS4 and restoring aggrecan. Western blotting confirmed suppressed ADAMTS4 and elevated aggrecan protein. CAP-LAMP2b-ADAMTS4 EVs, therefore, showed superior uptake and therapeutic efficacy in inflamed chondrocytes, attenuating inflammatory gene expression and preserving matrix integrity. These results support engineered EVs as a promising cell-free approach for cartilage repair and osteoarthritis treatment. Full article
(This article belongs to the Section Molecular Biology)
Show Figures

Figure 1

Back to TopTop