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

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (908)

Search Parameters:
Keywords = extracellular vesicles cargo

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 34589 KiB  
Article
Extracellular Vesicle-Mediated miR-155 from Visceral Adipocytes Induces Skeletal Muscle Dysplasia in Obesity
by Yunyan Ji, Zeen Gong, Rui Liang, Di Wu, Wen Sun, Xiaomao Luo, Yi Yan, Jiayin Lu, Juan Wang and Haidong Wang
Cells 2025, 14(17), 1302; https://doi.org/10.3390/cells14171302 - 22 Aug 2025
Abstract
Obesity poses a serious threat to human health, with induced skeletal muscle dysfunction significantly increasing the risk of metabolic syndrome. In obesity, it is known that visceral adipose tissue (VAT) mediates the dysregulation of the adipose–muscle axis through exosome-delivered miRNAs, but the associated [...] Read more.
Obesity poses a serious threat to human health, with induced skeletal muscle dysfunction significantly increasing the risk of metabolic syndrome. In obesity, it is known that visceral adipose tissue (VAT) mediates the dysregulation of the adipose–muscle axis through exosome-delivered miRNAs, but the associated regulatory mechanisms remain incompletely elucidated. This study established an AAV-mediated miR-155 obese mouse model and a co-culture system (HFD VAD-evs/RAW264.7 cells/C2C12 cells) to demonstrate that high-fat diet-induced VA-derived extracellular vesicles (HFD VAD-evs) preferentially accumulate in skeletal muscle and induce developmental impairment. HFD VAD-evs disrupt skeletal muscle homeostasis through dual mechanisms: the direct suppression of myoblast development via exosomal miR-155 cargo and the indirect inhibition of myogenesis through macrophage-mediated inflammatory responses in skeletal muscle. Notably, miR-155 inhibition in HFD VAD-evs reversed obesity-associated myogenic deficits. These findings provide novel mechanistic insights into obesity-induced skeletal muscle dysregulation and facilitate potential therapeutic strategies targeting exosomal miRNA signaling. Full article
Show Figures

Figure 1

39 pages, 1435 KiB  
Review
Extracellular Vesicles in Osteogenesis: A Comprehensive Review of Mechanisms and Therapeutic Potential for Bone Regeneration
by Sreyee Biswas, Prakash Gangadaran, Chandrajeet Dhara, Shreya Ghosh, Soumya Deep Phadikar, Akash Chakraborty, Atharva Anand Mahajan, Ranit Mondal, Debdeep Chattopadhyay, Trisha Banerjee, Anuvab Dey, Subhrojyoti Ghosh, Anand Krishnan, Byeong-Cheol Ahn and Ramya Lakshmi Rajendran
Curr. Issues Mol. Biol. 2025, 47(8), 675; https://doi.org/10.3390/cimb47080675 - 21 Aug 2025
Viewed by 58
Abstract
Extracellular vesicles (EVs) are nanoscale, membrane-bound particles secreted by diverse cell types and act as pivotal mediators of intercellular communication during bone regeneration. These vesicles transport bioactive cargo including proteins, lipids, mRNAs, and microRNAs that modulate osteogenesis, angiogenesis, and immune responses within the [...] Read more.
Extracellular vesicles (EVs) are nanoscale, membrane-bound particles secreted by diverse cell types and act as pivotal mediators of intercellular communication during bone regeneration. These vesicles transport bioactive cargo including proteins, lipids, mRNAs, and microRNAs that modulate osteogenesis, angiogenesis, and immune responses within the bone microenvironment. EVs originating from mesenchymal stem cells, osteoblasts, endothelial cells, and macrophages have demonstrated substantial potential to promote bone formation, inhibit bone resorption, and enhance vascularization. This review examines the biogenesis, classification, and cellular uptake mechanisms of EVs, focusing on their roles in osteogenesis and their therapeutic applications in fracture healing, osteoporosis, and bone tissue engineering. Despite their promise, significant challenges remain, including the need for standardization, scalable production, and assessment of long-term safety to enable clinical translation of EV-based therapies. Here, we provide a comprehensive overview of EV biology, elucidate the molecular mechanisms of EVs in bone regeneration, and discuss innovative strategies to optimize their therapeutic efficacy, highlighting their potential as next-generation orthobiologics. Full article
Show Figures

Graphical abstract

26 pages, 2840 KiB  
Article
Immune Tuning in Extreme Environments: Protein Citrullinome and Extracellular Vesicle Signatures Comparing Hibernating Versus Active States in the Heterothermic and Heterometabolic Tenrec (Tenrec ecaudatus)
by Gilbecca Rae Smith, Pinar Uysal-Onganer, Igor Kraev, Frank van Breukelen and Sigrun Lange
Biology 2025, 14(8), 1056; https://doi.org/10.3390/biology14081056 - 15 Aug 2025
Viewed by 427
Abstract
Tenrecs are heterothermic burrowing mammals, which are capable of withstanding extreme environmental stressors, including during hibernation. Their phylogenetic position as reminiscent of an ancestral placental mammal makes tenrecs a unique model for evolutionarily conserved traits, with potential translatability to human physiology and pathobiology, [...] Read more.
Tenrecs are heterothermic burrowing mammals, which are capable of withstanding extreme environmental stressors, including during hibernation. Their phylogenetic position as reminiscent of an ancestral placental mammal makes tenrecs a unique model for evolutionarily conserved traits, with potential translatability to human physiology and pathobiology, including adaptations to extreme environments. In this study, we compared tenrec plasma for post-translational protein citrullination profiles (citrullinomes) and extracellular vesicle (EV) characteristics, including selected microRNA cargoes (miR-21, miR-155, miR-206, miR-210), between baseline active and hibernating states at low (12 °C) and high (28 °C) ambient temperatures. Our findings show considerable changes in citrullinome plasma profiles and associated Gene Ontology and KEGG pathway analysis linked to physiological and inflammatory processes, comparing hibernating and active states, also differing between the two ambient temperature groups. We furthermore identified modified EV profiles with respect to stress-related (miR-21, miR-155), hypoxia (miR-210) and metabolic/muscle related (miR-206) microRNA cargoes, which showed significant differences between active and hibernating animals, also comparing the two ambient temperature groups. Our findings show novel roles for post-translational protein citrullination in regulating immune and metabolic associated pathways in the tenrec, and highlight EV profiles, based on microRNA cargoes, as indicators for stress and metabolic responses in active versus hibernating states, including at different temperatures. Collectively our data highlights the tenrec as an evolutionary model for regulating pathobiological responses in extreme environments and may have translatable potential for human physiology and pathologies. Full article
(This article belongs to the Section Immunology)
Show Figures

Figure 1

17 pages, 1476 KiB  
Review
Extracellular Vesicle-Derived Bioactive Molecules for Corneal and Ocular Surface Regeneration
by Ana Kolenc, Živa Dimnik, Miha Marzidovšek, Petra Schollmayer, Marko Hawlina, Elvira Maličev and Zala Lužnik Marzidovšek
J. Clin. Med. 2025, 14(15), 5594; https://doi.org/10.3390/jcm14155594 - 7 Aug 2025
Viewed by 349
Abstract
Cell-based therapies emerge as potential treatment options for various debilitating diseases. Preclinical research and clinical studies involving cells increased exponentially in the past decade. In addition to cell-based approaches, the use of extracellular vesicles (EVs), which are released by nearly all cell types, [...] Read more.
Cell-based therapies emerge as potential treatment options for various debilitating diseases. Preclinical research and clinical studies involving cells increased exponentially in the past decade. In addition to cell-based approaches, the use of extracellular vesicles (EVs), which are released by nearly all cell types, emerged as a promising cell-free alternative. Those approaches are also being explored in the field of ophthalmology. Several clinical trials involving EVs are underway to develop potential treatments for advanced ocular surface diseases, including corneal disorders, injuries, and dry eye disease. The cargo carried by EVs has been shown to include a diverse array of functional molecules such as transcription factors, cytokines, growth factors, mRNA, tRNA, rRNA, miRNA, and fragments of dsDNA. While the molecular composition of EVs is already well characterised, the specific activity of these molecules upon delivery to recipient cells remains poorly understood. In this review, we summarise recent studies investigating the bioactive molecules within EVs shown to influence or modulate cellular activity on the ocular surface. Among these, various miRNAs have most commonly been identified as therapeutic agents targeting distinct molecular pathways. The EVs studied were predominantly derived from various mesenchymal stem cells. Full article
(This article belongs to the Section Ophthalmology)
Show Figures

Figure 1

27 pages, 6602 KiB  
Article
Extracellular Vesicle-Mediated Delivery of AntimiR-Conjugated Bio-Gold Nanoparticles for In Vivo Tumor Targeting
by Parastoo Pourali, Eva Neuhöferová, Behrooz Yahyaei, Milan Svoboda, Adéla Buchnarová and Veronika Benson
Pharmaceutics 2025, 17(8), 1015; https://doi.org/10.3390/pharmaceutics17081015 - 5 Aug 2025
Viewed by 418
Abstract
Background/Objectives: Extracellular vesicles (EVs) are involved in cell-to-cell communication and delivery of signaling molecules and represent an interesting approach in targeted therapy. This project focused on EV-mediated facilitation and cell-specific delivery of effector antimiR molecules carried by biologically produced gold nanoparticles (AuNPs). Methods: [...] Read more.
Background/Objectives: Extracellular vesicles (EVs) are involved in cell-to-cell communication and delivery of signaling molecules and represent an interesting approach in targeted therapy. This project focused on EV-mediated facilitation and cell-specific delivery of effector antimiR molecules carried by biologically produced gold nanoparticles (AuNPs). Methods: First, we loaded EVs derived from cancer cells 4T1 with AuNPs-antimiR. The AuNPs were also decorated with or without transferrin (Tf) molecules. We examined parental cell-specific delivery of the AuNPs-Tf-antimiR within monocultures as well as co-cultures in vitro. Subsequently, we used autologous EVs containing AuNPs-Tf-antimiR to target tumor cells in a xenograft tumor model in vivo. Efficacy of the antimir transfer was assessed by qPCR and apoptosis assessment. Results: In vitro, EVs loaded with AuNPs-antimiR were internalized only by the parental cells and the AuNPs-antimiR transfer was successful and effective only in EVs that were decorated with Tf. We achieved effective delivery of the antimiR molecule into cancer cells in vivo, which was proved by specific silencing of the target oncogenic miRNA as well as induction of cancer cells apoptosis. Conclusions: EVs represent an interesting and potent way for targeted cargo delivery and personalized medicine. On the other hand, there are various safety and efficacy challenges that remain to be addressed. Full article
(This article belongs to the Special Issue Cell-Mediated Delivery Systems)
Show Figures

Graphical abstract

18 pages, 929 KiB  
Review
From Hypoxia to Bone: Reprogramming the Prostate Cancer Metastatic Cascade
by Melissa Santos, Sarah Koushyar, Dafydd Alwyn Dart and Pinar Uysal-Onganer
Int. J. Mol. Sci. 2025, 26(15), 7452; https://doi.org/10.3390/ijms26157452 - 1 Aug 2025
Viewed by 557
Abstract
Bone is the most frequent site of distant metastasis in advanced prostate cancer (PCa), contributing substantially to patient morbidity and mortality. Hypoxia, a defining feature of the solid tumour microenvironment, plays a pivotal role in driving bone-tropic progression by promoting epithelial-to-mesenchymal transition (EMT), [...] Read more.
Bone is the most frequent site of distant metastasis in advanced prostate cancer (PCa), contributing substantially to patient morbidity and mortality. Hypoxia, a defining feature of the solid tumour microenvironment, plays a pivotal role in driving bone-tropic progression by promoting epithelial-to-mesenchymal transition (EMT), cancer stemness, extracellular matrix (ECM) remodelling, and activation of key signalling pathways such as Wingless/Integrated (Wnt) Wnt/β-catenin and PI3K/Akt. Hypoxia also enhances the secretion of extracellular vesicles (EVs), enriched with pro-metastatic cargos, and upregulates bone-homing molecules including CXCR4, integrins, and PIM kinases, fostering pre-metastatic niche formation and skeletal colonisation. In this review, we analysed current evidence on how hypoxia orchestrates PCa dissemination to bone, focusing on the molecular crosstalk between HIF signalling, Wnt activation, EV-mediated communication, and cellular plasticity. We further explore therapeutic strategies targeting hypoxia-related pathways, such as HIF inhibitors, hypoxia-activated prodrugs, and Wnt antagonists, with an emphasis on overcoming therapy resistance in castration-resistant PCa (CRPC). By examining the mechanistic underpinnings of hypoxia-driven bone metastasis, we highlight promising translational avenues for improving patient outcomes in advanced PCa. Full article
(This article belongs to the Special Issue Hypoxia: Molecular Mechanism and Health Effects)
Show Figures

Graphical abstract

26 pages, 9475 KiB  
Article
Microalgae-Derived Vesicles: Natural Nanocarriers of Exogenous and Endogenous Proteins
by Luiza Garaeva, Eugene Tolstyko, Elena Putevich, Yury Kil, Anastasiia Spitsyna, Svetlana Emelianova, Anastasia Solianik, Eugeny Yastremsky, Yuri Garmay, Elena Komarova, Elena Varfolomeeva, Anton Ershov, Irina Sizova, Evgeny Pichkur, Ilya A. Vinnikov, Varvara Kvanchiani, Alina Kilasoniya Marfina, Andrey L. Konevega and Tatiana Shtam
Plants 2025, 14(15), 2354; https://doi.org/10.3390/plants14152354 - 31 Jul 2025
Viewed by 505
Abstract
Extracellular vesicles (EVs), nanoscale membrane-enclosed particles, are natural carriers of proteins and nucleic acids. Microalgae are widely used as a source of bioactive substances in the food and cosmetic industries and definitely have a potential to be used as the producers of EVs [...] Read more.
Extracellular vesicles (EVs), nanoscale membrane-enclosed particles, are natural carriers of proteins and nucleic acids. Microalgae are widely used as a source of bioactive substances in the food and cosmetic industries and definitely have a potential to be used as the producers of EVs for biomedical applications. In this study, the extracellular vesicles isolated from the culture medium of two unicellular microalgae, Chlamydomonas reinhardtii (Chlamy-EVs) and Parachlorella kessleri (Chlore-EVs), were characterized by atomic force microscopy (AFM), cryo-electronic microscopy (cryo-EM), and nanoparticle tracking analysis (NTA). The biocompatibility with human cells in vitro (HEK-293T, DF-2 and A172) and biodistribution in mouse organs and tissues in vivo were tested for both microalgal EVs. An exogenous therapeutic protein, human heat shock protein 70 (HSP70), was successfully loaded to Chlamy- and Chlore-EVs, and its efficient delivery to human glioma and colon carcinoma cell lines has been confirmed. Additionally, in order to search for potential therapeutic biomolecules within the EVs, their proteomes have been characterized. A total of 105 proteins were identified for Chlamy-EVs and 33 for Chlore-EVs. The presence of superoxide dismutase and catalase in the Chlamy-EV constituents allows for considering them as antioxidant agents. The effective delivery of exogenous cargo to human cells and the possibility of the particle yield optimization by varying the microalgae growth conditions make them favorable producers of EVs for biotechnology and biomedical application. Full article
(This article belongs to the Section Plant Cell Biology)
Show Figures

Figure 1

22 pages, 1588 KiB  
Article
Scaffold-Free Functional Deconvolution Identifies Clinically Relevant Metastatic Melanoma EV Biomarkers
by Shin-La Shu, Shawna Benjamin-Davalos, Xue Wang, Eriko Katsuta, Megan Fitzgerald, Marina Koroleva, Cheryl L. Allen, Flora Qu, Gyorgy Paragh, Hans Minderman, Pawel Kalinski, Kazuaki Takabe and Marc S. Ernstoff
Cancers 2025, 17(15), 2509; https://doi.org/10.3390/cancers17152509 - 30 Jul 2025
Viewed by 501
Abstract
Background: Melanoma metastasis, driven by tumor microenvironment (TME)-mediated crosstalk facilitated by extracellular vesicles (EVs), remains a major therapeutic challenge. A critical barrier to clinical translation is the overlap in protein cargo between tumor-derived and healthy cell EVs. Objective: To address this, we developed [...] Read more.
Background: Melanoma metastasis, driven by tumor microenvironment (TME)-mediated crosstalk facilitated by extracellular vesicles (EVs), remains a major therapeutic challenge. A critical barrier to clinical translation is the overlap in protein cargo between tumor-derived and healthy cell EVs. Objective: To address this, we developed Scaffold-free Functional Deconvolution (SFD), a novel computational approach that leverages a comprehensive healthy cell EV protein database to deconvolute non-oncogenic background signals. Methods: Beginning with 1915 proteins (identified by MS/MS analysis on an Orbitrap Fusion Lumos Mass Spectrometer using the IonStar workflow) from melanoma EVs isolated using REIUS, SFD applies four sequential filters: exclusion of normal melanocyte EV proteins, prioritization of metastasis-linked entries (HCMDB), refinement via melanocyte-specific databases, and validation against TCGA survival data. Results: This workflow identified 21 high-confidence targets implicated in metabolic-associated acidification, immune modulation, and oncogenesis, and were analyzed for reduced disease-free and overall survival. SFD’s versatility was further demonstrated by surfaceome profiling, confirming enrichment of H7-B3 (CD276), ICAM1, and MIC-1 (GDF-15) in metastatic melanoma EV via Western blot and flow cytometry. Meta-analysis using Vesiclepedia and STRING categorized these targets into metabolic, immune, and oncogenic drivers, revealing a dense interaction network. Conclusions: Our results highlight SFD as a powerful tool for identifying clinically relevant biomarkers and therapeutic targets within melanoma EVs, with potential applications in drug development and personalized medicine. Full article
(This article belongs to the Section Methods and Technologies Development)
Show Figures

Figure 1

13 pages, 8639 KiB  
Article
In-Depth Characterization of L1CAM+ Extracellular Vesicles as Potential Biomarkers for Anti-CD20 Therapy Response in Relapsing–Remitting Multiple Sclerosis
by Shamundeeswari Anandan, Karina Maciak, Regina Breinbauer, Laura Otero-Ortega, Giancarlo Feliciello, Nataša Stojanović Gužvić, Oivind Torkildsen and Kjell-Morten Myhr
Int. J. Mol. Sci. 2025, 26(15), 7213; https://doi.org/10.3390/ijms26157213 - 25 Jul 2025
Viewed by 1157
Abstract
The effective suppression of inflammation using disease-modifying therapies is essential in the treatment of multiple sclerosis (MS). Anti-CD20 monoclonal antibodies are commonly used long-term as maintenance therapies, largely due to the lack of reliable biomarkers to guide dosing and evaluate treatment response. However, [...] Read more.
The effective suppression of inflammation using disease-modifying therapies is essential in the treatment of multiple sclerosis (MS). Anti-CD20 monoclonal antibodies are commonly used long-term as maintenance therapies, largely due to the lack of reliable biomarkers to guide dosing and evaluate treatment response. However, prolonged use increases the risk of infections and other immune-mediated side effects. The unique ability of brain-derived blood extracellular vesicles (EVs) to cross the blood–brain barrier and reflect the central nervous system (CNS) immune status has sparked interest in their potential as biomarkers. This study aimed to assess whether blood-derived L1CAM+ EVs could serve as biomarkers of treatment response to rituximab (RTX) in patients with relapsing-remitting MS (RRMS). Serum samples (n = 25) from the baseline (month 0) and after 6 months were analyzed from the RTX arm of the ongoing randomized clinical trial OVERLORD-MS (comparing anti-CD20 therapies in RRMS patients) and were compared with serum samples from healthy controls (n = 15). Baseline cerebrospinal fluid (CSF) samples from the same study cohort were also included. EVs from both serum and CSF samples were characterized, considering morphology, size, and concentration, using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). The immunophenotyping of EV surface receptors was performed using flow cytometry with the MACSPlex exosome kit, while label-free quantitative proteomics of EV protein cargo was conducted using a proximity extension assay (PEA). TEM confirmed the presence of EVs with the expected round morphology with a diameter of 50–150 nm. NTA showed significantly higher concentrations of L1CAM+ EVs (p < 0.0001) in serum total EVs and EBNA1+ EVs (p < 0.01) in serum L1CAM+ EVs at baseline (untreated) compared to in healthy controls. After six months of RTX therapy, there was a significant reduction in L1CAM+ EV concentration (p < 0.0001) and the downregulation of TNFRSF13B (p = 0.0004; FC = −0.49) in serum total EVs. Additionally, non-significant changes were observed in CD79B and CCL2 levels in serum L1CAM+ EVs at baseline compared to in controls and after six months of RTX therapy. In conclusion, L1CAM+ EVs in serum showed distinct immunological profiles before and after rituximab treatment, underscoring their potential as dynamic biomarkers for individualized anti-CD20 therapy in MS. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

12 pages, 1604 KiB  
Article
Extracellular Vesicles of Adipose Multipotent Mesenchymal Stromal Cells Propagate Senescent Phenotype by Affecting PTEN Nuclear Import
by Elizaveta Chechekhina, Semyon Kamenkov, Vadim Chechekhin, Anna Zinoveva, Elizaveta Bakhchinyan, Anastasia Efimenko, Natalia Kalinina, Vsevolod Tkachuk, Konstantin Kulebyakin and Pyotr Tyurin-Kuzmin
Int. J. Mol. Sci. 2025, 26(15), 7164; https://doi.org/10.3390/ijms26157164 - 24 Jul 2025
Viewed by 368
Abstract
Replicative or stress-induced senescence disrupts the functioning of multipotent mesenchymal stromal cells (MSCs) required for tissue renewal and regeneration. Aged MSCs demonstrate reduced proliferation, impaired differentiation, and aberrant secretory activity, defined as “senescence-associated secretory phenotype” (SASP). SASP is characterized by elevated secretion of [...] Read more.
Replicative or stress-induced senescence disrupts the functioning of multipotent mesenchymal stromal cells (MSCs) required for tissue renewal and regeneration. Aged MSCs demonstrate reduced proliferation, impaired differentiation, and aberrant secretory activity, defined as “senescence-associated secretory phenotype” (SASP). SASP is characterized by elevated secretion of proinflammatory cytokines and specific extracellular vesicles (SASP-EVs), which affect the cellular microenvironment and promote tissue dysfunction. However, molecular mechanisms responsible for senescent phenotype propagation remain largely obscure. Earlier, we demonstrated suppression of adipogenic differentiation and insulin sensitivity of young MSCs by SASP-EVs. In this study, we elucidated potential mechanisms underlying SASP-EVs’ effects on MSCs. Bioinformatic analysis revealed that insulin signaling components are the most probable targets of SASP-EVs microRNA cargo. We demonstrated that SASP-EVs downregulated intracellular AGO1 levels, but surprisingly, PTEN levels were upregulated. Specifically, the increase in PTEN content was provided by its nuclear fraction. We have found that the intracellular PTEN distribution in young MSCs treated by SASP-EVs was similar to senescent MSCs. Furthermore, PTEN upregulation was accompanied by increased PTENP1 expression—a molecular sponge for PTEN-targeting microRNAs. Our findings indicate that nuclear PTEN could be a hallmark of senescent MSCs, and SASP-EVs propagate the senescent phenotype in young MSCs by promoting PTEN nuclear localization. Full article
Show Figures

Figure 1

18 pages, 2571 KiB  
Article
Selective MicroRNA Packaging Reveals Distinct Core Signatures in Human Mesenchymal-Stromal-Cell-Derived Extracellular Vesicles
by Rachel E. Crossland, Clara Sanjurjo-Rodríguez, Monica Reis, Anne M. Dickinson, Elena Jones and Xiao-Nong Wang
Int. J. Mol. Sci. 2025, 26(14), 7010; https://doi.org/10.3390/ijms26147010 - 21 Jul 2025
Viewed by 485
Abstract
Mesenchymal stromal cells (MSCs) have demonstrated therapeutic efficacy across numerous clinical applications, with evidence suggesting their paracrine effects, particularly through extracellular vesicles (EVs), possibly driving functional outcomes. In this study we perform the comprehensive characterization of microRNA expression profiles in human MSC-derived EVs [...] Read more.
Mesenchymal stromal cells (MSCs) have demonstrated therapeutic efficacy across numerous clinical applications, with evidence suggesting their paracrine effects, particularly through extracellular vesicles (EVs), possibly driving functional outcomes. In this study we perform the comprehensive characterization of microRNA expression profiles in human MSC-derived EVs (MSC-EV) compared to their parental cells, cultured under clinically relevant xeno-free conditions. MSCs were isolated from the bone marrows of healthy donors and characterised according to the International Society for Cellular Therapy criteria, while MSC-EVs were isolated using differential ultracentrifugation and validated according to the International Society for Extracellular Vesicle guidelines. NanoString profiling identified 590 mature microRNAs expressed across both populations, with 42 being significantly differentially expressed between MSC-EVs and parental MSCs. Five microRNAs were distinctly highly expressed in MSCs and five in MSC-EVs, while fifteen of the top twenty most abundant microRNAs showed high expression in both populations. MicroRNA expression patterns were validated in an independent cohort. Functional pathway analysis of differentially expressed microRNAs showed enrichment of key biological processes including cell proliferation, differentiation, and immune regulation. This standardised profiling approach develops our understanding of MSC/MSC-EV microRNA cargo, using a transparent methodological approach that allows for the improved comparability of datasets for the development and advancement of MSC-EV therapeutics. Full article
(This article belongs to the Special Issue MicroRNA Regulation in Human Health and Diseases)
Show Figures

Graphical abstract

19 pages, 2051 KiB  
Article
Urinary Extracellular Vesicle Signatures as Biomarkers in Prostate Cancer Patients
by Sigrun Lange, Darryl Ethan Bernstein, Nikolay Dimov, Srinivasu Puttaswamy, Ian Johnston, Igor Kraev, Sarah R. Needham, Nikhil Vasdev and Jameel M. Inal
Int. J. Mol. Sci. 2025, 26(14), 6895; https://doi.org/10.3390/ijms26146895 - 18 Jul 2025
Viewed by 785
Abstract
Urinary extracellular vesicles (U-EVs) are gaining increasing interest as non-invasive liquid biopsy tools for clinical use. Prostate cancer (PCa) is amongst the highest cancer-related cause of death in men, and therefore, the identification of non-invasive robust biomarkers is of high importance. This study [...] Read more.
Urinary extracellular vesicles (U-EVs) are gaining increasing interest as non-invasive liquid biopsy tools for clinical use. Prostate cancer (PCa) is amongst the highest cancer-related cause of death in men, and therefore, the identification of non-invasive robust biomarkers is of high importance. This study assessed U-EV profiles from individuals affected by PCa at Gleason scores 6–9, compared with healthy controls. U-EVs were characterised and assessed for proteomic cargo content by LC-MS/MS analysis. The U-EV proteomes were compared for enrichment of gene ontology (GO), KEGG, and Reactome pathways, as well as disease–gene associations. U-EVs ranged in size from 50 to 350 nm, with the majority falling within the 100–200 nm size range for all groups. U-EV protein cargoes from the PCa groups differed significantly from healthy controls, with 16 protein hits unique to the GS 6–7 and 88 hits to the GS 8–9 U-EVs. Pathway analysis showed increased enrichment in the PCa U-EVs of biological process GO (5 and 37 unique to GS 6–7 and GS 8–9, respectively), molecular function GO (3 and 6 unique to GS 6–7 and GS 8–9, respectively), and cellular component GO (10 and 22 unique to GS 6–7 and GS 8–9, respectively) pathways. A similar increase was seen for KEGG pathways (11 unique to GS 8–9) and Reactome pathways (102 unique to GS 8–9). Enrichment of disease–gene associations was also increased in the PCa U-EVs, with highest differences for the GS 8–9 U-EVs (26 unique terms). The pathway enrichment in the PCa U-EVs was related to several key inflammatory, cell differentiation, cell adhesion, oestrogen signalling, and infection pathways. Unique GO and KEGG pathways enriched for the GS 8–9 U-EVs were associated with cell–cell communication, immune and stress responses, apoptosis, peptidase activity, antioxidant activity, platelet aggregation, mitosis, proteasome, mRNA stability oxytocin signalling, cardiomyopathy, and several neurodegenerative diseases. Our findings highlight U-EVs as biomarkers to inform disease pathways in prostate cancer patients and offer a non-invasive biomarker tool for clinical use. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Functions of Extracellular Vesicles)
Show Figures

Figure 1

18 pages, 2644 KiB  
Article
Exploring the Potential of Extracellular Vesicles from Atlantic Cod (Gadus morhua L.) Serum and Mucus for Wound Healing In Vitro
by Stefania D’Alessio, Igor Kraev, Bergljót Magnadóttir and Sigrun Lange
Biology 2025, 14(7), 870; https://doi.org/10.3390/biology14070870 - 17 Jul 2025
Viewed by 1424
Abstract
Novel therapeutic approaches for wound healing have included biomaterials from the Atlantic cod (Gadus morhua L.), with promising results in wound management. The use of extracellular vesicles (EVs), which can be isolated from cod biofluids, remains to be studied. EVs play key [...] Read more.
Novel therapeutic approaches for wound healing have included biomaterials from the Atlantic cod (Gadus morhua L.), with promising results in wound management. The use of extracellular vesicles (EVs), which can be isolated from cod biofluids, remains to be studied. EVs play key roles in cellular communication, and their use both as biomarkers and as therapeutic agents is widely reported in human pathologies, particularly with respect to mesenchymal stem cells. This pilot study characterized the total proteomic cargo content of EVs from cod serum and mucus and assessed the EVs’ potential for regenerative activity in wound-healing processes, using human and mouse fibroblast and keratinocyte in vitro scratch injury models. The pro-regenerative potential of both cod serum EVs and mucus EVs was identified, with differing capacities for accelerating wound closure in fibroblast and keratinocyte cells. This was further supported by varying effects of the cod serum EVs and mucus EVs on cellular vimentin and FGF-2 levels. The serum EV and mucus EV protein cargoes differed with respect to abundance of protein hits and associated enriched functional GO and KEGG pathways, but both were associated with immune, stress and wound-healing processes. Cod EVs may present as innovative therapeutic options for regenerative medicine applications, and our reported findings provide valuable insights for future in-depth studies. Full article
Show Figures

Graphical abstract

26 pages, 1408 KiB  
Review
Liposomes and Extracellular Vesicles as Distinct Paths Toward Precision Glioma Treatment
by Wiktoria Fraczek, Maciej Szmidt, Kacper Kregielewski and Marta Grodzik
Int. J. Mol. Sci. 2025, 26(14), 6775; https://doi.org/10.3390/ijms26146775 - 15 Jul 2025
Viewed by 470
Abstract
Glioblastoma multiforme (GBM), the most aggressive and therapy-resistant glioma subtype, remains an urgent clinical challenge due to its invasive nature, molecular heterogeneity, and the protective constraints of the blood–brain barrier (BBB). Liposomes and extracellular vesicles (EVs) have emerged as two of the most [...] Read more.
Glioblastoma multiforme (GBM), the most aggressive and therapy-resistant glioma subtype, remains an urgent clinical challenge due to its invasive nature, molecular heterogeneity, and the protective constraints of the blood–brain barrier (BBB). Liposomes and extracellular vesicles (EVs) have emerged as two of the most promising nanocarrier systems capable of overcoming these limitations through improved drug delivery and cellular targeting. Their applications in glioma therapy span chemotherapy, immunotherapy, and gene therapy, each presenting distinct advantages and mechanisms of action. Liposomes offer structural flexibility, controlled release, and a well-established clinical framework, while EVs provide innate biocompatibility, low immunogenicity, and the ability to mimic natural intercellular communication. Both systems demonstrate the capacity to traverse the BBB and selectively accumulate in tumor tissue, yet they differ in scalability, cargo loading efficiency, and translational readiness. Comparative evaluation of their functions across therapeutic modalities reveals complementary strengths that may be leveraged in the development of more effective, targeted strategies for glioma treatment. Full article
(This article belongs to the Special Issue Molecular Advances in Liposome-Based Drug Delivery Systems)
Show Figures

Figure 1

21 pages, 2638 KiB  
Article
Inhibiting miR-200a-3p Increases Sirtuin 1 and Mitigates Kidney Injury in a Tubular Cell Model of Diabetes and Hypertension-Related Renal Damage
by Olga Martinez-Arroyo, Ana Flores-Chova, Marta Mendez-Debaets, Laia Garcia-Ferran, Lesley Escrivá, Maria Jose Forner, Josep Redón, Raquel Cortes and Ana Ortega
Biomolecules 2025, 15(7), 995; https://doi.org/10.3390/biom15070995 - 11 Jul 2025
Viewed by 487
Abstract
Hypertension and diabetes mellitus are key contributors to kidney damage, with the renal tubule playing a central role in the progression of kidney disease. MicroRNAs have important regulatory roles in renal injury and are among the most abundant cargos within extracellular vesicles (EVs), [...] Read more.
Hypertension and diabetes mellitus are key contributors to kidney damage, with the renal tubule playing a central role in the progression of kidney disease. MicroRNAs have important regulatory roles in renal injury and are among the most abundant cargos within extracellular vesicles (EVs), emerging as novel kidney disease biomarkers and therapeutic tools. Previously, we identified miR-200a-3p and its target SIRT1 as having a potential role in kidney injury. We aimed to evaluate miR-200a-3p levels in EVs from patient’s urine and delve into its function in causing tubular injury. We quantified miR-200a-3p urinary EV levels in hypertensive patients with and without diabetes (n = 69), 42 of which were with increased urinary albumin excretion (UAE). We analysed miR-200a-3p levels in EVs and cellular pellets, as well as their targets at mRNA and protein levels in renal tubule cells (RPTECs) subjected to high glucose and Angiotensin II treatments, and observed their influence on apoptosis, RPTEC markers and tubular injury markers. We conducted microRNA mimic and inhibitor transfections in treated RPTECs. Our findings revealed elevated miR-200a-3p levels in increased UAE patient urinary EVs, effectively discriminating UAE (AUC of 0.75, p = 0.003). In vitro, miR-200a-3p and renal injury markers increased, while RPTEC markers, SIRT1, and apoptosis decreased under treatments. Experiments using miR-200a-3p mimics and inhibitors revealed a significant impact on SIRT1 and decrease in tubular damage through miR-200a-3p inhibition. Increased levels of miR-200a-3p emerge as a potential disease marker, and its inhibition provides a therapeutic target aimed at reducing renal tubular damage linked to hypertension and diabetes. Full article
(This article belongs to the Special Issue New Insights into Kidney Disease Development and Therapy Strategies)
Show Figures

Graphical abstract

Back to TopTop