1
|
Aldali F, Deng C, Nie M, Chen H. Advances in therapies using mesenchymal stem cells and their exosomes for treatment of peripheral nerve injury: state of the art and future perspectives. Neural Regen Res 2025; 20:3151-3171. [PMID: 39435603 PMCID: PMC11881730 DOI: 10.4103/nrr.nrr-d-24-00235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 07/26/2024] [Accepted: 08/26/2024] [Indexed: 10/23/2024] Open
Abstract
"Peripheral nerve injury" refers to damage or trauma affecting nerves outside the brain and spinal cord. Peripheral nerve injury results in movements or sensation impairments, and represents a serious public health problem. Although severed peripheral nerves have been effectively joined and various therapies have been offered, recovery of sensory or motor functions remains limited, and efficacious therapies for complete repair of a nerve injury remain elusive. The emerging field of mesenchymal stem cells and their exosome-based therapies hold promise for enhancing nerve regeneration and function. Mesenchymal stem cells, as large living cells responsive to the environment, secrete various factors and exosomes. The latter are nano-sized extracellular vesicles containing bioactive molecules such as proteins, microRNA, and messenger RNA derived from parent mesenchymal stem cells. Exosomes have pivotal roles in cell-to-cell communication and nervous tissue function, offering solutions to changes associated with cell-based therapies. Despite ongoing investigations, mesenchymal stem cells and mesenchymal stem cell-derived exosome-based therapies are in the exploratory stage. A comprehensive review of the latest preclinical experiments and clinical trials is essential for deep understanding of therapeutic strategies and for facilitating clinical translation. This review initially explores current investigations of mesenchymal stem cells and mesenchymal stem cell-derived exosomes in peripheral nerve injury, exploring the underlying mechanisms. Subsequently, it provides an overview of the current status of mesenchymal stem cell and exosome-based therapies in clinical trials, followed by a comparative analysis of therapies utilizing mesenchymal stem cells and exosomes. Finally, the review addresses the limitations and challenges associated with use of mesenchymal stem cell-derived exosomes, offering potential solutions and guiding future directions.
Collapse
Affiliation(s)
- Fatima Aldali
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chunchu Deng
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Mingbo Nie
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hong Chen
- Department of Rehabilitation Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
2
|
Picchio V, Pontecorvi V, Dhori X, Bordin A, Floris E, Cozzolino C, Frati G, Pagano F, Chimenti I, De Falco E. The emerging role of artificial intelligence applied to exosome analysis: from cancer biology to other biomedical fields. Life Sci 2025; 375:123752. [PMID: 40409585 DOI: 10.1016/j.lfs.2025.123752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 05/06/2025] [Accepted: 05/20/2025] [Indexed: 05/25/2025]
Abstract
In recent years, exosomes versatility has prompted their study in the biomedical field for diagnostic, prognostic, and therapeutic applications. Exosomes are bi-lipid small extracellular vesicles (30-150 nm) secreted by various cell types, containing proteins, lipids, and DNA/RNA. They mediate intercellular communication and can influence multiple human physiological and pathological processes. So far, exosome analysis has revealed their role as promising diagnostic tools for human pathologies. Concurrently, artificial intelligence (AI) has revolutionised multiple sectors, including medicine, owing to its ability to analyse large datasets and identify complex patterns. The combination of exosome analysis with AI processing has displayed a novel diagnostic approach for cancer and other diseases. This review explores the current applications and prospects of the combined use of exosomes and AI in medicine. Firstly, we provide a biological overview of exosomes and their relevance in cancer biology. Then we explored exosome isolation techniques and Raman spectroscopy/SERS analysis. Finally, we present a summarised essential guide of AI methods for non-experts, emphasising the advancements made in AI applications for exosome characterisation and profiling in oncology research, as well as in other human diseases.
Collapse
Affiliation(s)
- Vittorio Picchio
- Department of Angio Cardio Neurology, IRCCS Neuromed, 86077 Pozzilli, Italy
| | - Virginia Pontecorvi
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Xhulio Dhori
- CINECA, Super Computing Applications and Innovation Department, 000185 Roma, Italy
| | - Antonella Bordin
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Erica Floris
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Claudia Cozzolino
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Giacomo Frati
- Department of Angio Cardio Neurology, IRCCS Neuromed, 86077 Pozzilli, Italy; Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy
| | - Francesca Pagano
- Institute of Biochemistry and Cell Biology, National Council of Research (IBBC-CNR), 00015 Monterotondo,Italy
| | - Isotta Chimenti
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy; Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy.
| | - Elena De Falco
- Department of Medical Surgical Sciences and Biotechnologies, Sapienza University, 04100 Latina, Italy; Maria Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Italy
| |
Collapse
|
3
|
Teerapong P, Yang YT, Bhargava Sreerangaraja Urs D, Liu JJ, Kao SH. Umbilical cord-derived mesenchymal stem cells secretomes promote embryo development and implantation. Life Sci 2025; 374:123693. [PMID: 40348173 DOI: 10.1016/j.lfs.2025.123693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/22/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
AIMS Successful implantation relies on high-quality blastocysts, uterine receptivity, and effective embryo-endometrium communication. This study investigated the effects of umbilical cord-derived mesenchymal stem cells (UC-MSC) secretomes on embryo development and implantation. MAIN METHODS Trophoblastic spheroids and murine embryos were used to evaluate the impact of UC-MSC secretomes. Embryos obtained through superovulation were cultured in vitro and divided into five groups: a control group and four experimental groups treated with varying concentrations of UC-MSC secretomes (2.5, 5, 10, and 50 μg/mL). Embryo development competence and implantation potential were assessed in each group, and the expression levels of related genes were analyzed. KEY FINDINGS Supplementation with UC-MSC secretomes significantly enhanced trophoblast cell migration. It also stimulated endometrial cell proliferation and upregulated key implantation-related genes (LIF, LIFR, VEGFA, ITGB3, and ITGAV), improving endometrial receptivity and adhesion in trophoblastic spheroid co-cultures. While morulation rates of murine embryos remained unchanged, UC-MSC secretomes supplement significantly increased blastulation, pluripotency gene expression, and hatching rates. Supplementation with 10 and 50 μg/mL significantly increased blastocyst diameter and blastomere number, as well as embryo adhesion, outgrowth areas, and implantation rates. Additionally, growth factor analysis showed elevated VEGF-A and PDGF-AA levels in the culture media. SIGNIFICANCE This study demonstrates that UC-MSC secretomes enhance both embryo development and endometrial cell function, facilitating implantation potential. These findings suggest their potential utility in supporting preimplantation embryos and improving maternal endometrial receptivity in ART.
Collapse
Affiliation(s)
- Prompunya Teerapong
- Ph. D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Veterinary Research and Development Center-Lower Northern Region, National Institute of Animal Health, Bangkok 10900, Thailand
| | - Yuan-Ting Yang
- Biomedical Department, High-Tek Harness Enterprise Co., Ltd., Taipei 115, Taiwan
| | - Dilip Bhargava Sreerangaraja Urs
- Ph. D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Jun-Jen Liu
- Ph. D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan
| | - Shu-Huei Kao
- Ph. D. Program in Medical Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei 110, Taiwan; Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei 110, Taiwan.
| |
Collapse
|
4
|
Cao LM, Qiu YZ, Li ZZ, Wang GR, Xiao Y, Luo HY, Liu B, Wu Q, Bu LL. Extracellular Vesicles: Hermes between cancers and lymph nodes. Cancer Lett 2025; 623:217735. [PMID: 40268131 DOI: 10.1016/j.canlet.2025.217735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/16/2025] [Accepted: 04/19/2025] [Indexed: 04/25/2025]
Abstract
Cancer is one of the main causes of death and a major obstacle to increasing life expectancy in all countries of the world. Lymph node metastasis (LNM) of in cancer patients indicates poor prognosis and it is an important indication to determine the therapeutic regime. Therefore, more attention should be given to the molecular mechanics of tumor lymphangiogenesis and LNM. Extracellular vesicles (EVs) are nanoscale cargo-bearing membrane vesicles that can serve as key mediators for the intercellular communication. Like Hermes, the messenger of the Greek gods, EVs can be secreted by tumor cells to regulate the LNM process. Many evidence has proved the clinical correlation between EVs and LNM in various cancer types. EVs plays an active role in the process of metastasis by expressing its connotative molecules, including proteins, nucleic acids, and metabolites. However, the clear role of EVs in the process of cancer LNM has not been thoroughly studied yet. In this review, we will summarize the clinical and mechanical findings of EVs regulating role on cancer LNM, and discuss the advanced modification of the research proposal. We propose the "PUMP" principle of EVs in LNM, including Preparation, Unleash, Migration, and Planting.
Collapse
Affiliation(s)
- Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yu-Zhong Qiu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Guang-Rui Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Han-Yue Luo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Qiuji Wu
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behavior, Hubei Provincial Clinical Research Center for Cancer, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China.
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
5
|
Hicks R, Gozal D, Ahmed S, Khalyfa A. Interplay between gut microbiota and exosome dynamics in sleep apnea. Sleep Med 2025; 131:106493. [PMID: 40203611 DOI: 10.1016/j.sleep.2025.106493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 03/19/2025] [Accepted: 03/29/2025] [Indexed: 04/11/2025]
Abstract
Sleep-disordered breathing (SDB) is characterized by recurrent reductions or interruptions in airflow during sleep, termed hypopneas and apneas, respectively. SDB impairs sleep quality and is linked to substantive health issues including cardiovascular and metabolic disorders, as well as cognitive decline. Recent evidence suggests a link between gut microbiota (GM) composition and sleep apnea. Indeed, GM, a community of microorganisms residing in the gut, has emerged as a potential player in various diseases, and several studies have identified associations between sleep apnea and GM diversity along with shifts in bacterial populations. Additionally, the concept of "leaky gut," a compromised intestinal barrier with potentially increased inflammation, has emerged as another key player in the potential bidirectional relationship between GM and sleep apnea. One of the potential effectors could be extracellular vesicles (EVs) underlying gut-brain communication pathways that are relevant to sleep regulation and function. Thus, therapeutic implications afforded by targeting the GM or exosomes for sleep apnea management have surfaced as promising areas of research. This review explores current understanding of the relationship between GM, exosomes and sleep apnea, highlighting key research dynamics and potential mechanisms. A comprehensive review of the literature was conducted, focusing on studies investigating GM composition, intestinal barrier function and gut-brain communication in relation to sleep apnea.
Collapse
Affiliation(s)
- Rebecca Hicks
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - David Gozal
- Department of Pediatrics and Office of the Dean, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Sarfraz Ahmed
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA
| | - Abdelnaby Khalyfa
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, WV, 25755, USA.
| |
Collapse
|
6
|
Nie P, Qin W, Nie WC, Li B. Progress in the application of mesenchymal stem cells to attenuate apoptosis in diabetic kidney disease. World J Diabetes 2025; 16:105711. [DOI: 10.4239/wjd.v16.i6.105711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/24/2025] [Accepted: 04/25/2025] [Indexed: 06/13/2025] Open
Abstract
Diabetic kidney disease (DKD) has a high incidence and mortality rate and lacks effective preventive and therapeutic methods. Apoptosis is one of the main reasons for the occurrence and development of DKD. Mesenchymal stem cells (MSCs) have shown great promise in tissue regeneration for DKD treatment and have protective effects against DKD, including decreased blood glucose and urinary protein levels and improved renal function. MSCs can directly differentiate into kidney cells or act via paracrine mechanisms to reduce apoptosis in DKD by modulating signaling pathways. MSC-derived extracellular vesicles (MSC-EVs) mitigate apoptosis and DKD-related symptoms by transferring miRNAs to target cells or organs. However, studies on the regulatory mechanisms of MSCs and MSC-EVs in apoptosis in DKD are insufficient. This review comprehensively examines the mechanisms of apoptosis in DKD and research progress regarding the roles of MSCs and MSC-EVs in the disease process.
Collapse
Affiliation(s)
- Ping Nie
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun 130041, Jilin Province, China
| | - Wei Qin
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun 130041, Jilin Province, China
| | - Wei-Chen Nie
- Basic Clinical Specialization in Integrative Chinese and Western Medicine, Changchun University of Chinese Medicine, Changchun 130117, Jilin Province, China
| | - Bing Li
- Department of Nephropathy, The Second Hospital of Jilin University, Changchun 130041, Jilin Province, China
| |
Collapse
|
7
|
Yu H, Xin C, Zhou Y, Ding X. Advances in the application of extracellular vesicles in precise diagnosis of pancreatic cancer. Eur J Med Res 2025; 30:478. [PMID: 40514731 DOI: 10.1186/s40001-025-02739-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 05/28/2025] [Indexed: 06/16/2025] Open
Abstract
Pancreatic cancer is a highly malignant tumor with poor prognosis, emphasizing the need for accurate early diagnosis. EVs, as mediators of intercellular communication, carry DNA, RNA, and proteins that show differential but not tumor-specific expression patterns in pancreatic cancer. Studies have shown that combining RNA markers in EVs (such as miRNA, circRNA, and lncRNA) with serum CA 19-9 testing can significantly enhance diagnostic accuracy for pancreatic cancer. EV-associated proteins have exhibited favorable diagnostic performance in early-stage pancreatic cancer in preliminary studies, though their clinical applicability remains to be further validated. Furthermore, mutations in KRAS, TP53, and SMAD4 genes within EVs offer a promising avenue for non-invasive liquid biopsy. However, challenges such as standardization, low sensitivity, and specificity still hinder the clinical application of EVs. Future research should focus on strategies including multi-omics integration, AI-assisted analysis, multi-marker combined detection, and large-scale clinical validation to further improve the diagnostic capability for pancreatic cancer. Overcoming these obstacles may position EVs as a vital tool in the diagnosis of pancreatic cancer.
Collapse
Affiliation(s)
- Haiyang Yu
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Congling Xin
- Department of Gynecology, Fudan University Shanghai Cancer Center Minhang District, Shanghai, 200240, China
| | - Yu Zhou
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiaoyi Ding
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
8
|
Gabaran SG, Nejati V, Dilsiz N, Rezaie J. An updated review on the inhibition of exosome biogenesis, release, and uptake: a potential anticancer approach. Biochem Pharmacol 2025; 239:117019. [PMID: 40499840 DOI: 10.1016/j.bcp.2025.117019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Revised: 04/20/2025] [Accepted: 06/02/2025] [Indexed: 06/16/2025]
Abstract
Extracellular vesicles, exosomes, have garnered significant attention in the field of cancer therapy, one of the world's deadliest diseases. Exosomes from cancer cells participate in the development of cancer. Inhibition of exosome biogenesis may be a promising way to combat cancer. Numerous drugs and agents have been assessed to inhibit exosome biogenesis, release, and uptake, which are the main factors contributing to cancer progression. Different drugs target several intracellular mechanisms to stop the exosome signalling pathway. They affect various intracellular pathways; for example, they can disrupt the endosomal sorting complex or interfere with the intracellular trafficking of exosomes Furthermore, some of them suppress or modulate genes and proteins involved in exosome generation and release. Exosome inhibition may also be associated with different side and non-targeting effects. Pre-clinical studies show promising outcomes; however, some challenges need to be addressed in future studies. This review describes the properties of exosome inhibitor agents, focusing on the specific pathways involved in exosome biogenesis, release, and uptake.
Collapse
Affiliation(s)
| | - Vahid Nejati
- Department of Biology, Urmia University, Urmia, Iran
| | - Nihat Dilsiz
- University of Health Sciences, Experimental Medicine Practice and Research Center (EMPRC), Validebag Research Park, Istanbul, Turkey
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
9
|
Lo KJ, Wang MH, Kuo CY, Pan MH. Optimizing Isolation Methods and Exploring the Therapeutic Potential of Lotus-Derived Extracellular Vesicles in Modulating Inflammation and Promoting Wound Healing. ACS Biomater Sci Eng 2025. [PMID: 40490711 DOI: 10.1021/acsbiomaterials.5c00377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
In the past decade, with the rise of research on plant-derived extracellular vesicles (PDEVs), scientists have been continuously exploring the bioactivity of PDEVs. Many PDEVs have been shown to possess a variety of biological activities. Given that the specific characteristics of EVs are believed to be related to their source cells, PDEVs from traditional Chinese medicinal herbs hold significant potential for development. In this study, lotus (Nelumbo nucifera Gaertn.) leaves were selected as the source of PDEVs, and the impact of different isolation methods on their characteristics was evaluated, while their potential biological activities were also assessed. Lotus-derived EVs (LDEVs) were isolated by using tangential flow filtration (TFF), ultracentrifugation (UC), density gradient ultracentrifugation (DGU), and size-exclusion chromatography (SEC), respectively. The mean sizes of LDEVs isolated by various methods were in the range of 130-160 nm. Although the LDEVs isolated by the TFF method had a lower zeta potential, it exhibited the highest purity, with a yield of 3.69 ± 0.43 × 109 particles/g lotus leaves. Notably, LDEVs isolated by different methods all demonstrated the ability to attenuate LPS-induced inflammation in RAW264.7 cells, significantly decreasing the nitrite concentration in the culture medium. Furthermore, LDEVs also showed potential for wound healing, promoting the migration of HaCaT cells in vitro. LDEVs also demonstrated internalization by RAW264.7 and HaCaT cells. These results support the potential of LDEVs for biomedical applications while also suggesting that TFF is a promising and viable strategy for large-scale PDEV isolation.
Collapse
Affiliation(s)
- Kai-Jiun Lo
- Institute of Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan
| | - Mu-Hui Wang
- Department of Medical Research, National Taiwan University Hospital, Taipei 100225, Taiwan
| | - Ching-Yao Kuo
- BO HUI BIOTECH CO., LTD., New Taipei City 248016, Taiwan
| | - Min-Hsiung Pan
- Institute of Food Science and Technology, National Taiwan University, Taipei 10617, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
10
|
Voros C, Athanasiou D, Mavrogianni D, Varthaliti A, Bananis K, Athanasiou A, Athanasiou A, Papadimas G, Gkirgkinoudis A, Papapanagiotou I, Migklis K, Vaitsis D, Koulakmanidis AM, Mazis Kourakos D, Ivanidou S, Daskalaki MA, Theodora M, Antsaklis P, Loutradis D, Daskalakis G. Exosomal Communication Between Cumulus-Oocyte Complexes and Granulosa Cells: A New Molecular Axis for Oocyte Competence in Human-Assisted Reproduction. Int J Mol Sci 2025; 26:5363. [PMID: 40508172 PMCID: PMC12155520 DOI: 10.3390/ijms26115363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2025] [Revised: 05/26/2025] [Accepted: 06/02/2025] [Indexed: 06/16/2025] Open
Abstract
Exosomal microRNAs (ex-miRs), encapsulated in extracellular vesicles (EVs), play a vital role in facilitating paracrine communication among granulosa cells (GCs), cumulus cells (CCs), and the oocyte inside follicular fluid (FF). These small non-coding RNAs are crucial for regulating folliculogenesis, oocyte maturation, and early embryonic development via modulating intracellular signaling networks. Dysregulation o has been associated with reproductive disorders such as polycystic ovarian syndrome (PCOS), diminished ovarian reserve (DOR), and inadequate ovarian response (POR), impacting oocyte quality and fertility outcomes. This narrative review consolidates molecular data from current human and animal studies regarding ex-miR expression patterns, functional targets, and pathway involvement within the context of assisted reproductive technologies (ARTs). A literature-based analysis was undertaken, focusing on signaling pathways, pathogenic processes, and clinical implications. Specifically, ex-miRs-such as miR-21, miR-34c, miR-143-3p, miR-155-5p, miR-339-5p, and miR-424-5p-were identified as regulators of critical pathways including phosphoinositide 3-kinase (PI3K)-AKT, ERK1/2, TGF-β/SMAD, and Rb-E2F1. These ex-miRs regulate apoptosis, glycolysis, mitochondrial function, and cell cycle expansion to influence oocyte competence. Pathological patterns in PCOS and POR are associated with altered ex-miR expression that disrupts metabolic and developmental signaling. Research utilizing animal models confirmed that modifications in EV-associated miRNA influence in vitro maturation (IVM) efficiency and blastocyst quality. Ex-miRs serve as intriguing non-invasive biomarkers and potential therapeutic targets for ARTs. Their mechanical involvement in oocyte and follicular physiology positions them for integration into forthcoming precision-based infertility therapies. For its implementation in reproductive medicine, EV profiling requires standardization and further functional validation in clinical environments.
Collapse
Affiliation(s)
- Charalampos Voros
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.V.); (A.G.); (A.-M.K.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Diamantis Athanasiou
- IVF Athens Reproduction Center V. Athanasiou, 15123 Maroussi, Greece; (D.A.); (A.A.); (A.A.)
| | - Despoina Mavrogianni
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.V.); (A.G.); (A.-M.K.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Antonia Varthaliti
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.V.); (A.G.); (A.-M.K.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Kyriakos Bananis
- King’s College Hospitals NHS Foundation Trust, London SE5 9RS, UK;
| | - Antonia Athanasiou
- IVF Athens Reproduction Center V. Athanasiou, 15123 Maroussi, Greece; (D.A.); (A.A.); (A.A.)
| | - Aikaterini Athanasiou
- IVF Athens Reproduction Center V. Athanasiou, 15123 Maroussi, Greece; (D.A.); (A.A.); (A.A.)
| | - Georgios Papadimas
- Athens Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece; (G.P.); (I.P.); (K.M.); (D.V.); (D.M.K.); (S.I.); (D.L.)
| | - Athanasios Gkirgkinoudis
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.V.); (A.G.); (A.-M.K.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Ioannis Papapanagiotou
- Athens Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece; (G.P.); (I.P.); (K.M.); (D.V.); (D.M.K.); (S.I.); (D.L.)
| | - Kyriaki Migklis
- Athens Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece; (G.P.); (I.P.); (K.M.); (D.V.); (D.M.K.); (S.I.); (D.L.)
| | - Dimitrios Vaitsis
- Athens Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece; (G.P.); (I.P.); (K.M.); (D.V.); (D.M.K.); (S.I.); (D.L.)
| | - Aristotelis-Marios Koulakmanidis
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.V.); (A.G.); (A.-M.K.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Dimitris Mazis Kourakos
- Athens Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece; (G.P.); (I.P.); (K.M.); (D.V.); (D.M.K.); (S.I.); (D.L.)
| | - Sofia Ivanidou
- Athens Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece; (G.P.); (I.P.); (K.M.); (D.V.); (D.M.K.); (S.I.); (D.L.)
| | - Maria Anastasia Daskalaki
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.V.); (A.G.); (A.-M.K.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Marianna Theodora
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.V.); (A.G.); (A.-M.K.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Panagiotis Antsaklis
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.V.); (A.G.); (A.-M.K.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| | - Dimitrios Loutradis
- Athens Medical School, National and Kapodistrian University of Athens, 15772 Athens, Greece; (G.P.); (I.P.); (K.M.); (D.V.); (D.M.K.); (S.I.); (D.L.)
- Fertility Institute-Assisted Reproduction Unit, Paster 15, 11528 Athens, Greece
| | - Georgios Daskalakis
- 1st Department of Obstetrics and Gynecology, ‘Alexandra’ General Hospital, National and Kapodistrian University of Athens, 80 VasilissisSofias Avenue, 11528 Athens, Greece; (D.M.); (A.V.); (A.G.); (A.-M.K.); (M.A.D.); (M.T.); (P.A.); (G.D.)
| |
Collapse
|
11
|
Seth G, Singh S, Sharma G, Suvedi D, Kumar D, Nagraik R, Sharma A. Harnessing the power of stem cell-derived exosomes: a rejuvenating therapeutic for skin and regenerative medicine. 3 Biotech 2025; 15:184. [PMID: 40417660 PMCID: PMC12102458 DOI: 10.1007/s13205-025-04345-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 05/04/2025] [Indexed: 05/27/2025] Open
Abstract
Exosomes are small extracellular vesicles produced by most cell types and contain proteins, lipids, and nucleic acids (non-coding RNAs, mRNA, and DNA) that can be released by donor cells to influence the function of recipient cells. Skin photoaging is the premature aging of skin structures caused by prolonged exposure to ultraviolet (UV), as demonstrated by depigmentation, roughness, rhytides, elastosis, and precancerous alterations. Exosomes are associated with aging processes such as oxidative damage, inflammation, and senescence. Exosomes' anti-aging properties have been linked to various in vitro and preclinical investigations. There are still several unanswered questions about the use of MSC exosomes for skin rejuvenation, despite encouraging results. Uncertainty surrounds the precise processes by which exosomes stimulate the creation of collagen, skin tissue via a variety of mechanisms, including reduced matrix metalloproteinase (MMP) expression, increased collagen and elastin production, and modulation of intracellular signaling pathways and intercellular communication. These findings suggest the therapeutic potential of exosomes in skin aging. This review provides information on the molecular mechanisms and consequences of exosome anti-aging.
Collapse
Affiliation(s)
- Gracy Seth
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
| | - Siddharth Singh
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
| | - Geetansh Sharma
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
| | - Divyesh Suvedi
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
| | - Dinesh Kumar
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
| | - Rupak Nagraik
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
- Department of Biotechnology, Graphic Era (Deemed to Be University), Dehradun, 248002 India
| | - Avinash Sharma
- Faculty of Applied Sciences and Biotechnology, Shoolini University, Solan, Himachal Pradesh 173229 India
- Department of Biotechnology, Graphic Era (Deemed to Be University), Dehradun, 248002 India
| |
Collapse
|
12
|
Bavafa A, Izadpanahi M, Hosseini E, Hajinejad M, Abedi M, Forouzanfar F, Sahab-Negah S. Exosome: an overview on enhanced biogenesis by small molecules. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6473-6508. [PMID: 39862264 DOI: 10.1007/s00210-024-03762-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025]
Abstract
Exosomes are extracellular vesicles that received attention for their potential use in the treatment of various injuries. They communicate intercellularly by transferring genetic and bioactive molecules from parent cells. Although exosomes hold immense promise for treating neurodegenerative and oncological diseases, their actual clinical use is very limited because of their biogenesis and secretion. Recent studies have shown that small molecules can significantly enhance exosome biogenesis, thereby remarkably improving yield, functionality, and therapeutic effects. These molecules modulate critical pathways toward optimum exosome production in a mode that is either ESCRT dependent or ESCRT independent. Improved exosome biogenesis may provide new avenues for targeted cancer therapy, neuroprotection in neurodegenerative diseases, and regenerative medicine in wound healing. This review explores the role of small molecules in enhancing exosome biogenesis and secretion, highlights their underlying mechanisms, and discusses emerging clinical applications. By addressing current challenges and focusing on translational opportunities, this study provides a foundation for advancing cell-free therapies in regenerative medicine and beyond.
Collapse
Affiliation(s)
- Amir Bavafa
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Izadpanahi
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Hosseini
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mehrdad Hajinejad
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Qaen Faculty of Medical Sciences, Birjand University of Medical Sciences, Birjand, Iran
| | - Mahsa Abedi
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Institute for Molecular and Clinical Immunology, Medical Faculty, Otto-von-Guericke University, Magdeburg, Germany
| | - Fatemeh Forouzanfar
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Sajad Sahab-Negah
- Neuroscience Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
- Department of Neuroscience, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
- Shefa Neuroscience Research Center, Khatam Alanbia Hospital, Tehran, Iran.
| |
Collapse
|
13
|
Wang C, Zhou L, Kang X, Huang CH, Gao Z, Shen J, Wu S, Wu S, Cai Y, Chen W, Dai S, Chen P. A nanoplasmonic cell-on-a-chip for in situ monitoring of PD-L1 + exosome-mediated immune modulation. Biosens Bioelectron 2025; 277:117293. [PMID: 39999609 PMCID: PMC11996229 DOI: 10.1016/j.bios.2025.117293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 02/11/2025] [Accepted: 02/20/2025] [Indexed: 02/27/2025]
Abstract
Despite the transformative impact of immune checkpoint inhibitors (ICIs) targeting the PD-1/PD-L1 pathway in cancer therapy, up to 80% of patients fail to respond, necessitating reliable predictive biomarkers to guide treatment decisions. Recent studies highlight the critical role of tumor-derived exosomal PD-L1 in immune evasion, and its potential as a diagnostic and prognostic biomarker in cancer immunotherapy. However, significant challenges remain in elucidating the functional roles of PD-L1+ exosomes in immune suppression, as current methods lack the ability to precisely and simultaneously characterize and monitor exosome secretion and the corresponding immune modulation on site. To address this, we developed an integrated microfluidic platform that combines a digital nanoplasmonic immunoassay with a cell-on-a-chip system, enabling in situ monitoring of PD-L1+ exosome secretion and exosome-mediated T cell immune responses. This nanoplasmonic immunoassay integrated cell-on-a-chip (NIIC) creates a localized co-cultured microenvironment that facilitates exosome-mediated cellular interactions without direct contact. The NIIC employs machine-learning assisted signal processing for highly sensitive detection of both exosomes and cytokines, providing spatial and quantitative analysis of immune modulation in situ. Using this system, we demonstrated that PD-L1+ exosomes from cancer cells significantly suppressed IFN-γ and IL-2 secretion in neighboring T cells, offering direct insights into exosome-mediated immune suppression. The NIIC platform represents a powerful tool for advancing the understanding of exosome-driven immune modulation and holds potential for predicting clinical responses to anti-PD-1/PD-L1 therapies, paving the way for more personalized cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Chuanyu Wang
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Lang Zhou
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Xuejia Kang
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849; Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Chung-Hui Huang
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, 36849, USA
| | - Zhuangqiang Gao
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Jialiang Shen
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Shuai Wu
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Siqi Wu
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Yuxin Cai
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Weiqiang Chen
- Department of Mechanical and Aerospace Engineering, New York University, Brooklyn, NY, 11201, USA; Department of Biomedical Engineering, New York University, Brooklyn, NY, 11201, USA
| | - Siyuan Dai
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849
| | - Pengyu Chen
- Materials Engineering, Department of Mechanical Engineering, Auburn University, Auburn, AL, USA, 36849.
| |
Collapse
|
14
|
Widjaja Lomanto MY, Wanandi SI, Jayusman AM, Lukmanto D, Prayitno YH, Sutandyo N. Smoking induces different expression of miR-320b and miR-10b-5p in plasma extracellular vesicles of non-small cell lung cancer patients. THE JOURNAL OF LIQUID BIOPSY 2025; 8:100291. [PMID: 40224902 PMCID: PMC11984573 DOI: 10.1016/j.jlb.2025.100291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 04/15/2025]
Abstract
Background Previous studies found that cigarette smoke (CS) exposure could induce NSCLC malignancy and miRNA dysregulation. Yet, the association of CS-induced miRNA dysregulation and NSCLC malignancy has not been clearly understood. This study aimed to evaluate the effect of CS exposure in smokers on the expression of miR-10b-5p and miR-320b in extracellular vesicles (EVs) from NSCLC patients. Material and methods Bioinformatic analysis was conducted to validate miRNA candidates. Blood and tissue samples were collected from NSCLC patients (n = 21) with smoking and non-smoking history. EVs were isolated from plasma and miRNAs were extracted from the isolated EVs. The miRNAs relative expression was analyzed and compared. Results In silico analysis identified miR-320b and miR-10b-5p as potential biomarkers for diagnosing NSCLC in smokers. Experimental analysis revealed differential expression of EVs-associated miRNAs in NSCLC patients with smoking and non-smoking histories. EVs-associated miR-10b-5p was significantly overexpressed in smoker NSCLC patients (p = 0.000), while miR-320b expression was significantly lower in this group (p = 0.018). Additionally, smoking intensity influenced miRNA expression, with higher smoking intensity correlating with increased miR-10b-5p expression and decreased miR-320b expression. ROC analysis demonstrated that EVs were a superior source of miRNAs compared to plasma for NSCLC diagnostics. miR-10b-5p and miR-320b in EVs showed higher diagnostic performance (AUC 0.878; 0.739) compared to plasma (AUC 0.628; 0.559). Conclusion CS exposure induces different expression of miR-10b-5p and miR-320b in EVs of NSCLC patients with smoking history. EV-related miR-10b-5p and miR-320b showed potential to be utilized as prognostic biomarker for smokers NSCLC patients.
Collapse
Affiliation(s)
| | - Septelia Inawati Wanandi
- Master's Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Molecular Biology and Proteomics Core Facilities, Indonesian Medical Education and Research Institute (IMERI), Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | - Donny Lukmanto
- Laboratory of Advanced Vision Sciences, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Yuniar Harris Prayitno
- Department of Hematology and Medical Oncology, Dharmais Hospital National Cancer Center, Jakarta, Indonesia
| | - Noorwati Sutandyo
- Master's Program in Biomedical Sciences, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
- Department of Hematology and Medical Oncology, Dharmais Hospital National Cancer Center, Jakarta, Indonesia
| |
Collapse
|
15
|
Zhang H, Jiang N, Xu M, Jing D, Dong T, Liu Q, Lv Q, Huo R, Chen P, Li L, Wang X. M2 macrophage derived exosomal miR-20a-5p ameliorates trophoblast pyroptosis and placental injuries in obstetric antiphospholipid syndrome via the TXNIP/NLRP3 axis. Life Sci 2025; 370:123561. [PMID: 40127859 DOI: 10.1016/j.lfs.2025.123561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
AIM Obstetric antiphospholipid syndrome (OAPS) is a pregnancy-related complication characterized by trophoblast pyroptosis and placental injury induced by antiphospholipid antibodies (aPLs). M2-polarized macrophage-derived exosomes (M2-exos) exert anti-inflammatory, immunomodulatory, and growth-promoting effects in various autoimmune diseases and tumors. However, their role in OAPS is not yet clear. Therefore, in this study, we isolated M2-exos from M2 macrophages and investigated their effects on trophoblast proliferation, death, migration, invasion, and pyroptosis following stimulation using aPLs. MAIN METHODS First, we established an animal model of OAPS and thereafter treated the OAPS mice with exogenous M2-exos via injection through the tail vein. Then to clarify the roles of miR-20a-5p and thioredoxin-interacting protein (TXNIP) in OAPS, we performed gain- or loss-of-function assays, and used GraphPad Prism software to analyze the collected data with statistical significance set at P < 0.05. KEY FINDINGS MicroRNAs (miRNAs) sequencing revealed the enrichment of miR-20a-5p in M2-exos, and these M2-exos significantly alleviated aPLs-induced trophoblast dysfunction. Our results also indicated that M2-exos delivered miR-20a-5p to trophoblast cells directly targeted thioredoxin-interacting protein (TXNIP), and thus suppressed the TXNIP/NLRP3 pathway, reduced pyroptosis and inflammation in trophoblast cells, and improved placental function and fetal development. SIGNIFICANCE M2-exos improve pregnancy outcomes in OAPS via the miR-20a-5p/TXNIP/NLRP3 axis, and thus represent as a novel therapeutic approach for aPLs-induced OAPS.
Collapse
Affiliation(s)
- Hongyuan Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China
| | - Ning Jiang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Mingyang Xu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Die Jing
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Tingting Dong
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Qian Liu
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Feixian County People's Hospital, Linyi 273400, Shandong, China
| | - Qingfeng Lv
- The Affiliated Taian City Central Hospital of Qingdao University, Taian 271000, Shandong, China
| | - Ruiheng Huo
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China
| | - Pengzheng Chen
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China.
| | - Lei Li
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China.
| | - Xietong Wang
- Department of Obstetrics and Gynecology, Shandong Provincial Hospital, Shandong University, Jinan 250021, Shandong, China; Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, Shandong, China; The Laboratory of Medical Science and Technology Innovation Center (Institute of Translational Medicine), Shandong First Medical University (Shandong Academy of Medical Sciences) of China, Jinan 250117, Shandong, China.
| |
Collapse
|
16
|
Abuzan M, Surugiu R, Wang C, Mohamud-Yusuf A, Tertel T, Catalin B, Doeppner TR, Giebel B, Hermann DM, Popa-Wagner A. Extracellular Vesicles Obtained from Hypoxic Mesenchymal Stromal Cells Induce Neurological Recovery, Anti-inflammation, and Brain Remodeling After Distal Middle Cerebral Artery Occlusion in Rats. Transl Stroke Res 2025; 16:817-830. [PMID: 39243323 PMCID: PMC12045817 DOI: 10.1007/s12975-024-01266-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 05/18/2024] [Accepted: 05/30/2024] [Indexed: 09/09/2024]
Abstract
Small extracellular vesicles (sEVs) obtained from mesenchymal stromal cells (MSCs) have shown considerable promise as restorative stroke treatment. In a head-to-head comparison in mice exposed to transient proximal middle cerebral artery occlusion (MCAO), sEVs obtained from MSCs cultured under hypoxic conditions particularly potently enhanced long-term brain tissue survival, microvascular integrity, and angiogenesis. These observations suggest that hypoxic preconditioning might represent the strategy of choice for harvesting MSC-sEVs for clinical stroke trials. To test the efficacy of hypoxic MSCs in a second stroke model in an additional species, we now exposed 6-8-month-old Sprague-Dawley rats to permanent distal MCAO and intravenously administered vehicle, platelet sEVs, or sEVs obtained from hypoxic MSCs (1% O2; 2 × 106 or 2 × 107 cell equivalents/kg) at 24 h, 3, 7, and 14 days post-MCAO. Over 28 days, motor-coordination recovery was evaluated by rotating pole and cylinder tests. Ischemic injury, brain inflammatory responses, and peri-infarct angiogenesis were assessed by infarct volumetry and immunohistochemistry. sEVs obtained from hypoxic MSCs did not influence infarct volume in this permanent MCAO model, but promoted motor-coordination recovery over 28 days at both sEV doses. Ischemic injury was associated with brain ED1+ macrophage infiltrates and Iba1+ microglia accumulation in the peri-infarct cortex of vehicle-treated rats. Hypoxic MSC-sEVs reduced brain macrophage infiltrates and microglia accumulation in the peri-infarct cortex. In vehicle-treated rats, CD31+/BrdU+ proliferating endothelial cells were found in the peri-infarct cortex. Hypoxic MSC-sEVs increased the number of CD31+/BrdU+ proliferating endothelial cells. Our results provide evidence that hypoxic MSC-derived sEVs potently enhance neurological recovery, reduce neuroinflammation. and increase angiogenesis in rat permanent distal MCAO.
Collapse
Affiliation(s)
- Mihaela Abuzan
- Experimental Research Center in Normal and Pathological Aging (ARES), University of Medicine and Pharmacy, Craiova, Romania
| | - Roxana Surugiu
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Experimental Research Center in Normal and Pathological Aging (ARES), University of Medicine and Pharmacy, Craiova, Romania
| | - Chen Wang
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Ayan Mohamud-Yusuf
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bogdan Catalin
- Experimental Research Center in Normal and Pathological Aging (ARES), University of Medicine and Pharmacy, Craiova, Romania
| | - Thorsten R Doeppner
- Department of Neurology, University Hospital Gießen and Marburg, Campus Gießen, Giessen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dirk M Hermann
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
- Experimental Research Center in Normal and Pathological Aging (ARES), University of Medicine and Pharmacy, Craiova, Romania.
| | - Aurel Popa-Wagner
- Department of Neurology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
- Experimental Research Center in Normal and Pathological Aging (ARES), University of Medicine and Pharmacy, Craiova, Romania.
| |
Collapse
|
17
|
Samanta A, Yoo MJ, Koh J, Lufkin SC, Lufkin T, Kraus P. Proteomic profiling of small extracellular vesicles from bovine nucleus pulposus cells. PLoS One 2025; 20:e0324179. [PMID: 40440285 PMCID: PMC12121814 DOI: 10.1371/journal.pone.0324179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/21/2025] [Indexed: 06/02/2025] Open
Abstract
Small extracellular vesicles (small EV) are a conserved means of communication across the domains of life and lately gained more interest in mammalian non-cancerous work as non-cellular, biological therapeutic with encouraging results in recent studies of chronic degenerative diseases. The nucleus pulposus (NP) is the avascular and aneural center of an intervertebral disc (IVD), home to unique niche conditions and affected in IVD degeneration. We investigated autologous and mesenchymal stem cell (MSC) small EVs for their potential to contribute to cell and tissue homeostasis in the NP niche via mass spectrometric proteome and functional enrichment analysis using adult and fetal donors. We compared these findings to published small EV databases and MSC small EV data. We propose several mechanisms associated with NP small EVs: Membrane receptor trafficking to modify signal responses promoting niche homeostasis; Redox and energy homeostasis via metabolic enzymes delivery; Cell homeostasis via proteasome delivery and immunomodulation beyond an association with a serum protein corona. The proteome signature of small EVs generated by NP parent cells is similar to previously published small EV data, yet with a focus on supplementing anaerobic metabolism and redox balance while contributing to the maintenance of an aneural and avascular microniche.
Collapse
Affiliation(s)
- Ankita Samanta
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Mi-Jeong Yoo
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Jin Koh
- The Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, Florida, United States of America
| | - Sina Charlotte Lufkin
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Thomas Lufkin
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| | - Petra Kraus
- Department of Biology, Clarkson University, Potsdam, New York, United States of America
| |
Collapse
|
18
|
Zhou M, Guan B, Liu Y, Gu Q, Chen W, Xie B, Zhou M, Xiang J, Zhao S, Zhao Q, Yan D. Fibrinogen-like 2 in tumor-associated macrophage-derived extracellular vesicles shapes an immunosuppressive microenvironment in colorectal liver metastases by promoting tumor stemness and neutrophil extracellular traps formation. Cancer Lett 2025; 618:217642. [PMID: 40097065 DOI: 10.1016/j.canlet.2025.217642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 03/01/2025] [Accepted: 03/14/2025] [Indexed: 03/19/2025]
Abstract
Investigating the mechanisms underlying the development of an immunosuppressive microenvironment within colorectal liver metastases (CRLM) is important for identifying synergistic targets for immunotherapy. The regulatory role of tumor-associated macrophage-derived extracellular vesicles (TAM-EVs) in the immune microenvironment of CRLM has not yet been fully explored. Here, we found that TAM-EVs shaped the immunosuppressive microenvironment at the invasive front in murine CRLM models, thus dampening anti-PD-1 immunotherapy. This environment is characterized by an increased tumor stemness potential and abundant neutrophil extracellular traps (NETs) formation. Mechanistically, TAM-EVs-derived fibrinogen-like 2 (FGL2) interacts with the FCGR2B receptor in tumor cells, which further activates a p-STAT3/IL-1β positive feedback loop to increase the stemness potential of cancer cells, whereas IL-1β mediates the communication between cancer cells and neutrophils. The use of an anti-IL-1β monoclonal antibody can reduce NETs production and synergize with anti-PD-1 immunotherapy, which offers clinical translational significance for CRLM therapy. The FGL2/p-STAT3/IL-1β loop correlates with an immunosuppressive microenvironment and poor prognosis in human patients with CRLM. Our results revealed the potential of enhancing the efficacy of immunotherapy via the targeted clearance of NETs using anti-IL-1β monoclonal antibodies, which have significant clinical translational value in the treatment of CRLM.
Collapse
Affiliation(s)
- Menghua Zhou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bingjie Guan
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Youdong Liu
- Department of Gastric Surgery, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qi Gu
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiwei Chen
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bowen Xie
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mantang Zhou
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianjun Xiang
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Senlin Zhao
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China; Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qian Zhao
- Department of Pathophysiology, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Dongwang Yan
- Department of General Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
19
|
Gan L, Guo X, Dong S, Sun C. The biology of exosomes and exosomal non-coding RNAs in cardiovascular diseases. Front Pharmacol 2025; 16:1529375. [PMID: 40492132 PMCID: PMC12147041 DOI: 10.3389/fphar.2025.1529375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 04/07/2025] [Indexed: 06/11/2025] Open
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of death worldwide, both in developed and developing countries. Despite the implementation of various measures in clinical practice that have shown certain curative effects, poor prognosis and irreversible pathological cardiac remodeling continue to limit the therapeutic effect of CVDs. There are still many new mechanisms worth exploring for the regulation of CVDs. Previous studies have highlighted the potential applicability of exosomes in CVDs, and significant research has been conducted in this area. In this review, we summarize the physiological mechanisms of exosomes and the basic research achievements in regulating CVDs via exosomal non-coding RNAs. We also discuss the limitations and prospects of exosome application in CVD treatment.
Collapse
Affiliation(s)
- Lu Gan
- Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaofei Guo
- Department of Pharmacy, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shichao Dong
- Department of Pharmacy, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Chuan Sun
- Department of Pharmacy, Children’s Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
20
|
Xu K, Wu Q, Lingyun Z, Nguyen R, Safri F, Yang W, Xu Y, Ye Y, Kwan HY, Wang Q, Liang X, Shiddiky MJA, Warkiani ME, George J, Bao J, Qiao L. Extracellular vesicles as a promising platform of precision medicine in liver cancer. Pharmacol Res 2025:107800. [PMID: 40419123 DOI: 10.1016/j.phrs.2025.107800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 05/19/2025] [Accepted: 05/22/2025] [Indexed: 05/28/2025]
Abstract
Extracellular vesicles (EVs) are natural carriers of biological information and play pivotal roles in intercellular communication. EVs are biocompatible, have low immunogenicity, and are capable of traversing biological barriers, making them ideal tools for disease diagnosis and therapy. Despite their promising prospects, the full realization of EVs potential faces several challenges. This article aims to comprehensively review the biological and molecular features of EVs, their applications in liver cancer and possible underlying mechanisms, and the critical challenges affecting the clinical translation of EVs-based therapies in liver cancer.
Collapse
Affiliation(s)
- Keyang Xu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Qibiao Wu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Zhao Lingyun
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Romario Nguyen
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Fatema Safri
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - William Yang
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Yikun Xu
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Yun Ye
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong, China
| | - Hiu Yee Kwan
- Centre for Cancer & Inflammation Research, School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong, China
| | - Qiang Wang
- Division of Medical Imaging and Technology, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden
| | - Xiuming Liang
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine Karolinska Institute, Stockholm, Sweden
| | - Muhammad J A Shiddiky
- Rural Health Research Institute (RHRI), Charles Sturt University, Orange NSW 2800, Australia
| | - Majid E Warkiani
- School of Biomedical Engineering, the University of Technology Sydney, Ultimo NSW 2007, Australia
| | - Jacob George
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia
| | - Jianfeng Bao
- Hangzhou Xixi Hospital affiliated to Zhejiang Chinese Medical University, Zhejiang, China.
| | - Liang Qiao
- Storr Liver Centre, Westmead Institute for Medical Research, the University of Sydney and Westmead Hospital, Westmead, NSW 2145, Australia.
| |
Collapse
|
21
|
Manna OM, Caruso Bavisotto C, Gratie MI, Damiani P, Tomasello G, Cappello F. The Role of Helicobacter pylori Heat Shock Proteins in Gastric Diseases' Pathogenesis. Int J Mol Sci 2025; 26:5065. [PMID: 40507876 PMCID: PMC12155366 DOI: 10.3390/ijms26115065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/19/2025] [Accepted: 05/22/2025] [Indexed: 06/16/2025] Open
Abstract
Helicobacter pylori (H. pylori) is a Gram-negative bacterium that colonizes the human stomach and is associated with several gastric diseases, including gastritis, peptic ulcer disease, and gastric cancer. The bacterium's ability to thrive in the harsh gastric environment is due, to some extent, to its stress response mechanisms, with its heat shock proteins (HSPs) playing a putative, yet not fully understood, role in these adaptive processes. HSPs are a family of molecules, highly conserved throughout phylogenesis, that assist in protein folding, prevent aggregation, and ensure cellular homeostasis under stressful conditions. In H. pylori, HSPs contribute to survival in the stomach's acidic environment and oxidative stress. Furthermore, they aid in the bacterium's ability to adhere to gastric epithelial cells, modulate the host immune response, and form biofilms, all contributing to chronic infection and pathogenicity. The role of microbial HSPs in antibiotic resistance has also emerged as a critical area of research, as these proteins help stabilize efflux pumps, protect essential proteins targeted by antibiotics, and promote biofilm formation, thereby reducing the efficacy of antimicrobial treatments. Among bacterial HSPs, GroEL and DnaK are probably the major proteins that control most of the H. pylori's functioning. Indeed, both proteins possess remarkable acid resistance, high substrate affinity, and dual roles in protein homeostasis and host interaction. These features make them critical for H. pylori's adaptation, persistence, and pathogenicity in the gastric niche. In addition, recent findings have also highlighted the involvement of HSPs in the crosstalk between H. pylori and gastric epithelial cells mediated by the release of bacterial outer membrane vesicles and host-derived exosomes, both of these extracellular vesicles being part of the muco-microbiotic layer of the stomach and influencing cellular signalling and immune modulation. Considering their critical role in the survival and persistence of bacteria, microbial HSPs also represent potential therapeutic targets. Strategies aimed at inhibiting microbial HSP function, combined with conventional antibiotics or developing vaccines targeting microbial HSPs, could provide new avenues for the treatment of H. pylori infections and combat antibiotic resistance. This review explores the multifaceted roles of microbial HSPs in the pathogenesis of H. pylori, highlighting their contributions to bacterial adhesion, immune evasion, stress response, and antibiotic resistance.
Collapse
Affiliation(s)
- Olga Maria Manna
- Pathologic Anatomy Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy;
| | - Celeste Caruso Bavisotto
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy; (C.C.B.); (M.I.G.)
| | - Melania Ionelia Gratie
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BIND), Institute of Human Anatomy and Histology, University of Palermo, 90127 Palermo, Italy; (C.C.B.); (M.I.G.)
| | - Provvidenza Damiani
- Risk Management and Quality Unit, Hospital University “Paolo Giaccone”, 90127 Palermo, Italy; (P.D.); (G.T.)
| | - Giovanni Tomasello
- Risk Management and Quality Unit, Hospital University “Paolo Giaccone”, 90127 Palermo, Italy; (P.D.); (G.T.)
| | - Francesco Cappello
- Risk Management and Quality Unit, Hospital University “Paolo Giaccone”, 90127 Palermo, Italy; (P.D.); (G.T.)
| |
Collapse
|
22
|
Samuels M, Karakostas C, Besta S, Lauer Betrán A, Tsilingiri K, Turner C, Shirazi Nia R, Poudine N, Goodyear R, Jones W, Klinakis A, Giamas G. LMTK3 regulation of EV biogenesis and cargo sorting promotes tumour growth by reducing monocyte infiltration and driving pro-tumourigenic macrophage polarisation in breast cancer. Mol Cancer 2025; 24:149. [PMID: 40405280 PMCID: PMC12100856 DOI: 10.1186/s12943-025-02346-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 04/28/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Lemur Tail Kinase 3 (LMTK3) promotes cell proliferation, invasiveness and therapy resistance, and its expression correlates with poor survival in several different malignancies, including breast cancer. Crosstalk through extracellular vesicles (EVs) is an increasingly appreciated mechanism of cell communication within the tumour immune microenvironment, which contributes to different aspects of cancer progression and plays a pivotal role in shaping tumour fate. METHODS Nanoparticle tracking analysis and transmission electron microscopy were used to study the effects of LMTK3 on EV size, while single particle interferometry allowed us to examine LMTK3-dependent effects on the subpopulation distribution of EVs. Quantitative mass spectrometry was used to profile LMTK3-dependent proteomics changes in breast cancer-derived EVs. Bioinformatics analysis of clinical data along with in vitro and cell-based assays were implemented to explore the effects of LMTK3-dependent EV protein cargo on the tumour immune microenvironment. To elucidate the mechanism through which LMTK3 impacts endosomal trafficking and regulates EV biogenesis, we used a variety of approaches, including in vitro kinase assays, confocal and electron microscopy, as well as in vivo subcutaneous and orthotopic breast cancer mouse models. RESULTS Here, we report that LMTK3 increases the average size of EVs, modulates immunoregulatory EV proteomic cargo and alters the subpopulation distribution of EVs released by breast cancer cells. Mechanistically, we provide evidence that LMTK3 phosphorylates Rab7, a key regulator of multivesicular body (MVB) trafficking, thereby reducing the fusion of MVBs with lysosomes and subsequent degradation of intralumenal vesicles, resulting in altered EV release. Moreover, LMTK3 causes increased packaging of phosphoserine aminotransferase 1 (PSAT1) in EVs, leading to a paracrine upregulation of phosphoglycerate dehydrogenase (PHGDH) in monocytes when these EVs are taken up. PSAT1 and PHGDH play key roles in the serine biosynthesis pathway, which is closely linked to cancer progression and regulation of monocyte behaviour. LMTK3 EV-induced elevated PHGDH expression in monocytes reduces their infiltration into breast cancer 3D spheroids and in vivo breast cancer mouse models. Furthermore, these infiltrating monocytes preferentially differentiate into pro-tumourigenic M2-like macrophages. Additional breast cancer mouse studies highlight the contribution of LMTK3-dependent EVs in the observed immunosuppressive macrophage phenotype. Finally, in vitro experiments show that pharmacological inhibition of LMTK3 reverses the pro-tumourigenic and immunomodulatory effects mediated by EVs derived from LMTK3 overexpressing cells. CONCLUSION Overall, this study advances our knowledge on the mechanisms of EV biogenesis and highlights a novel oncogenic role of LMTK3 in the breast TME, further supporting it as a target for cancer therapy.
Collapse
Affiliation(s)
- Mark Samuels
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University. Oncology department of the first affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK
| | - Christos Karakostas
- Center of Basic Research Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
| | - Simoni Besta
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University. Oncology department of the first affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK
| | - Andrea Lauer Betrán
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK
| | - Katerina Tsilingiri
- Center of Basic Research Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
| | - Charlotte Turner
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK
| | - Reza Shirazi Nia
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University. Oncology department of the first affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Niloufar Poudine
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University. Oncology department of the first affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Richard Goodyear
- Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton, UK
| | - William Jones
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK
| | - Apostolos Klinakis
- Center of Basic Research Biomedical Research Foundation of the Academy of Athens, Athens, 11527, Greece
| | - Georgios Giamas
- International Oncology Institute, The First Affiliated Hospital of Zhejiang Chinese Medical University. Oncology department of the first affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Department of Biochemistry and Biomedicine, School of Life Sciences, University of Sussex, JMS Building, Falmer, Brighton, BN1 9QG, UK.
| |
Collapse
|
23
|
Park KC, Jaafari A, Smith CA, Lobo AR, Errichelli L, Şimşek G, Gunadasa-Rohling M, Marchant A, Levitin MO, Castilla-Llorente V, Vilela P, Swietach P. A Langendorff-heart discovery pipeline demonstrates cardiomyocyte targeting by extracellular vesicles functionalized with beta-blockers using click-chemistry. J Mol Cell Cardiol 2025:S0022-2828(25)00089-6. [PMID: 40414416 DOI: 10.1016/j.yjmcc.2025.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 05/18/2025] [Accepted: 05/21/2025] [Indexed: 05/27/2025]
Abstract
Extracellular vesicles (EVs) are widely explored as vehicles for delivering therapeutic or experimental cargo to cardiomyocytes. Efforts to improve EV bioavailability in the heart, and reduce their off-target actions, require screening methods that can replicate the physiological and anatomical barriers present in the myocardium. Additionally, discovery pipelines must exercise control over EV dosage and timing, and provide a means of assessing cargo incorporation into cardiomyocytes specifically. These criteria are not generally met by experiments on cultured cells or animals. Here, we present a Langendorff-heart discovery pipeline that combines the strengths of in vivo and in vitro approaches. Langendorff-mode perfusion enables controlled exposure of beating hearts to re-circulated EVs. Following perfusion, cardiomyocytes can be isolated enzymatically for analysis, such as imaging. We tested this discovery pipeline by functionalizing EVs with beta-blockers (atenolol, metoprolol) using click-chemistry and incorporating the fluorescent protein NeonGreen2 to track the fate of EV cargo. Fluorescence in cardiomyocytes, including their nuclear regions, increased after Langendorff-treatment with beta-blocker decorated EVs, but only if these contained NeonGreen2, implicating the fluorescent cargo as the source of signal. Superior binding efficacy of beta-blockers was confirmed by referencing to the substantially lower signals obtained using wild-type EVs or EVs presenting myomaker or myomixer proteins, motifs that modestly enrich cardiac EV uptake in mice. Our findings demonstrate successful cardiomyocyte targeting using EVs decorated with beta-receptor binders. We propose the Langendorff-perfused heart as an intermediate step, nested between in vitro characterisation and animal testing, in discovery pipelines for seeking improved EV designs for the heart.
Collapse
Affiliation(s)
- Kyung Chan Park
- Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, UK
| | - Amir Jaafari
- Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, UK
| | | | | | - Lorenzo Errichelli
- Evox Therapeutics, Medawar Centre, Robert Robinson Ave, Oxford OX4 4HG, UK
| | - Gül Şimşek
- Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, UK; Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | | | - Alexander Marchant
- Evox Therapeutics, Medawar Centre, Robert Robinson Ave, Oxford OX4 4HG, UK
| | - Maria O Levitin
- Evox Therapeutics, Medawar Centre, Robert Robinson Ave, Oxford OX4 4HG, UK
| | | | - Patrick Vilela
- Evox Therapeutics, Medawar Centre, Robert Robinson Ave, Oxford OX4 4HG, UK
| | - Pawel Swietach
- Department of Physiology, Anatomy & Genetics, Parks Road, Oxford OX1 3PT, UK.
| |
Collapse
|
24
|
Zhang H, Gong L, Yu L, Xian C, Ma Z, Wang X, Xia R. Emerging roles of non-coding RNA derived from extracellular vesicles in regulating PD-1/PD-L1 pathway: insights into cancer immunotherapy and clinical applications. Cancer Cell Int 2025; 25:188. [PMID: 40410719 PMCID: PMC12103061 DOI: 10.1186/s12935-025-03809-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 05/05/2025] [Indexed: 05/25/2025] Open
Abstract
Numerous studies have demonstrated that extracellular vesicles (EVs) carry a variety of noncoding RNAs (ncRNAs), which can be taken up by neighboring cells or transported to distant sites via bodily fluids, thereby facilitating intercellular communication and regulating multiple cellular functions. Within the tumor microenvironment, EV-ncRNA, on the one hand, regulate the expression of PD-L1, thereby influencing tumor immune evasion, promoting tumor cell proliferation, and enhancing tumor growth, invasion, and metastasis in vivo. On the other hand, these specific EV-ncRNAs can also modulate the functions of immune cells (such as CD8 + T cells, macrophages, and NK cells) through various molecular mechanisms, inducing an immunosuppressive microenvironment and promoting resistance to anti-PD-1 therapy. Therefore, delving into the molecular mechanisms underlying EV-ncRNA regulation of immune checkpoints presents compelling therapeutic prospects for strategies that selectively target EV-ncRNAs. In this review, we elaborate on the cutting-edge research progress related to EV-ncRNAs in the context of cancer and dissect their pivotal roles in the PD-1/PD-L1 immune checkpoint pathway. We also highlight the promising clinical applications of EV-ncRNAs in anti-PD-1/PD-L1 immunotherapy, bridging basic research with practical clinical applications.
Collapse
Affiliation(s)
- Haixia Zhang
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
| | - Lianfeng Gong
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
| | - Li Yu
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China
- Department of Urology, General Hospital of The Yangtze River Shipping, Wuhan, 430010, China
| | - Chenge Xian
- Naidong District People's Hospital, Shannan, 856004, Tibet Autonomous Region, China
| | - Zhaowu Ma
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
| | - Xianwang Wang
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
- Shannan Maternal and Child Health Hospital, Shannan, 856099, Tibet Autonomous Region, China.
| | - Ruohan Xia
- Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou, 434023, Hubei, China.
| |
Collapse
|
25
|
Zhang Y, Wang C, Shao H. Nanoplasmonic Sensing of Heterogeneous Extracellular Vesicles: From Bulk to Single Vesicles. SMALL METHODS 2025:e2500097. [PMID: 40391615 DOI: 10.1002/smtd.202500097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/16/2025] [Indexed: 05/22/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous nanoscale membrane vesicles released by almost all cell types into the circulation. Depending on their biogenesis and cells of origin, EVs show considerable heterogeneity in their biophysical and biomolecular composition and can serve as reflective and dynamic blood biomarkers for personalized medicine. Conventional analytical technologies, however, often lack the compatibility to reveal nanoscale EV features and resolve vesicle heterogeneity. The past decade has since witnessed the development of various nanoplasmonic technologies to empower EV analysis, through bulk and single-vesicle characterization, at an unprecedented scale and resolution. These platforms achieve versatile measurements that are not only size-matched to EV dimensions but can also probe multiplexed biomolecular contents, thereby providing new insights into EV heterogeneity and enabling transformative clinical opportunities. In this review, key characteristics of EVs and their remarkable heterogeneity are introduced. The sensing principles of plasmonic platforms are also discussed, with recent technology developments highlighted to resolve EV heterogeneity, through bulk analyses of EV subpopulations as well as high-resolution single-EV measurements. An outlook is further provided on emerging opportunities, at the interface of biomarker discovery and technology innovation, to develop empowering nanoplasmonic EV platforms for personalized medicine. biosensing; bulk analysis; extracellular vesicles; nanoplasmonics; single-vesicle analysis.
Collapse
Affiliation(s)
- Yan Zhang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Chao Wang
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Huilin Shao
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117583, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
- Department of Materials Science and Engineering, College of Design and Engineering, National University of Singapore, Singapore, 117575, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, 138673, Singapore
| |
Collapse
|
26
|
Longo A, Manganelli V, Misasi R, Riitano G, Caglar TR, Fasciolo E, Recalchi S, Sorice M, Garofalo T. Extracellular Vesicles in the Crosstalk of Autophagy and Apoptosis: A Role for Lipid Rafts. Cells 2025; 14:749. [PMID: 40422252 DOI: 10.3390/cells14100749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 05/13/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025] Open
Abstract
Autophagy and apoptosis are two essential mechanisms regulating cell fate. Although distinct, their signaling pathways are closely interconnected through various crosstalk mechanisms. Lipid rafts are described to act as both physical and functional platforms during the early stages of autophagic and apoptotic processes. Only recently has a role for lipid raft-associated molecules in regulating EV biogenesis and release begun to emerge. In particular, lipids of EV membranes are essential components in conferring stability to these vesicles in different extracellular environments and/or to facilitate binding or uptake into recipient cells. In this review we highlight these aspects, focusing on the role of lipid molecules during apoptosis and secretory autophagy pathways. We describe the molecular machinery that connects autophagy and apoptosis with vesicular trafficking and lipid metabolism during the release of EVs, and how their alterations contribute to the development of various diseases, including autoimmune disorders and cancer. Overall, these findings emphasize the complexity of autophagy/apoptosis crosstalk and its key role in cellular dynamics, supporting the role of lipid rafts as new therapeutic targets.
Collapse
Affiliation(s)
- Agostina Longo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Valeria Manganelli
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Roberta Misasi
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Gloria Riitano
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Tuba Rana Caglar
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Elena Fasciolo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Serena Recalchi
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Maurizio Sorice
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| | - Tina Garofalo
- Department of Experimental Medicine, "Sapienza" University of Rome, 00161 Rome, Italy
| |
Collapse
|
27
|
Wu J, Jin Z, Fu T, Qian Y, Bian X, Zhang X, Zhang J. Extracellular Vesicle-Based Drug Delivery Systems in Cancer Therapy. Int J Mol Sci 2025; 26:4835. [PMID: 40429976 PMCID: PMC12112466 DOI: 10.3390/ijms26104835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Revised: 05/05/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed particles secreted by cells and ubiquitously present in various biofluids. They not only mediate intercellular communication but also serve as promising drug carriers that are capable of delivering therapeutic agents to target cells through their inherent physicochemical properties. In this review, we summarized the recent advances in EV isolation techniques and innovative drug-loading strategies. Furthermore, we emphasized the distinct advantages and therapeutic applications of EVs derived from different cellular sources in cancer treatment. Finally, we critically evaluated the ongoing clinical trials utilizing EVs for drug delivery and systematically assessed both the opportunities and challenges associated with implementing EV-based drug delivery systems in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China; (J.W.); (Z.J.); (T.F.); (Y.Q.); (X.B.)
| | - Jiahui Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China; (J.W.); (Z.J.); (T.F.); (Y.Q.); (X.B.)
| |
Collapse
|
28
|
Hegeman CV, Elsharkasy OM, Driedonks TAP, Friesen KRJ, Vader P, de Jong OG. Modulating binding affinity of aptamer-based loading constructs enhances extracellular vesicle-mediated CRISPR/Cas9 delivery. J Control Release 2025; 384:113853. [PMID: 40393529 DOI: 10.1016/j.jconrel.2025.113853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 04/07/2025] [Accepted: 05/14/2025] [Indexed: 05/22/2025]
Abstract
The CRISPR/Cas9 toolbox consists of modular nucleases that can be employed to efficiently modify genomic sequences with high specificity. However, delivery of the large Cas9-sgRNA ribonucleoprotein (RNP) complexes remains challenging due to their immunogenicity, size, and overall negative charge. An approach to overcome these limitations is the use of extracellular vesicles (EVs) as intracellular delivery vehicles. EVs exhibit the natural ability to carry and deliver RNA and proteins across biological barriers, and can be engineered to load and deliver a variety of biotherapeutic molecules. Previous studies have shown that efficient EV-mediated cargo delivery does not only require active loading strategies, but also benefits from strategies to release cargo from the EV membrane. Here, we load Cas9 RNP complexes into EVs by expressing sgRNAs containing MS2 aptamers (MS2-sgRNAs), alongside Cas9 and a fusion protein of CD63 and tandem MS2 coat proteins (MCPs). We demonstrate that efficient Cas9 RNP delivery can also be facilitated by modulating the binding affinity between MS2 aptamers and the MCPs. To study the effect of altering the binding affinity between the MS2 hairpin and the MCP on Cas9 RNP delivery, various mutations affecting the binding affinity were made in both the interacting MS2-hairpin and the RNA-binding domain of the MCPs. Comparing Cas9 RNP delivery of the modulated MS2-sgRNAs revealed that adapting binding affinity highly affects functional RNP delivery. Mutations resulting in high affinity did not facilitate efficient RNP delivery unless combined with a photo-inducible release strategy, showing that cargo release was a limiting factor in RNP delivery. Mutations that decreased affinity resolved this issue, resulting in Cas9 RNP delivery without the requirement of additional release strategies. However, further decreasing affinity resulted in decreased Cas9 gene-editing efficiency due to decreased levels of Cas9 RNP loading into EVs. A similar effect on functional delivery was seen after modification of the RNA-binding domain of the MCPs. Our results demonstrate that EVs are capable of functional Cas9-sgRNA complex delivery, and that modulation of binding affinity can be used to increase efficient functional delivery with non-covalent loading constructs, without the need for additional engineering strategies for cargo release.
Collapse
Affiliation(s)
- Charlotte V Hegeman
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands
| | - Omnia M Elsharkasy
- CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Tom A P Driedonks
- CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Kate R J Friesen
- Department of Oncology, University of Oxford, Oxford, Oxfordshire, United Kingdom
| | - Pieter Vader
- CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands; Department of Experimental Cardiology, University Medical Center Utrecht, the Netherlands
| | - Olivier G de Jong
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Utrecht University, Utrecht, the Netherlands; CDL Research, University Medical Center Utrecht, Utrecht, the Netherlands.
| |
Collapse
|
29
|
Sonar S, Das A, Yeong Zher L, Narayanan Ravi R, Zheng Kong EQ, Dhar R, Narayanan K, Gorai S, Subramaniyan V. Exosome-Based Sensor: A Landmark of the Precision Cancer Diagnostic Era. ACS APPLIED BIO MATERIALS 2025. [PMID: 40366154 DOI: 10.1021/acsabm.5c00288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Extracellular vesicles are nanoscale vesicles released by a diversity of cells that mediate intercellular communication by transporting an array of biomolecules. They are gaining increasing attention in cancer research due to their ability to carry specific biomarkers. This characteristic makes them potentially useful for highly sensitive, noninvasive diagnostic procedures and more precise prognostic assessments. Consequently, EVs are emerging as a transformative tool in cancer treatment, facilitating early detection and personalized medicine. Despite significant progress, clinical implementation is hindered by challenges in EV isolation, purification, and characterization. However, developing advanced biosensor technologies offers promising solutions to these obstacles. This review highlights recent progress in biosensors for EV detection and analysis, focusing on various sensing modalities including optical, electrochemical, microfluidic, nanomechanical, and biological sensors. We also explore techniques for EV isolation, characterization, and analysis, such as electron microscopy, atomic force microscopy, nanoparticle tracking analysis, and single-particle analysis. Furthermore, the review critically assesses the challenges associated with EV detection and put forward future directions, aiming to usher in a cutting-edge era of precision medicine through advanced, sensor-based, noninvasive early cancer diagnosis by detecting EV-carried biomarkers.
Collapse
Affiliation(s)
- Swarup Sonar
- Department of Oncology, Neuron Institute of Applied Research, Amravati, Maharashtra 444605, India
| | - Asmit Das
- Department of Oncology, Neuron Institute of Applied Research, Amravati, Maharashtra 444605, India
| | - Lee Yeong Zher
- Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Ram Narayanan Ravi
- Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Eason Qi Zheng Kong
- Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Rajib Dhar
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, Subang Jaya 47500, Selangor (Darul Ehsan), Malaysia
| | - Kumaran Narayanan
- Monash University Malaysia, Bandar Sunway, Subang Jaya 47500, Selangor, Malaysia
| | - Sukhamoy Gorai
- Department of Neurological Sciences, Rush University Medical Center, 1620 W Harrison Street, Chicago, Illinois 60612, United States
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, Subang Jaya 47500, Selangor (Darul Ehsan), Malaysia
| |
Collapse
|
30
|
Staller DW, Gawargi FI, Panigrahi SS, Mishra PK, Mahato RI. Pharmaceutical perspectives on oligonucleotide therapeutics and delivery systems. Pharmacol Rev 2025; 77:100065. [PMID: 40513184 DOI: 10.1016/j.pharmr.2025.100065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/09/2025] [Accepted: 05/07/2025] [Indexed: 06/16/2025] Open
Abstract
Gene therapy has a pivotal role in treating new diseases. In addition to the recent mRNA-based COVID-19 vaccines produced by Pfizer-BioNTech and Moderna against severe acute respiratory syndrome corona virus 2, several new gene therapies have recently been approved as effective treatments for fatal genetic disorders such as Duchenne's muscular dystrophy, familial transthyretin amyloidosis, hemophilia A, hemophilia B, spinal muscle atrophy, early cerebral autoleukodystrophy, and β-thalassemia. This review provides novel insights into RNA therapeutics focusing on endogenous RNA species, RNA structure and function, and chemical modifications that improve the stability and distribution of RNAs. Furthermore, it includes updated knowledge on clinically approved gene therapies rendering a comprehensive understanding of the biochemical basis and clinical application of gene therapies. SIGNIFICANCE STATEMENT: There have recently been significant advances in clinical translation of RNA therapeutics. This review discusses the diverse types of RNA species, RNA structure and function, backbone and chemical modifications to RNAs, and every RNA therapeutic approved for clinical use at the time of writing.
Collapse
Affiliation(s)
- Dalton W Staller
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Flobater I Gawargi
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Sanjali S Panigrahi
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska
| | - Paras K Mishra
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Ram I Mahato
- Department of Cellular & Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska; Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, Nebraska.
| |
Collapse
|
31
|
Li L, Zheng Z, Lan W, Tang N, Zhang D, Ling J, Wu Y, Yang P, Fu L, Liu J, Zhang J, Yu P, Huang T. Role of Exosomes in Cardiovascular Disease: A Key Regulator of Intercellular Communication in Cardiomyocytes. ACS OMEGA 2025; 10:18145-18169. [PMID: 40385188 PMCID: PMC12079207 DOI: 10.1021/acsomega.4c11423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/27/2025] [Accepted: 04/22/2025] [Indexed: 05/20/2025]
Abstract
In the cardiovascular system, different types of cardiovascular cells can secrete specific exosomes and participate in the maintenance of cardiovascular function and the occurrence and development of diseases. Exosomes carry biologically active substances such as proteins and nucleic acids from cells of origin and can be used as biomarkers for disease diagnosis and prognosis assessment. In addition, exosome-mediated intercellular communication plays a key role in the occurrence and development of cardiovascular diseases and has become a potential therapeutic target. This article emphasizes the importance of understanding the mechanism of exosomes in cardiovascular diseases and systematically details the current understanding of exosomes as regulators of intercellular communication in cardiomyocytes, providing a basis for future research and therapeutic intervention.
Collapse
Affiliation(s)
- Liuxin Li
- Department of Endocrinology and Metabolism, second Affiliated Hospital
of Nanchang University, Nanchang, People’s Republic of China, The second Clinical Medical College, Nanchang University, Nanchang 330006, Republic of China
| | - Zhidong Zheng
- Department of Endocrinology and Metabolism, second Affiliated Hospital
of Nanchang University, Nanchang, People’s Republic of China, The second Clinical Medical College, Nanchang University, Nanchang 330006, Republic of China
| | - Wenyu Lan
- The
Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Nan Tang
- The
Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Deju Zhang
- Food
and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong 0000, Hong Kong
| | - Jitao Ling
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Yuting Wu
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Pingping Yang
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Linhua Fu
- Department
of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Jianping Liu
- Department
of Endocrinology and Metabolism, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| | - Jing Zhang
- Department
of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical
College, Nanchang University, Nanchang 330006, Jiangxi, China
| | - Peng Yu
- Department
of Metabolism and Endocrinology, The Second
Affiliated Hospital of Nanchang University, Nanchang 330006, Jiangxi, China
| | - Tieqiu Huang
- Department
of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi
Medical College, Nanchang University, Nanchang 330006, Jiangxi,China
| |
Collapse
|
32
|
Gao Y, Li X, Ding Y, Wang Y, Du J, Chen Y, Xu J, Liu Y. MiR-451a-Enriched Small Extracellular Vesicles Derived from Mg 2+-Activated DPSCs Induce Vascularized Bone Regeneration through the AKT/eNOS/NO Axis. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40364481 DOI: 10.1021/acsami.5c02551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Blood vessel formation is a necessary part of bone tissue regeneration. MSCs-sEVs play a vital role in the in vivo bone regeneration strategy. However, natural MSCs-sEVs suffer from limited blood vessel formation potency, which makes it difficult to induce vascularized bone regeneration. Here, sEVs derived from magnesium cation-activated DPSCs (Mg2+-EVs) are purified and found to have superior potential in promoting endothelial cell migration and angiogenesis, as well as BMSC proliferation and osteogenesis. The beneficial effects of Mg2+-EVs could be attributed to the enrichment of miR-451a and the subsequent regulation and activation of AKT/eNOS signaling pathways. On this basis, Mg2+-EVs are delivered on β-TCP-modified GelMA scaffolds for slow release and better bioavailability. The rat cranial defect model verifies that GelMA/β-TCP with Mg2+-EVs has enhanced potential of inducing vascularized bone regeneration. The present study provides a cation-activated strategy to modulate the cargos and contents of MSC-derived sEVs, obtaining desirable vascular promotion and bone regeneration potential. Furthermore, the developed β-TCP-modified delivery scaffolds represent a promising strategy for efficient loading and slow-release delivery of sEVs for clinical translation.
Collapse
Affiliation(s)
- Yike Gao
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, No. 9 Fanjiacun Road, Beijing 100071, China
| | - Xiaoyan Li
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, No. 9 Fanjiacun Road, Beijing 100071, China
| | - Yichen Ding
- Department of Endodontics, School of Stomatology, Capital Medical University, No. 9 Fanjiacun Road, Beijing 100071, China
| | - Yanxue Wang
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, No. 9 Fanjiacun Road, Beijing 100071, China
| | - Juan Du
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, No. 9 Fanjiacun Road, Beijing 100071, China
| | - Yingyi Chen
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, No. 9 Fanjiacun Road, Beijing 100071, China
| | - Junji Xu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, No. 9 Fanjiacun Road, Beijing 100071, China
| | - Yi Liu
- Laboratory of Tissue Regeneration and Immunology and Department of Periodontics, Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Capital Medical University, No. 9 Fanjiacun Road, Beijing 100071, China
| |
Collapse
|
33
|
Philippon M, Labib R, Ley MBRG, Kaplan LD, Mendez AJ, Best TM, Kouroupis D. Characterization of Extracellular Vesicles from Infrapatellar Fat Pad Mesenchymal Stem/Stromal Cells Expanded Using Regulatory-Compliant Media and Inflammatory/Hormonal Priming. Cells 2025; 14:706. [PMID: 40422209 PMCID: PMC12109853 DOI: 10.3390/cells14100706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/23/2025] [Accepted: 05/08/2025] [Indexed: 05/28/2025] Open
Abstract
Osteoarthritis (OA) remains a leading cause of disability worldwide, with no disease-modifying therapies currently available for treatment. The infrapatellar fat pad (IFP) harbors mesenchymal stem/stromal cells (MSC) with potent immunomodulatory and regenerative properties, making them a promising candidate for OA treatment. A growing body of evidence suggests that the therapeutic effects of MSC are largely mediated by their extracellular vesicles (EVs), which carry bioactive cargo that modulates inflammation and tissue repair. However, optimizing MSC-derived EVs as a cell-free therapeutic approach requires an in-depth understanding of how culture conditions and inflammatory/hormonal priming influence their functional properties. In this study, IFP-MSC were expanded in regulatory-compliant human platelet lysate (HPL) and xeno-/serum-free (XFSF) media and primed with an inflammatory/fibrotic cocktail (TIC) with oxytocin (OXT) to assess the impact on their immunophenotypic profile and EV cargo. The immunophenotype confirmed that TIC+OXT-primed MSC retained key immunomodulatory surface markers, while EV characterization verified the successful isolation of CD63+/CD9+ vesicles. Pathway enrichment analysis of both HPL- and XFSF- TIC+OXT EVs cargo identified key miRNAs associated with immune regulation, tissue repair, and anabolic signaling. Functional assays revealed that TIC+OXT EVs promoted M2-like anti-inflammatory macrophage polarization and exhibited chondroprotective properties in chondrocytes/synoviocytes inflammatory osteoarthritic assay. These findings highlight the therapeutic potential of TIC+OXT-primed IFP-MSC-derived EVs as immunomodulatory and chondroprotective agents, offering a promising strategy for OA treatment through a clinically viable, cell-free approach.
Collapse
Affiliation(s)
- Marc Philippon
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Ramy Labib
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Michelle Bellas Romariz Gaudie Ley
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biomedical Engineering, University of Miami, Miami, FL 33146, USA
| | - Lee D. Kaplan
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
| | - Armando J. Mendez
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| | - Thomas M. Best
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
| | - Dimitrios Kouroupis
- Department of Orthopaedics, UHealth Sports Medicine Institute, Miller School of Medicine, University of Miami, Miami, FL 33146, USA; (M.P.J.); (R.L.); (M.B.R.G.L.); (L.D.K.); (T.M.B.)
- Diabetes Research Institute & Cell Transplant Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
| |
Collapse
|
34
|
Hagos B, Brasov I, Branscome H, Rashid S, Bradford R, Leonelli J, Kashanchi F, Ben Mamoun C, Molestina RE. Activation of macrophages by extracellular vesicles derived from Babesia-infected red blood cells. Infect Immun 2025; 93:e0033324. [PMID: 40172538 PMCID: PMC12070731 DOI: 10.1128/iai.00333-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 02/24/2025] [Indexed: 04/04/2025] Open
Abstract
Babesia microti is the primary cause of human babesiosis in North America. Despite the emergence of the disease in recent years, the pathogenesis and immune response to B. microti infection remain poorly understood. Studies in laboratory mice have shown a critical role for macrophages in the elimination of parasites and infected red blood cells (iRBCs). Importantly, the underlying mechanisms that activate macrophages are still unknown. Recent evidence identified the release of extracellular vesicles (EVs) from Babesia iRBCs. EVs are spherical particles released from cell membranes under natural or pathological conditions that have been suggested to play roles in host-pathogen interactions among diseases caused by protozoan parasites. The present study examined whether EVs released from cultured Babesia iRBCs could activate macrophages and alter cytokine secretion. An analysis of vesicle size in EV fractions from Babesia iRBCs showed diverse populations in the <100 nm size range compared to EVs from uninfected RBCs. In co-culture experiments, EVs released by B. microti iRBCs appeared to be associated with macrophage membranes and cytoplasm, indicating uptake of these vesicles in vitro. Interestingly, the incubation of macrophages with EVs isolated from Babesia iRBC culture supernatants resulted in the activation of NF-κB and modulation of pro-inflammatory cytokines. These results support a role for Babesia-derived EVs in macrophage activation and provide new insights into the mechanisms involved in the induction of the innate immune response during babesiosis.
Collapse
Affiliation(s)
- Biniam Hagos
- American Type Culture Collection, Manassas, Virginia, USA
| | - Ioana Brasov
- American Type Culture Collection, Manassas, Virginia, USA
| | | | - Sujatha Rashid
- American Type Culture Collection, Manassas, Virginia, USA
| | | | | | - Fatah Kashanchi
- School of Systems Biology, George Mason University, Manassas, Virginia, USA
| | - Choukri Ben Mamoun
- Section of Infectious Disease, Yale University School of Medicine, New Haven, Connecticut, USA
| | | |
Collapse
|
35
|
Phan N, Li Y, Yang M, Liu F. Tear fluid derived extracellular vesicles for new biomarker discovery. Ocul Surf 2025; 37:314-322. [PMID: 40368029 DOI: 10.1016/j.jtos.2025.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 04/23/2025] [Accepted: 05/05/2025] [Indexed: 05/16/2025]
Abstract
Various cell types release extracellular vesicles (EVs) containing proteins, DNA, and RNA essential for intercellular communication. The bioactive molecules from EVs can reflect disease status and monitor progression, while their communication abilities suggest therapeutic potential. We will review various EV isolation methods, EV-enriched fluids, and studies analyzing differential mi-RNA and protein levels extracted from EVs. Specifically, tear-derived EVs, which protect their molecular content and allow for real-time monitoring of ocular conditions such as Dry Eye Disease (DED), Sjögren's disease (SJD), Ocular graft-versus-host disease (oGVHD), and Diabetic Retinopathy (DR), which all currently remain undiagnosed in patients. EVs also provide potential as carriers for gene transfer, and mesenchymal stem cell (MSCs)-derived EVs are shown to be immunomodulatory, demonstrating promise for autoimmune ocular diseases. Through the multi-omic analysis of tear-fluid content, EVs are promising biomarkers and therapeutic agents in ocular diseases.
Collapse
Affiliation(s)
- Natalie Phan
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Molecular and Cell Biology, University of California at Berkeley, Berkeley, CA, 94720, USA
| | - Yi Li
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Menglu Yang
- Department of Ophthalmology, Schepens Eye Research Institute of Mass Eye and Ear, Harvard Medical School, Boston, MA, 02114, USA.
| | - Fei Liu
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA.
| |
Collapse
|
36
|
Tanrıkulu MD, Çevi K M, Yüce M, Neslihan Taşlı P, Yıldırım K. Cryoprotective effects of mesenchymal stem cell and seminal plasma-derived extracellular vesicles on canine sperm. Theriogenology 2025; 244:117480. [PMID: 40381592 DOI: 10.1016/j.theriogenology.2025.117480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 05/01/2025] [Accepted: 05/09/2025] [Indexed: 05/20/2025]
Abstract
In this study, the lack of standardized freezing protocols for sperm cryopreservation in dogs and the limited research on using exosomes in cryopreservation were considered. Additionally, unlike previous studies on sperm cryopreservation, we introduced an innovative approach using the Aqueous Two-Phase System (ATPS) method for exosome isolation. This study aimed to evaluate the sperm-protective effects of adipose tissue-derived mesenchymal stem cell exosomes (MSC-exo) and seminal plasma exosomes (SP-exo) in dog sperm cryopreservation. Ejaculates from six dogs were processed with Tris-based diluents and divided into four groups: MSC-exo, 1.5 % SP-exo, 2 % SP-exo, and control, and frozen. After thawing, sperm motility, viability, membrane integrity, chromatin integrity, morphological integrity, and gene expression levels were analyzed. The results showed that the MSC-exo group had significantly higher total motility (%60.31 ± 6.12), progressive motility (%22.09 ± 3.34), plasma membrane integrity (%66.94 ± 2.24), and viability (%70.88 ± 1.95) compared to the other groups (P < 0.05). Additionally, the normal chromatin packaging rate was highest in the MSC-exo group (%91.33 ± 0.61, P < 0.05). While some improvements were observed in the SP-exo groups, they were not as pronounced as in the MSC-exo group. No significant differences were found in gene expression levels, although an improvement trend was observed in the MSC-exo group. In conclusion, MSC-exo reduced cryopreservation-induced sperm damage and provided overall protection in sperm parameters. These findings suggest that MSC-exo could be a potential biological additive in dog sperm freezing protocols.
Collapse
Affiliation(s)
- Merve Deniz Tanrıkulu
- Ondokuz Mayıs University, Faculty of Veterinary Medicine, Department of Reproduction and Artificial Insemination, Samsun, Turkey.
| | - Mesut Çevi K
- Ondokuz Mayıs University, Faculty of Veterinary Medicine, Department of Reproduction and Artificial Insemination, Samsun, Turkey
| | - Melek Yüce
- Ondokuz Mayıs University, School of Health Services, Samsun, Turkey
| | - Pakize Neslihan Taşlı
- Yeditepe University, Faculty of Engineering, Department of Genetics and Bioengineering, Istanbul, Turkey
| | - Kubilay Yıldırım
- Ondokuz Mayıs University, Faculty of Science, Department of Molecular Biology and Genetics, Samsun, Turkey
| |
Collapse
|
37
|
Bharti PS, Rani K, Singh R, Rai S, Rastogi S, Batra M, Mishra A, Zehra S, Gorai PK, Sasidhar MV, Modi GP, Malik G, Rani N, Dev K, Reddy TJ, Inampudi KK, Nikolajeff F, Kumar S. A simplified and efficient method for isolating small extracellular vesicles for comparative and comprehensive translational research. Sci Rep 2025; 15:16367. [PMID: 40350518 PMCID: PMC12066714 DOI: 10.1038/s41598-025-99822-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 04/23/2025] [Indexed: 05/14/2025] Open
Abstract
Small extracellular vesicles (sEVs) can provide information about the pathophysiology of the cells; therefore, sEVs have attracted considerable interest as possible diagnostic biomarkers. A key challenge lies in the necessity for simple and cost-effective sEV isolation methods to achieve high purity and yield suitable for research and clinical applications. We are introducing a comprehensive study on isolating sEVs using a novel cocktail strategy that integrates chemical precipitation and ultrafiltration with a two-step filtering process to ensure a highly pure and homogeneous population and further compared with PEG-based precipitation, ultra-centrifugation, and size-exclusion-chromatography columns. The isolated sEVs from each protocol are quantified for size and yield using nanoparticle tracking analysis, morphologically characterized through transmission electron microscopy, and validated by quantifying the expression profiles of sEV surface biomarkers. Furthermore, the study explores the applicability of our method for downstream multi-omics analyses. The results highlight the efficacy of the proposed protocol, demonstrating the ease and efficiency of isolating sEVs from different biofluids with minimal laboratory requirements and confirming the compatibility with multi-omics analyses. These findings position our method as particularly valuable for translational research, offering a promising avenue for advancing the study and application of sEVs in diagnostic and therapeutic research.
Collapse
Affiliation(s)
- Prahalad Singh Bharti
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Komal Rani
- Department of Pathology & Laboratory Medicine, All India Institute of Medical Sciences Bibinagar, Hyderabad, Telangana, 508126, India
| | - Rishabh Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Sanskriti Rai
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Simran Rastogi
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Manya Batra
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Abhay Mishra
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Sadaqa Zehra
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Priya Kumari Gorai
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Manda Venkata Sasidhar
- Apollo Hospitals Educational and Research Foundation, Hyderabad, Telangana, 500033, India
| | - Gyan Prakash Modi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh, 221005, India
| | - Garima Malik
- Indian Council of Medical Research, New Delhi, 110029, India
| | - Neerja Rani
- Department of Anatomy, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Kapil Dev
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, 110025, India
| | - Thota Jagadeshwar Reddy
- Analytical Department, CSIR-Indian Institute of Chemical Technology, Hyderabad, Telangana, 500007, India
| | - Krishna Kishore Inampudi
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India
| | - Fredrik Nikolajeff
- Department of Health, Education and Technology, Lulea University of Technology, 97187, Lulea, Sweden
| | - Saroj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, Delhi, 110029, India.
- Department of Health, Education and Technology, Lulea University of Technology, 97187, Lulea, Sweden.
| |
Collapse
|
38
|
Jin Y, Xu C, Zhu Y, Gu Z. Extracellular vesicle as a next-generation drug delivery platform for rheumatoid arthritis therapy. J Control Release 2025; 381:113610. [PMID: 40058499 DOI: 10.1016/j.jconrel.2025.113610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 03/01/2025] [Accepted: 03/04/2025] [Indexed: 03/16/2025]
Abstract
Rheumatoid arthritis (RA) is a systemic autoimmune disorder characterized by chronic inflammation and progressive damage to connective tissue. It is driven by dysregulated cellular homeostasis, often leading to autoimmune destruction and permanent disability in severe cases. Over the past decade, various drug delivery systems have been developed to enable targeted therapies for disease prevention, reduction, or suppression. As an emerging therapeutic platform, extracellular vesicles (EVs) offer several advantages over conventional drug delivery systems, including biocompatibility and low immunogenicity. Consequently, an increasing number of studies have explored EV-based delivery systems in the treatment of RA, leveraging their natural ability to evade phagocytosis, prolong in vivo half-life, and minimize the immunogenicity of therapeutic agents. In this review, we first provide an in-depth overview of the pathogenesis of RA and the current treatment landscape. We then discuss the classification and biological properties of EVs, their potential therapeutic mechanisms, and the latest advancements in EVs as drug delivery platforms for RA therapy. We emphasize the significance of EVs as carriers in RA treatment and their potential to revolutionize therapeutic strategies. Furthermore, we examine key technological innovations and the future trajectory of EV research, focusing on the challenges and opportunities in translating these platforms into clinical practice. Our discussion aims to offer a comprehensive understanding of the current state and future prospects of EV-based therapeutics in RA.
Collapse
Affiliation(s)
- Yi Jin
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China
| | - Cong Xu
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, United States
| | - Yujuan Zhu
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| | - Zhifeng Gu
- Department of Rheumatology, Research Center of Clinical Medicine, Research Center of Immunology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, China.
| |
Collapse
|
39
|
Chen Y, Ghavami S, Park PC, Irobi J. Editorial: Decoding cell fate: the critical roles of extracellular vesicles. Front Cell Dev Biol 2025; 13:1615951. [PMID: 40406418 PMCID: PMC12095917 DOI: 10.3389/fcell.2025.1615951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/26/2025] Open
Affiliation(s)
- Yongqiang Chen
- Manitoba Chemosensory Biology Research Group, Department of Oral Biology, Dr. Gerald Niznick College of Dentistry, University of Manitoba, Winnipeg, MB, Canada
- Children’s Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Saeid Ghavami
- Children’s Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
- Department of Human Anatomy and Cell Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
- Paul Albrechtsen Research Institute, CancerCare Manitoba, University of Manitoba, Winnipeg, MB, Canada
- Akademia Śląska, Katowice, Poland
| | - Paul C. Park
- Department of Pathology, Max Rady College of Medicine, University of Manitoba, Winnipeg, MB, Canada
| | - Joy Irobi
- Department of Immunology and Infections, Biomedical Research Institute, Hasselt University, Hasselt, Belgium
| |
Collapse
|
40
|
Mariante RM, Muñoz-Caro T, Saraiva EM. Editorial: Neutrophil extracellular traps (NETs) triggered by helminths and protozoan parasites. Front Immunol 2025; 16:1613886. [PMID: 40406102 PMCID: PMC12095302 DOI: 10.3389/fimmu.2025.1613886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2025] [Accepted: 04/28/2025] [Indexed: 05/26/2025] Open
Affiliation(s)
- Rafael M. Mariante
- Laboratório de Biologia Estrutural, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil
| | - Tamara Muñoz-Caro
- Escuela de Medicina Veterinaria, Facultad de Medicina Veterinaria y Recursos Naturales, Universidad Santo Tomás, Talca, Chile
| | - Elvira M. Saraiva
- Laboratório de Imunidade Inata, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
41
|
Balic N, Nikolac Perkovic M, Milos T, Vuic B, Kurtovic Kodzoman M, Svob Strac D, Nedic Erjavec G. Extracellular vesicles as a promising tool in neuropsychiatric pharmacotherapy application and monitoring. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111393. [PMID: 40340017 DOI: 10.1016/j.pnpbp.2025.111393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/23/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
This review deals with the application of extracellular vesicles (EVs) in the treatment of various neuropsychiatric disorders, including mood disorders, neurodegeneration, psychosis, neurological insults and injuries, epilepsy and substance use disorders. The main challenges of most neuropsychiatric pharmaceuticals nowadays are how to reach the central nervous system at therapeutic concentration and maintain it long enough and how to avoid undesirable side effects caused by unsatisfying toxicity. Extracellular vesicles, as very important mediators of intercellular communication, can have a variety of therapeutic qualities. They can act neuroprotective, regenerative and anti-inflammatory, but they also have characteristics of a good drug delivery system, including their nano- scale size, biological safety and abilities to cross BBB, to pack drugs within the lipid bilayer, and not to trigger an immunological response. Besides, due to their presence in readily accessible biofluids, they are good candidates for biomarkers of the disease, its progression and therapy response monitoring. Alternations in EVs' cargo profiles can reflect the pathogenesis of neuropsychiatric disorders, but they could also affect the disease outcomes. In the future, EVs could help physicians to tailor treatment strategies for individual patients, however, more extensive studies are needed to standardize isolation, purification and production procedures, increase efficacy of drug loading and limit unwanted effects of innate EVs' content.
Collapse
Affiliation(s)
- Nikola Balic
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | | | - Tina Milos
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | - Barbara Vuic
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | | | | | | |
Collapse
|
42
|
Xu F, Wang K, Xu C, Xu J, Zhu C, Zhu Y, Zhu C, Zhang W, Zhang J, Li Z, Guan X. Enrichment and Detection of HER2-Expressing Extracellular Vesicles Based on DNA Tetrahedral Nanostructures: A New Strategy for Liquid Biopsy in Breast Cancer. Anal Chem 2025; 97:9212-9219. [PMID: 40116571 DOI: 10.1021/acs.analchem.4c06417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Extracellular vesicles (EVs) play a crucial role as important mediators of intercellular communication in the progression of tumors. The capture and analysis of tumor-derived EVs offer new possibilities for the application of cancer liquid biopsies. This study aims to construct a DNA tetrahedral nanostructure that specifically recognizes HER2 and CD63, enabling the effective enrichment and detection of HER2-expressing EVs (HEVs). We enriched HEVs from cell lines and 13 random clinical samples and validated their characteristics by dynamic light scattering, transmission electron microscopy, and Western blotting. Further, we detected HEVs levels in clinical samples. The HEVs levels in HER2-positive breast cancer patients were significantly higher than those in healthy/benign controls (mean, 4.737 vs 4.160 vs 4.144 U/μL, P < 0.0001), displaying a concentration gradient across different HER2 expression levels. This study establishes an approach for HEV detection, thus providing a new tool for the diagnosis of HER2-positive breast cancer.
Collapse
Affiliation(s)
- Feng Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ke Wang
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chi Xu
- Department of Breast Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, China
| | - Jingtong Xu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chengjun Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ye Zhu
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Chuandong Zhu
- Department of Oncology, Nanjing Second Hospital, Nanjing 210003, China
| | - Wenwen Zhang
- Department of Oncology, Nanjing First Hospital, Nanjing 210006, China
| | - Jian Zhang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center, Shanghai 200032, China
| | - Zhe Li
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing 210000, China
- State Key Laboratory of Analytical Chemistry for Life Science, Nanjing University, Nanjing 210000, China
| | - Xiaoxiang Guan
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
- Jiangsu Key Lab of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Personalized Cancer Medicine, Nanjing Medical University, Nanjing 211100, China
| |
Collapse
|
43
|
Bairagi RD, Reon RR, Hasan MM, Sarker S, Debnath D, Rahman MT, Rahman S, Islam MA, Siddique MAT, Bokshi B, Rahman MM, Acharzo AK. Ocular drug delivery systems based on nanotechnology: a comprehensive review for the treatment of eye diseases. DISCOVER NANO 2025; 20:75. [PMID: 40317427 PMCID: PMC12049359 DOI: 10.1186/s11671-025-04234-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/07/2025] [Indexed: 05/07/2025]
Abstract
Ocular drug delivery is a significant challenge due to the intricate anatomy of the eye and the various physiological barriers. Conventional therapeutic approaches, while effective to some extent, often fall short in effectively targeting ocular diseases, resulting in suboptimal therapeutic outcomes due to factors such as poor ocular bioavailability, frequent dosing requirements, systemic side effects, and limited penetration through ocular barriers. This review elucidates the eye's intricate anatomy and physiology, prevalent ocular diseases, traditional therapeutic modalities, and the inherent pharmacokinetic and pharmacodynamic limitations associated with these modalities. Subsequently, it delves into nanotechnology-based solutions, presenting breakthroughs in nanoformulations such as nanocrystals, liposomes, dendrimers, and nanoemulsions that have demonstrated enhanced drug stability, controlled release, and deeper ocular penetration. Additionally, it explores a range of nanosized carriers, including nano-structured lipid carriers, hydrogels, nanogels, nanoenzymes, microparticles, conjugates, exosomes, nanosuspensions, viral vectors, and polymeric nanoparticles, and their applications. Unique insights include emerging innovations such as nanowafers and transcorneal iontophoresis, which indicate paradigm shifts in non-invasive ocular drug delivery. Furthermore, it sheds light on the advantages and limitations of these nanotechnology-based platforms in addressing the challenges of ocular drug delivery. Though nano-based drug delivery systems are drawing increasing attention due to their potential to enhance bioavailability and therapeutic efficacy, the review ends up emphasizing the imperative need for further research to drive innovation and improve patient outcomes in ophthalmology.
Collapse
Affiliation(s)
- Rahul Dev Bairagi
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | - Raiyan Rahman Reon
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | - Md Mahbub Hasan
- Department of Biomedical Engineering, Khulna University of Engineering and Technology (KUET), Khulna, 9203, Bangladesh
| | - Sumit Sarker
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, Bara Phool, Punjab, 140001, India
| | - Dipa Debnath
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology BHU, Varanasi, Uttar Pradesh, 221005, India
| | - Md Tawhidur Rahman
- Department of Pharmacy, Northern University of Bangladesh, Dhaka, 1230, Bangladesh
| | - Sinthia Rahman
- Department of Chemistry, University of Wyoming, Laramie, WY, USA
| | - Md Amirul Islam
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
- Department of Pharmacy, East West University, Dhaka, 1212, Bangladesh
| | - Md Abu Talha Siddique
- Department of Pharmaceutics, College of Pharmacy, University of Florida, Gainesville, FL, 32610, USA
| | - Bishwajit Bokshi
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | - Md Mustafizur Rahman
- Pharmacy Discipline, School of Life Sciences, Khulna University, Khulna, 9208, Bangladesh
| | | |
Collapse
|
44
|
Botto A, De Cesari C, Ndimurwanko N, Finamore F, Greco F, Cappello V, Casieri V, Immordino B, Lionetti V, Gemmi M, Tonazzini I, Giovannetti E, McDonnell LA. Novel PPT+SEC Workflow for High-Sensitivity Extracellular Vesicle Proteomics from Cell Media. J Proteome Res 2025. [PMID: 40315925 DOI: 10.1021/acs.jproteome.5c00082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
Size exclusion chromatography (SEC) is a well-established method for the isolation of extracellular vesicles (EVs), but the large elution volumes necessitate a concentration step prior to proteomics analysis. This concentration step can lead to a significant EV loss. Here we report an EV proteomics approach that enables the isolation of EVs into just 80 μL, which is directly compatible with proteomics analysis without the need for a prior concentration. EVs were characterized by transmission electron microscopy, Western blot, and nanoparticle tracking analysis, all of which confirmed the presence of small EVs. Proteomics analysis of the EVs was performed and benchmarked against those isolated by using an automated UHPLC-SEC platform. The novel workflow identified more proteins and more EV markers, including 96 of the 100 top exosomal proteins from the ExoCarta database, compared to 91 identified using EV samples isolated by UHPLC-SEC. When applied to EVs isolated from pancreatic cancer cell lines, the workflow demonstrated higher sensitivity for previously reported EV markers of pancreatic cancer.
Collapse
Affiliation(s)
- Asia Botto
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
- Department of Chemistry and Industrial Chemistry, University of Pisa, 56124 Pisa, Italy
| | - Chiara De Cesari
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
- Nanoscience Institute, National Research Council, @NEST, 56127 Pisa, Italy
| | - Noa Ndimurwanko
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
- Scuola Normale Superiore, 56126 Pisa, Italy
| | - Francesco Finamore
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
| | - Francesco Greco
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
| | - Valentina Cappello
- Center for Materials Interfaces, Istituto Italiano di Tecnologia, 56025 Pontedera, Italy
| | | | - Benoit Immordino
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
- Scuola Superiore Sant'Anna, 56127 Pisa, Italy
| | - Vincenzo Lionetti
- Scuola Superiore Sant'Anna, 56127 Pisa, Italy
- UOSVD Anesthesia and Intensive Care, Fondazione Toscana G. Monasterio, 56124 Pisa, Italy
| | - Mauro Gemmi
- Center for Materials Interfaces, Istituto Italiano di Tecnologia, 56025 Pontedera, Italy
| | - Ilaria Tonazzini
- Nanoscience Institute, National Research Council, @NEST, 56127 Pisa, Italy
| | - Elisa Giovannetti
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
- Department of Medical Oncology, Cancer Center Amsterdam, VU University, 1081 HV Amsterdam, The Netherlands
| | - Liam A McDonnell
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
| |
Collapse
|
45
|
Tomiyama E, Fujita K, Matsuzaki K, Narumi R, Matsushita M, Hayashi Y, Hashimoto M, Kato T, Hatano K, Kawashima A, Minami T, Takao T, Takada S, Uemura H, Adachi J, Tomonaga T, Nonomura N. EIF2S1 in Urinary Extracellular Vesicles as a Novel Diagnostic Marker for Bladder Cancer. Cancer Med 2025; 14:e70964. [PMID: 40365898 PMCID: PMC12076193 DOI: 10.1002/cam4.70964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 04/17/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
BACKGROUND Urinary extracellular vesicles (uEVs), directly secreted from bladder cancer (BCa) cells, harbor potential for biomarker discovery. METHODS We performed proteomic analysis to explore and validate uEV-based diagnostic markers for BCa, with a focus on cytoplasmic EV proteins. Among the 1960 proteins identified by shotgun proteomics (tandem mass tag-labeled liquid chromatography-tandem mass spectrometry [LC-MS/MS]) of uEVs from seven patients with BCa and four healthy individuals, 17 cytoplasmic EV proteins were significantly elevated in the patients' urine (fold change > 1.5; p < 0.05). These 17 proteins were subsequently validated using targeted proteomics (selected reaction monitoring/multiple reaction monitoring) using urine samples from 49 and 48 patients with and without BCa, respectively, including those with non-BCa hematuria. RESULTS Ten measurable EV proteins remained significantly elevated in the urine of patients with BCa, with EV-EIF2S1 demonstrating the best diagnostic performance (area under the receiver operating characteristic [ROC] curve [AUC] [ROCAUC]: 0.83). Additionally, EV-EIF2S1 distinguished patients with BCa from those without BCa and hematuria in a suitable manner (ROCAUC: 0.92). Functional analysis of EIF2S1 in the BCa cell lines (T24 and 5637) showed that EIF2S1 knockdown markedly inhibited cell proliferation and induced cell cycle arrest and apoptosis, suggesting its essentiality for BCa cell growth and survival. CONCLUSIONS This study identified EV-EIF2S1 as a novel, uEV-based BCa diagnostic marker and demonstrated its functional significance in BCa cell growth and survival.
Collapse
Affiliation(s)
- Eisuke Tomiyama
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Kazutoshi Fujita
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
- Department of UrologyKindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Kyosuke Matsuzaki
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Ryohei Narumi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design ResearchNational Institutes of Biomedical Innovation, Health and NutritionIbarakiOsakaJapan
| | - Makoto Matsushita
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Yujiro Hayashi
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Mamoru Hashimoto
- Department of UrologyKindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Taigo Kato
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Koji Hatano
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Atsunari Kawashima
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| | - Takafumi Minami
- Department of UrologyKindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Tetsuya Takao
- Department of UrologyOsaka General Medical CenterOsakaOsakaJapan
| | - Shingo Takada
- Department of UrologyOsaka Police HospitalOsakaOsakaJapan
| | - Hirotsugu Uemura
- Department of UrologyKindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Jun Adachi
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design ResearchNational Institutes of Biomedical Innovation, Health and NutritionIbarakiOsakaJapan
| | - Takeshi Tomonaga
- Laboratory of Proteomics for Drug Discovery, Center for Drug Design ResearchNational Institutes of Biomedical Innovation, Health and NutritionIbarakiOsakaJapan
| | - Norio Nonomura
- Department of UrologyOsaka University Graduate School of MedicineSuitaOsakaJapan
| |
Collapse
|
46
|
Tao K, Tao K, Wang J. The potential mechanisms of extracellular vesicles in transfusion-related adverse reactions: Recent advances. Transfus Clin Biol 2025; 32:205-227. [PMID: 40180029 DOI: 10.1016/j.tracli.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Blood transfusion is an irreplaceable clinical treatment. Blood components are differentiated and stored according to specific guidelines. Storage temperatures and times vary depending on the blood component, but they all release extracellular vesicles (EVs) during storage. Although blood transfusions can be life-saving, they can also cause many adverse transfusion reactions, among which the effects of EVs are of increasing interest to researchers. EVs are submicron particles that vary in size, composition, and surface biomarkers, are encapsulated by a lipid bilayer, and are not capable of self-replication. EVs released by blood cells are important contributors to pathophysiologic states through proinflammatory, coagulant, and immunosuppressive effects, which in turn promote or inhibit the associated disease phenotype. Therefore, this review explores the potential mechanisms of hematopoietic-derived EVs in transfusion-associated adverse reactions and discusses the potential of the latest proteomics tools to be applied to the analysis of EVs in the field of transfusion medicine with a view to reducing the risk of blood transfusion.
Collapse
Affiliation(s)
- Keyi Tao
- Panzhihua University, Panzhihua 617000 Sichuan, China
| | - Keran Tao
- Institute of Medicine and Nursing, Hubei University of Medicine, Shiyan 442000 Hubei, China
| | - Jing Wang
- Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000 China.
| |
Collapse
|
47
|
Le Meur M, Pignatelli J, Blasi P, Palomo V. Nanoparticles targeting the central circadian clock: Potential applications for neurological disorders. Adv Drug Deliv Rev 2025; 220:115561. [PMID: 40120723 DOI: 10.1016/j.addr.2025.115561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Circadian rhythms and their involvement with various human diseases, including neurological disorders, have become an intense area of research for the development of new pharmacological treatments. The location of the circadian clock machinery in the central nervous system makes it challenging to reach molecular targets at therapeutic concentrations. In addition, a timely administration of the therapeutic agents is necessary to efficiently modulate the circadian clock. Thus, the use of nanoparticles in circadian clock dysfunctions may accelerate their clinical translation by addressing these two key challenges: enhancing brain penetration and/or enabling their formulation in chronodelivery systems. This review describes the implications of the circadian clock in neurological pathologies, reviews potential molecular targets and their modulators and suggests how the use of nanoparticle-based formulations could improve their clinical success. Finally, the potential integration of nanoparticles into chronopharmaceutical drug delivery systems will be described.
Collapse
Affiliation(s)
- Marion Le Meur
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain; Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum - Università di Bologna, 40127 Bologna, Italy
| | - Jaime Pignatelli
- Cajal Institute, Consejo Superior de Investigaciones Científicas (CSIC), 28002 Madrid, Spain; Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Paolo Blasi
- Dipartimento di Farmacia e Biotecnologie (FaBiT), Alma Mater Studiorum - Università di Bologna, 40127 Bologna, Italy.
| | - Valle Palomo
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), 28049 Madrid, Spain; Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28029 Madrid, Spain; Unidad de Nanobiotecnología asociada al Centro Nacional de Biotecnología (CNB-CSIC), 28049 Madrid, Spain.
| |
Collapse
|
48
|
Abbad L, Esteve E, Chatziantoniou C. Advances and challenges in kidney fibrosis therapeutics. Nat Rev Nephrol 2025; 21:314-329. [PMID: 39934355 DOI: 10.1038/s41581-025-00934-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
Chronic kidney disease (CKD) is a major global health burden that affects more than 10% of the adult population. Current treatments, including dialysis and transplantation, are costly and not curative. Kidney fibrosis, defined as an abnormal accumulation of extracellular matrix in the kidney parenchyma, is a common outcome in CKD, regardless of disease aetiology, and is a major cause of loss of kidney function and kidney failure. For this reason, research efforts have focused on identifying mediators of kidney fibrosis to inform the development of effective anti-fibrotic treatments. Given the prominent role of the transforming growth factor-β (TGFβ) family in fibrosis, efforts have focused on inhibiting TGFβ signalling. Despite hopes raised by the efficacy of this approach in preclinical models, translation into clinical practice has not met expectations. Antihypertensive and antidiabetic drugs slow the decline in kidney function and could slow fibrosis but, owing to the lack of technologies for in vivo renal imaging, their anti-fibrotic effect cannot be truly assessed at present. The emergence of new drugs targeting pro-fibrotic signalling, or enabling cell repair and cell metabolic reprogramming, combined with better stratification of people with CKD and the arrival of nanotechnologies for kidney-specific drug delivery, open up new perspectives for the treatment of this major public health challenge.
Collapse
Affiliation(s)
- Lilia Abbad
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Emmanuel Esteve
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Christos Chatziantoniou
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France.
| |
Collapse
|
49
|
Izhar M, Lesniak MS. Role of Extracellular Vesicles in the Pathogenesis of Brain Metastasis. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70051. [PMID: 40330713 PMCID: PMC12053894 DOI: 10.1002/jex2.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/03/2025] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Extracellular vesicles (EVs) are small particles released by various cells, including cancer cells. They play a significant role in the development of different cancers, including brain metastasis. These EVs transport biomolecular materials such as RNA, DNA, and proteins from tumour cells to other cells, facilitating the spread of primary tumours to the brain tissue. EVs interact with the endothelial cells of the blood-brain barrier (BBB), compromising its integrity and allowing metastatic cells to pass through easily. Additionally, EVs interact with various cells in the brain's microenvironment, creating a conducive environment for incoming metastatic cells. They also influence the immune system within this premetastatic environment, promoting the growth of metastatic cells. This review paper focuses on the research regarding the role of EVs in the development of brain metastasis, including their impact on disrupting the BBB, preparing the premetastatic environment, and modulating the immune system. Furthermore, the paper discusses the potential of EVs as diagnostic and prognostic biomarkers for brain metastasis.
Collapse
Affiliation(s)
- Muhammad Izhar
- Department of NeurosurgeryMassachusetts General Hospital, Harvard Medical SchoolBostonMassachusettsUSA
- Department of NeurosurgeryStanford University School of MedicineStanfordCaliforniaUSA
| | - Maciej S. Lesniak
- Department of Neurological SurgeryLou and Jean Malnati Brain Tumor Institute, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern UniversityChicagoIllinoisUSA
| |
Collapse
|
50
|
Yang Y, Zhang C, Li H, He Q, Xie J, Liu H, Cui F, Lei Z, Qin X, Liu Y, Xu M, Huang S, Zhang X. A review of molecular interplay between inflammation and cancer: The role of lncRNAs in pathogenesis and therapeutic potential. Int J Biol Macromol 2025; 309:142824. [PMID: 40187457 DOI: 10.1016/j.ijbiomac.2025.142824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 04/07/2025]
Abstract
The inflammatory microenvironment (IME) has been demonstrated to facilitate the initiation and progression of tumors throughout the inflammatory process. Simultaneously, cancer can initiate or intensify the inflammatory response, thereby promoting tumor progression. This review examines the dual role of long non-coding RNAs (lncRNAs) in the interplay between inflammation and cancer. LncRNA modulate inflammation-induced cancer by influencing the activation of signaling pathways (NF-κB, Wnt/β-catenin, mTOR, etc), microRNA (miRNA) sponging, protein interactions, interactions with immune cells, and encoding short peptides. In contrast, lncRNAs also impact cancer-induced inflammatory processes by regulating cytokine expression, mediating tumor-derived extracellular vesicles (EVs), modulating intracellular reactive oxygen species (ROS) levels, and facilitating metabolic reprogramming. Furthermore, the therapeutic potential of lncRNA and the challenges of clinical translation were explicitly discussed as well. Overall, this review aims to provide a comprehensive and systematic resource for future researchers investigating the impact of lncRNAs on inflammation and cancer.
Collapse
Affiliation(s)
- Yan Yang
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China; School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Chuxi Zhang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China
| | - Huacui Li
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China; Tangshan Institute of Southwest Jiaotong University, Tangshan, China
| | - Qin He
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Jiang Xie
- Department of Pediatrics, The Third People's Hospital of Chengdu, Chengdu, China
| | - Hongmei Liu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Fenfang Cui
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Ziqin Lei
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Xiaoyan Qin
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Ying Liu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China
| | - Min Xu
- Department of Pharmacy, The Third People's Hospital of Chengdu, Chengdu, China.
| | - Shuai Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, China.
| | - Xu Zhang
- Department of Pharmacy, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu University of TCM, Chengdu, China.
| |
Collapse
|