1
|
Lin Y, Wang J, Bu F, Zhang R, Wang J, Wang Y, Huang M, Huang Y, Zheng L, Wang Q, Hu X. Bacterial extracellular vesicles in the initiation, progression and treatment of atherosclerosis. Gut Microbes 2025; 17:2452229. [PMID: 39840620 DOI: 10.1080/19490976.2025.2452229] [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: 08/21/2024] [Revised: 11/13/2024] [Accepted: 01/07/2025] [Indexed: 01/23/2025] Open
Abstract
Atherosclerosis is the primary cause of cardiovascular and cerebrovascular diseases. However, current anti-atherosclerosis drugs have shown conflicting therapeutic outcomes, thereby spurring the search for novel and effective treatments. Recent research indicates the crucial involvement of oral and gastrointestinal microbiota in atherosclerosis. While gut microbiota metabolites, such as choline derivatives, have been extensively studied and reviewed, emerging evidence suggests that bacterial extracellular vesicles (BEVs), which are membrane-derived lipid bilayers secreted by bacteria, also play a significant role in this process. However, the role of BEVs in host-microbiota interactions remains insufficiently explored. This review aims to elucidate the complex communication mediated by BEVs along the gut-heart axis. In this review, we summarize current knowledge on BEVs, with a specific focus on how pathogen-derived BEVs contribute to the promotion of atherosclerosis, as well as how BEVs from gut symbionts and probiotics may mitigate its progression. We also explore the potential and challenges associated with engineered BEVs in the prevention and treatment of atherosclerosis. Finally, we discuss the benefits and challenges of using BEVs in atherosclerosis diagnosis and treatment, and propose future research directions to address these issues.
Collapse
Affiliation(s)
- Yuling Lin
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingyu Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fan Bu
- Institute of Hematology, Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Zhejiang University, Hangzhou, China
| | - Ruyi Zhang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Junhui Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yubing Wang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mei Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiyi Huang
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qian Wang
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Center for Clinical Laboratory, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiumei Hu
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
2
|
Batur A, Novak R, Salai G, Hrkač S, Ćosić V, Grgurević L. Extracellular vesicles in the pathogenesis and future diagnostics of oral squamous cell carcinoma. Future Sci OA 2025; 11:2461940. [PMID: 39920887 PMCID: PMC11812389 DOI: 10.1080/20565623.2025.2461940] [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: 09/11/2024] [Accepted: 01/13/2025] [Indexed: 02/09/2025] Open
Abstract
Extracellular vesicles are a group of heterogeneous particles secreted during both physiological and pathological conditions which serve in intercellular communication and play a role in the development and progression of oral squamous cell carcinoma, the most common malignant tumor of the head and neck with a high mortality rate. Extensive research is being conducted in order to determine the precise role of extracellular vesicles in oncogenic processes and to explore the possible application of extracellular vesicles as early tumor biomarkers. In this review, we aimed to systematize observed roles extracellular vesicles might play in organizing of tumor microenvironment, tumor invasion and metastasis, as well as the impact of extracellular vesicles on immune dysregulation and development of resistance to chemotherapeutics. Additionally, we summarized findings involving the potential use of extracellular vesicles cargo proteins as early disease biomarkers.
Collapse
Affiliation(s)
- Anđela Batur
- School of Dental Medicine, University of Zagreb, Zagreb, Croatia
| | - Ruđer Novak
- Center for Translational and Clinical Research, Department of Proteomics, University of Zagreb, School of Medicine, Zagreb, Croatia
- BIMIS – Biomedical Research Center Šalata, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Grgur Salai
- Department of Pulmonology, University Hospital Dubrava, Zagreb, Croatia
| | - Stela Hrkač
- Department of Clinical Immunology, Allergology and Rheumatology, University Hospital Dubrava, Zagreb, Croatia
| | - Vesna Ćosić
- Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Lovorka Grgurević
- Center for Translational and Clinical Research, Department of Proteomics, University of Zagreb, School of Medicine, Zagreb, Croatia
- BIMIS – Biomedical Research Center Šalata, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Anatomy, “Drago Perović”, University of Zagreb, School of Medicine, Zagreb, Croatia
| |
Collapse
|
3
|
Li LY, Liang SY, Cai MP, Ge JC, Tan HS, Wang CB, Xu B. Engineered extracellular vesicles as imaging biomarkers and therapeutic applications for urological diseases. Mater Today Bio 2025; 32:101646. [PMID: 40160248 PMCID: PMC11953971 DOI: 10.1016/j.mtbio.2025.101646] [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: 01/01/2025] [Revised: 02/23/2025] [Accepted: 03/07/2025] [Indexed: 04/02/2025] Open
Abstract
With the ever-increasing burden of urological diseases, the need for developing novel imaging biomarkers and therapeutics to manage these disorders has never been greater. Extracellular vesicles (EVs) are natural membranous nanoparticles and widely applied in both diagnostics and therapeutics for many diseases. A growing body of research has demonstrated that EVs can be engineered to enhance their efficiency, specificity, and safety. We systematically examine the strategies for achieving targeted delivery of EVs as well as the techniques for engineering them in this review, with a particular emphasis on cargo loading and transportation. Additionally, this review highlights and summarizes the wide range of imaging biomarkers and therapeutic applications of engineered EVs in the context of urological diseases, emphasizing the potential applications in urological malignancy and kidney diseases.
Collapse
Affiliation(s)
- Liao-Yuan Li
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Si-Yuan Liang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Mao-Ping Cai
- Department of Urology, Cancer Center, Fudan University, Shanghai, China
| | - Jian-Chao Ge
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hai-Song Tan
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Cheng-Bang Wang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Bin Xu
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
4
|
Chan HY, Wang Q, Howie A, Bucci J, Graham P, Li Y. Extracellular vesicle biomarkers redefine prostate cancer radiotherapy. Cancer Lett 2025; 616:217568. [PMID: 39978570 DOI: 10.1016/j.canlet.2025.217568] [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: 12/08/2024] [Revised: 02/04/2025] [Accepted: 02/16/2025] [Indexed: 02/22/2025]
Abstract
Radiotherapy (RT) remains a cornerstone in the treatment of prostate cancer (PCa). Extracellular vesicles (EVs), nano-sized particles secreted by cells, play important roles in intercellular communication within the tumour microenvironment (TME) and contribute to tumour growth, metastasis, and therapy resistance. Recent advancements demonstrate the potential of EVs as biomarkers for cancer diagnosis, prognosis, and treatment monitoring. Accumulating evidence supports the role of EVs in modulating RT outcomes by shaping the TME, mediating radioresistance, and influencing cancer metastasis. Despite substantial progress, challenges remain, including the heterogeneity of EV biogenesis, variability in cargo composition, and the absence of standardised methods for EV isolation and characterisation. While the therapeutic and diagnostic prospects of EVs in PCa management are promising, further research is needed to clarify the mechanisms through which EVs impact RT and to translate these findings into clinical practice. Incorporating EV research into PCa treatment paradigms could enhance diagnostic accuracy, enable real-time monitoring of RT responses, and support the development of new targeted therapeutic strategies. This review discusses recent progress in understanding EVs in the context of RT for PCa, focuses on their roles in modulating tumour growth, contributing to radioresistance within the TME, and facilitating the monitoring of RT efficacy and recurrence. In addition, the potential of EVs as biomarkers for liquid biopsy and their applications in enhancing radiosensitivity or overcoming radioresistance is also explored.
Collapse
Affiliation(s)
- Hei Yeung Chan
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Qi Wang
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Andrew Howie
- Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Joseph Bucci
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Peter Graham
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia
| | - Yong Li
- St George and Sutherland Clinical Campuses, School of Clinical Medicine, UNSW Sydney, Kensington, NSW, 2052, Australia; Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia.
| |
Collapse
|
5
|
Liao Z, Zeng J, Lin A, Zou Y, Zhou Z. Pre-treated mesenchymal stem cell-derived exosomes: A new perspective for accelerating spinal cord injury repair. Eur J Pharmacol 2025; 992:177349. [PMID: 39921061 DOI: 10.1016/j.ejphar.2025.177349] [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: 11/13/2024] [Revised: 01/24/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Spinal cord injury (SCI) is a devastating event for the central nervous system (CNS), often resulting in the loss of sensory and motor functions. It profoundly affects both the physiological and psychological well-being of patients, reducing their quality of life while also imposing significant economic pressure on families and the healthcare system. Due to the complex pathophysiology of SCI, effective treatments for promoting recovery remain scarce. Mesenchymal stem cell-derived exosomes (MSC-Exos) offer advantages such as low immunogenicity, good biocompatibility, and the ability to cross the blood-spinal cord barrier (BSCB). In preclinical studies, they have progressively shown efficacy in promoting SCI repair and functional recovery. However, the low yield and insufficient targeting of MSC-Exos limit their therapeutic efficacy. Currently, genetic engineering and other preprocessing techniques are being employed to optimize both the yield and functional properties of exosomes, thereby enhancing their therapeutic potential. Therefore, this paper provides an overview of the pathophysiology of SCI and the biogenesis of exosomes. It also summarizes current approaches to optimizing exosome performance. Additionally, it details the mechanisms through which optimized exosomes provide neuroprotection and explores the potential of combined treatments involving MSC-Exos and hydrogels.
Collapse
Affiliation(s)
- Zhiqiang Liao
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Junjian Zeng
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Aiqing Lin
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Yu Zou
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China
| | - Zhidong Zhou
- Department of Anesthesiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, 330006, Nanchang, Jiangxi, China; Jiangxi Province Key Laboratory of Anesthesiology, 1# Minde Road, 330006, Nanchang City, Jiangxi Province, China.
| |
Collapse
|
6
|
Khorrami-Nejad M, Hashemian H, Majdi A, Jadidi K, Aghamollaei H, Hadi A. Application of stem cell-derived exosomes in anterior segment eye diseases: A comprehensive update review. Ocul Surf 2025; 36:209-219. [PMID: 39884389 DOI: 10.1016/j.jtos.2025.01.012] [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: 09/12/2024] [Revised: 01/11/2025] [Accepted: 01/23/2025] [Indexed: 02/01/2025]
Abstract
Mesenchymal stem cell (MSC) therapy has emerged as a promising approach for addressing various eye-related conditions. Yet, its clinical application faces challenges due to issues such as limited biocompatibility and difficulties in effectively delivering treatment to specific ocular tissues. Recent studies have shifted attention towards MSC-derived exosomes, which share similar regenerative, reparative, and immunomodulatory capabilities with their origin cells. This review delves into the latest research on the use of MSC-derived exosomes for treating anterior segment diseases of the eye. It explores the exosomes' composition, biological functions, and the methods used for their isolation, as well as their roles in disease progression, diagnosis, and therapy. The review critically assesses the therapeutic advantages and mechanisms of action of MSC-derived exosomes in treating conditions like dry eye disease, Sjogren's syndrome, keratoconus, corneal lesions, and corneal allograft rejection. Additionally, it discusses the obstacles and future prospects of employing MSC-derived exosomes as innovative therapies for anterior segment eye diseases. This comprehensive overview underscores the significant potential of MSC-derived exosomes in transforming the treatment paradigm for anterior segment eye disorders, while also highlighting the necessity for further research to enhance their clinical application.
Collapse
Affiliation(s)
- Masoud Khorrami-Nejad
- Optometry Department, School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran; Translational Ophthalmology Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| | - Hesam Hashemian
- Translational Ophthalmology Research Center, Farabi Eye Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Majdi
- Optical Techniques Department, College of Health and Medical Techniques, Al-Mustaqbal University, 51001, Babylon, Iraq
| | - Khosrow Jadidi
- Vision Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Hossein Aghamollaei
- Vision Health Research Center, Semnan University of Medical Sciences, Semnan, Iran
| | - Ali Hadi
- Optometry Department, School of Rehabilitation, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Yadav K, Sahu KK, Sucheta, Minz S, Pradhan M. Unlocking exosome therapeutics: The critical role of pharmacokinetics in clinical applications. Tissue Cell 2025; 93:102749. [PMID: 39904192 DOI: 10.1016/j.tice.2025.102749] [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: 10/17/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Exosomes are microscopic vesicles released by cells that transport various biological materials and play a vital role in intercellular communication. When they are engineered, they serve as efficient delivery systems for therapeutic agents, making it possible to precisely deliver active pharmaceutical ingredients to organs, tissues, and cells. Exosomes' pharmacokinetics, or how they are transported and metabolized inside the body, is affected by several factors, including their source of origination and the proteins in their cell membranes. The pharmacokinetics and mobility of both native and modified exosomes are being observed in living organisms using advanced imaging modalities such as in vitro-in vivo simulation, magnetic resonance imaging, and positron emission tomography. Establishing comprehensive criteria for the investigation of exosomal pharmacokinetic is essential, given its increasing significance in both therapy and diagnostics. To obtain a thorough understanding of exosome intake, distribution, metabolism, and excretion, molecular imaging methods are crucial. The development of industrial processes and therapeutic applications depends on the precise measurement of exosome concentration in biological samples. To ensure a seamless incorporation of exosomes into clinical practice, as their role in therapeutics grows, it is imperative to conduct a complete assessment of their pharmacokinetics. This review provides a brief on how exosome-based research is evolving and the need for pharmacokinetic consideration to realize the full potential of these promising new therapeutic approaches.
Collapse
Affiliation(s)
- Krishna Yadav
- Rungta College of Pharmaceutical Sciences and Research, Kohka Road, Kurud, Bhilai, Chhattisgarh 491024, India
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 11 122103, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | | |
Collapse
|
8
|
Otieno MO, Powrózek T, Garcia-Foncillas J, Martinez-Useros J. The crosstalk within tumor microenvironment and exosomes in pancreatic cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189308. [PMID: 40180303 DOI: 10.1016/j.bbcan.2025.189308] [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: 10/01/2024] [Revised: 03/26/2025] [Accepted: 03/27/2025] [Indexed: 04/05/2025]
Abstract
Pancreatic cancer is one of the most malignant tumors with a grim prognosis. Patients develop chemoresistance that drastically decreases their survival. The chemoresistance is mainly attributed to deficient vascularization of the tumor, intratumoral heterogeneity and pathophysiological barrier due to the highly desmoplastic tumor microenvironment. The interactions of cells that constitute the tumor microenvironment change its architecture into a cancer-permissive environment and stimulate cancer development, metastasis and treatment response. The cell-cell communication in the tumor microenvironment is often mediated by exosomes that harbour a diverse repertoire of molecular cargo, such as proteins, lipids, and nucleic acid, including messenger RNAs, non-coding RNAs and DNA. Therefore, exosomes can serve as potential targets as biomarkers and improve the clinical management of pancreatic cancer to overcome chemoresistance. This review critically elucidates the role of exosomes in cell-cell communication within the tumor microenvironment and how these interactions can orchestrate chemoresistance.
Collapse
Affiliation(s)
- Michael Ochieng' Otieno
- Translational Oncology Division, OncoHealth Institute, Health Research Institute Fundación Jimenez Diaz, Fundación Jimenez Díaz University Hospital, Universidad Autonoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain
| | - Tomasz Powrózek
- Department of Human Physiology of the Chair of Preclinical Sciences, Medical University in Lublin, 20-080 Lublin, Poland
| | - Jesus Garcia-Foncillas
- Translational Oncology Division, OncoHealth Institute, Health Research Institute Fundación Jimenez Diaz, Fundación Jimenez Díaz University Hospital, Universidad Autonoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Medical Oncology Department, Fundación Jimenez Diaz University Hospital, 28040, Madrid, Spain
| | - Javier Martinez-Useros
- Translational Oncology Division, OncoHealth Institute, Health Research Institute Fundación Jimenez Diaz, Fundación Jimenez Díaz University Hospital, Universidad Autonoma de Madrid (IIS-FJD, UAM), 28040 Madrid, Spain; Area of Physiology, Department of Basic Health Sciences, Faculty of Health Sciences, Rey Juan Carlos Univer-Sity, 28922 Madrid, Spain.
| |
Collapse
|
9
|
Wang W, Cheng Z, Yu M, Liu K, Duan H, Zhang Y, Huang X, Li M, Li C, Hu Y, Luo Z, Liu M. Injectable ECM-mimetic dynamic hydrogels abolish ferroptosis-induced post-discectomy herniation through delivering nucleus pulposus progenitor cell-derived exosomes. Nat Commun 2025; 16:3131. [PMID: 40169595 PMCID: PMC11961689 DOI: 10.1038/s41467-025-58447-5] [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: 09/04/2024] [Accepted: 03/21/2025] [Indexed: 04/03/2025] Open
Abstract
Discectomy-induced ferroptosis of nucleus pulposus cells (NPCs) contributes to postoperative lumbar disc herniation (LDH) recurrence and intervertebral disc degeneration (IDD). We discover that nucleus pulposus progenitor cells (NPPCs) could imprint ferroptosis resistance into NPCs through exosome-dependent intercellular transmission of miR-221-3p. Based on these findings, we first develop synthetically-tailored NPPC-derived exosomes with enhanced miR-221-3p expression and NPC uptake capacity, which are integrated into an injectable hydrogel based on extracellular matrix (ECM) analogues. The ECM-mimetic hydrogel (HACS) serves as a biomimetic filler for the post-operative care of herniated discs, which could be facilely injected into the discectomy-established nucleus pulposus (NP) cavity for localized treatment. HACS-mediated in-situ exosome release in the NP cavity enables marked ferroptosis inhibition in NPCs that not only prevents LDH recurrence but also reverses the IDD symptoms, leading to robust restoration of NP structure and functions. In summary, this study offers a promising approach for treating disc herniation.
Collapse
Affiliation(s)
- Wenkai Wang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- Department of Orthopedics, General Hospital of PLA Xizang Military Area Command, Lhasa, Xizang, China
| | - Zhuo Cheng
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Miao Yu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ke Liu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hongli Duan
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yang Zhang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xinle Huang
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Menghuan Li
- School of Life Science, Chongqing University, Chongqing, China
| | - Changqing Li
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China.
| | - Zhong Luo
- School of Life Science, Chongqing University, Chongqing, China.
| | - Minghan Liu
- Department of Orthopedics, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
10
|
Gao B, Huang X, Fu J, Chen L, Deng Z, Wang S, Zhu Y, Xu C, Zhang Y, Zhang M, Chen L, Cui M, Zhang M. Oral administration of Momordica charantia-derived extracellular vesicles alleviates ulcerative colitis through comprehensive renovation of the intestinal microenvironment. J Nanobiotechnology 2025; 23:261. [PMID: 40170075 PMCID: PMC11959773 DOI: 10.1186/s12951-025-03346-6] [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: 02/07/2025] [Accepted: 03/23/2025] [Indexed: 04/03/2025] Open
Abstract
BACKGROUND Ulcerative colitis (UC) is an inflammatory bowel disease (IBD), accompanied by intense inflammation, oxidative stress, and intestinal microbiota dysbiosis. Current treatments using chemotherapeutic drugs or immunosuppressants have limited effectiveness and side effects. Therefore, the development of safe, effective, and multi-targeting therapies for IBD is of great importance. Momordica charantia exhibits antioxidant, anti-inflammatory, and intestinal microbiota-regulating properties, suggesting that Momordica charantia-derived extracellular vesicles (MCEVs) have the potential for UC management. RESULTS We extracted MCEVs using differential centrifugation and density gradient centrifugation. The results showed that MCEVs possessed high purity, even particle size, and excellent stability. In vitro, MCEVs were shown to inhibit macrophage inflammatory responses, scavenge reactive oxygen species (ROS), and protect cells from oxidative damage. Transcriptomics analysis revealed that MCEVs may alleviate mitochondria-dependent apoptosis by safeguarding the integrity of the mitochondrial structure and regulating the expression of apoptosis-related proteins. Furthermore, all components of MCEVs contributed to their pharmacological activity. In vivo, MCEVs had better retention in the inflamed colon and significantly alleviated UC through a comprehensive renovation of the intestinal microenvironment. CONCLUSION These findings suggested that MCEVs own considerable potential as natural nanotherapeutics for UC treatment.
Collapse
Affiliation(s)
- Bowen Gao
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Xiaoling Huang
- Department of Gastroenterology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang Uygur Autonomous Region, 830001, China
| | - Junlong Fu
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Liyuan Chen
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Zhichao Deng
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Shuhui Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yuanyuan Zhu
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Chenxi Xu
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Yujie Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Mingxin Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, 710077, China
| | - Lina Chen
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China
| | - Manli Cui
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi, 710077, China.
| | - Mingzhen Zhang
- School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, China.
| |
Collapse
|
11
|
Sabatke B, Rossi IV, Bonato L, Fucio S, Cortés A, Marcilla A, Ramirez MI. Host-Pathogen Cellular Communication: The Role of Dynamin, Clathrin, and Macropinocytosis in the Uptake of Giardia-Derived Extracellular Vesicles. ACS Infect Dis 2025. [PMID: 40155351 DOI: 10.1021/acsinfecdis.4c00996] [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: 04/01/2025]
Abstract
Giardia intestinalis, a protozoan causing giardiasis, disrupts gastrointestinal health through complex host-parasite interactions. This study explores the differential uptake mechanisms of extracellular vesicles (EVs) derived from Giardia (gEVs), host cells (hEVs), and the host-parasite interaction (intEVs) in intestinal Caco-2 cells. Results show that intEVs are internalized more rapidly than gEVs and hEVs, underscoring their pivotal role in pathogenesis. To delineate uptake pathways, various endocytosis inhibitors were applied, and clathrin-mediated endocytosis inhibition using monodansylcadaverine (MDC) significantly reduced intEV and gEV uptake, confirming the role of clathrin-mediated endocytosis (CME). The use of dynasore, a dynamin inhibitor, strongly reduced the internalization of all EV types, demonstrating that uptake is dynamin-dependent. In contrast, methyl-β-cyclodextrin (MβCD), which disrupts lipid rafts and caveolae-mediated pathways, had no effect on EV uptake, indicating that caveolae are not involved in this process. Furthermore, inhibition of Na+/H+ exchange and phosphoinositide 3-kinase activity, both essential for macropinocytosis, also led to a significant reduction in intEV internalization. These findings strongly support that gEVs are internalized primarily through a dynamin- and clathrin-dependent pathway, independent of caveolae and lipid rafts, but modulated by tyrosine kinase signaling and macropinocytosis. These insights into selective and comprehensive inhibition pathways offer promising therapeutic targets to mitigate giardiasis.
Collapse
Affiliation(s)
- Bruna Sabatke
- Graduate Program in Microbiology, Pathology and Parasitology, Federal University of Paraná, Curitiba, PR 81350-010, Brazil
- EVAHPI-Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, PR 81350-010, Brazil
| | - Izadora V Rossi
- Graduate Program in Microbiology, Pathology and Parasitology, Federal University of Paraná, Curitiba, PR 81350-010, Brazil
- EVAHPI-Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, PR 81350-010, Brazil
| | - Leticia Bonato
- Graduate Program in Microbiology, Pathology and Parasitology, Federal University of Paraná, Curitiba, PR 81350-010, Brazil
- EVAHPI-Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, PR 81350-010, Brazil
| | - Sarah Fucio
- EVAHPI-Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, PR 81350-010, Brazil
- Graduate Program in Cell and Molecular Biology, Federal University of Paraná, Curitiba, PR 81350-010, Brazil
| | - Alba Cortés
- Department of Parasitology and Cellular Biology, Faculty of Pharmacy, University of Valencia, Valencia 46010, Spain
| | - Antonio Marcilla
- Department of Parasitology and Cellular Biology, Faculty of Pharmacy, University of Valencia, Valencia 46010, Spain
| | - Marcel I Ramirez
- EVAHPI-Extracellular Vesicles and Host-Parasite Interactions Research Group, Carlos Chagas Institute (Fiocruz-PR), Curitiba, PR 81350-010, Brazil
| |
Collapse
|
12
|
Abdulmonem WA, Ahsan M, Mallick AK, Mohamed AH, Waggiallah HA, Shafie A, Alzahrani HS, Ashour AA, Rab SO, Mirdad MT, Ali HTO. The Role of Exosomal miRNAs in Female Infertility: Therapeutic Potential and Mechanisms of Action. Stem Cell Rev Rep 2025:10.1007/s12015-025-10869-w. [PMID: 40126819 DOI: 10.1007/s12015-025-10869-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 03/26/2025]
Abstract
Reproductive disorders, including preeclampsia (PE), endometriosis, premature ovarian failure (POF), and polycystic ovary syndrome (PCOS), present substantial challenges to women's reproductive health. Exosomes (EXOs) are cell-derived vesicles containing molecules that influence target cells' gene expression and cellular behavior. Among their cargo, microRNAs (miRNAs)-short, non-coding RNAs typically 19-25 nucleotides in length-play a crucial role in post-transcriptional gene regulation and have been extensively studied for their therapeutic potential. miRNAs are considered therapeutic targets because they regulate key cellular pathways such as proliferation, apoptosis, angiogenesis, and tissue repair. This review examines the role of exosomal miRNAs from sources such as mesenchymal stem cells (MSCs), plasma, and amniotic fluid in female reproductive disorders, including PE, POF, PCOS, and endometriosis. We discuss their biological origins, mechanisms of miRNA sorting and packaging, and their therapeutic applications in modulating disease progression. By categorizing miRNAs according to their beneficial or detrimental effects in specific conditions, we aim to simplify the understanding of their roles in female infertility.
Collapse
Affiliation(s)
- Waleed Al Abdulmonem
- Department of Pathology, College of Medicine, Qassim University, Buraydah, Saudi Arabia
| | - Marya Ahsan
- Department of Pharmacology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, 13317, Saudi Arabia
| | - Ayaz Khurram Mallick
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Asma'a H Mohamed
- Department of Optometry Techniques, Technical College Al-Mussaib, Al-Furat Al-Awsat Technical University, Najaf, Iraq.
| | - Hisham Ali Waggiallah
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Alkharj, 11942, Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, P.O.Box 11099, Taif, 21944, Saudi Arabia
| | - Hassan Swed Alzahrani
- Counseling Healthy Marriage, Jeddah Regional Laboratory, Jeddah First Cluster , Jeddah, Saudi Arabia
| | - Amal Adnan Ashour
- Department of Oral & Maxillofacial Surgery and Diagnostic Sciences, Faculty of Dentistry, Taif University, P.O. Box 11099, Taif, 21944, Saudi Arabia
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed Tarek Mirdad
- Medical Intern MBBS, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Hatim T O Ali
- Obstetrics and Gynecology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
13
|
Kundu S, Guo J, Islam MS, Rohokale R, Jaiswal M, Guo Z. A New Strategy to Functionalize Exosomes via Enzymatic Engineering of Surface Glycans and its Application to Profile Exosomal Glycans and Endocytosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2415942. [PMID: 40106306 DOI: 10.1002/advs.202415942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 03/01/2025] [Indexed: 03/22/2025]
Abstract
Exosomes are membrane-enclosed nanoparticles secreted by cells to mediate intercellular communication. Hence, functionalized exosomes are powerful tools in biology and medicine, and efficient methods to functionalize exosomes are highly desired. In this work, a novel approach is developed to modify and functionalize exosomes based on enzymatic engineering of their surface glycans. It employs a sialyltransferase and an azide-modified sialyl donor to enzymatically install azido-sialic acids onto exosomal glycans. The azide tags serve as universal molecular handles to attach various probes, e.g., biotin, protein, fluorophore, etc., by simple and biocompatible click chemistry. This approach is easy and effective, and the modified exosomes are readily retrieved from the plate, enabling the production of functional exosomes in practical scales for various studies and applications. The functionalized exosomes obtained are employed to profile exosomal glycans, disclosing the diverse glycosylation patterns of exosomes of different origins. They also facilitated comprehensive investigations on the cellular uptake of exosomes to disclose macropinocytosis as the main and general uptake route, while other endocytosis pathways are also partially involved in specific exosomes. Additionally, the new exosome functionalization approach has been demonstrated to be widely applicable to exosomes of different origins.
Collapse
Affiliation(s)
- Sayan Kundu
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
| | - Jiatong Guo
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
| | - Md Shamiul Islam
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
| | - Rajendra Rohokale
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
| | - Mohit Jaiswal
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
| | - Zhongwu Guo
- Department of Chemistry, University of Florida, Gainesville, FL, 32611, USA
- UF Health Cancer Center, University of Florida, Gainesville, FL, 32611, USA
| |
Collapse
|
14
|
Yoo D, Jung SY, Go D, Park JY, You DG, Jung WK, Li Y, Ding J, Park JH, Um W. Functionalized extracellular vesicles of mesenchymal stem cells for regenerative medicine. J Nanobiotechnology 2025; 23:219. [PMID: 40102934 PMCID: PMC11921732 DOI: 10.1186/s12951-025-03300-6] [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: 06/09/2024] [Accepted: 03/06/2025] [Indexed: 03/20/2025] Open
Abstract
Stem cell-derived extracellular vesicles (EVs) have emerged as a safe and potent alternative to regenerative medicine in recent decades. Furthermore, the adjustment of EV functions has been recently enabled by certain stem cell preconditioning methods, providing an exceptional opportunity to enhance the therapeutic potential or confer additional functions of stem cell-derived EVs. In this review, we discuss the recent progress of functionalized EVs, based on stem cell preconditioning, for treating various organ systems, such as the musculoskeletal system, nervous system, integumentary system, cardiovascular system, renal system, and respiratory system. Additionally, we summarize the expected outcomes of preconditioning methods for stem cells and their EVs. With recent progress, we suggest considerations and future directions for developing personalized medicine based on preconditioned stem cell-derived EVs.
Collapse
Affiliation(s)
- Donghyeon Yoo
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Se Young Jung
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dabin Go
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Ji Yeong Park
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea
| | - Dong Gil You
- Department of Chemical Engineering & Biotechnology, Tech University of Korea, Siheung, 15073, Republic of Korea
| | - Won-Kyo Jung
- Marine Integrated Biomedical Technology Center, The National Key Research Institutes in Universities, Pukyong National University, Busan, 48513, Republic of Korea
- Major of Biomedical Engineering, Division of Smart Healthcare, College of Information Technology and Convergence and New-senior Healthcare Innovation Center (BK21 Plus), Pukyong National University, Busan, 48513, Republic of Korea
| | - Yuce Li
- College of Life Science and Health, Wuhan University of Science and Technology (WUST), Wuhan, 430065, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, P. R. China
| | - Jae Hyung Park
- School of Chemical Engineering, College of Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea.
| | - Wooram Um
- Department of Biotechnology, College of Fisheries Science, Pukyong National University, Busan, 48513, Republic of Korea.
| |
Collapse
|
15
|
Alfandari D, Rosenhek-Goldian I, Kozela E, Nevo R, Senprún MB, Moisieiev A, Sogauker N, Azuri I, Gelman S, Kiper E, Ben Hur D, Dharan R, Sorkin R, Porat Z, Morandi MI, Regev-Rudzki N. Host Immune Cell Membrane Deformability Governs the Uptake Route of Malaria-Derived Extracellular Vesicles. ACS NANO 2025; 19:9760-9778. [PMID: 40030053 PMCID: PMC11924330 DOI: 10.1021/acsnano.4c07503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
The malaria parasite, Plasmodium falciparum, secretes extracellular vesicles (EVs) to facilitate its growth and to communicate with the external microenvironment, primarily targeting the host's immune cells. How parasitic EVs enter specific immune cell types within the highly heterogeneous pool of immune cells remains largely unknown. Using a combination of imaging flow cytometry and advanced fluorescence analysis, we demonstrated that the route of uptake of parasite-derived EVs differs markedly between host T cells and monocytes. T cells, which are components of the adaptive immune system, internalize parasite-derived EVs mainly through an interaction with the plasma membrane, whereas monocytes, which function in the innate immune system, take up these EVs via endocytosis. The membranal/endocytic balance of EV internalization is driven mostly by the amount of endocytic incorporation. Integrating atomic force microscopy with fluorescence data analysis revealed that internalization depends on the biophysical properties of the cell membrane rather than solely on molecular interactions. In support of this, altering the cholesterol content in the cell membrane tilted the balance in favor of one uptake route over another. Our results provide mechanistic insights into how P. falciparum-derived EVs enter into diverse host cells. This study highlights the sophisticated cell-communication tactics used by the malaria parasite.
Collapse
Affiliation(s)
- Daniel Alfandari
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Irit Rosenhek-Goldian
- Department of Chemical Research Support, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ewa Kozela
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Reinat Nevo
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Marcela Bahlsen Senprún
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Anton Moisieiev
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Noam Sogauker
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ido Azuri
- Bioinformatics Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Samuel Gelman
- Bioinformatics Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Edo Kiper
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Daniel Ben Hur
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Raviv Dharan
- Raymond and Beverly Sackler Faculty of Exact Sciences, School of Chemistry, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Raya Sorkin
- Raymond and Beverly Sackler Faculty of Exact Sciences, School of Chemistry, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ziv Porat
- Flow cytometry Unit, Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Mattia I Morandi
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Science, Prague 160-00, Czech Republic
- IMol Polish Academy of Sciences, Warsaw 02-247, Poland
| | - Neta Regev-Rudzki
- Department of Biomolecular Sciences, Faculty of Biochemistry, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
16
|
Wang Z, Li F, Liu W. Extracellular vesicles in endometrial-related diseases: role, potential and challenges. PeerJ 2025; 13:e19041. [PMID: 40093416 PMCID: PMC11910146 DOI: 10.7717/peerj.19041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Endometrial dysfunction underlies many common gynecologic disorders, such as endometriosis, endometrial cancer, intrauterine adhesions, and endometritis, which affect many women around the world. Extracellular vesicles play an important role in the pathophysiologic process of endometrial-related diseases. Extracellular vesicles are released by cells, which usually act as a form of intercellular communication, affecting biological processes such as fibrosis, angiogenesis, cell proliferation, and inflammatory responses by transferring their own proteins, lipids, RNA transcripts, and DNA for messaging, and play a key role in physiological dynamic homeostasis and disease development. This review combines the studies of the last decade, using the sub-description method to introduce the application of different sources of extracellular vesicles in the diagnosis and treatment of related diseases, and discusses the challenges faced by extracellular vesicles in the diagnostic and therapeutic application of endometriosis-related diseases, with the aim of contributing to our understanding of the mechanism of action of extracellular vesicles and their therapeutic roles, so as to provide a reference for the development of endometriosis-related diseases, as well as their prognosis and treatment.
Collapse
Affiliation(s)
- Zilu Wang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Feng Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Wenqiong Liu
- Shandong Provincial Hospital of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
17
|
Hirosawa KM, Sato Y, Kasai RS, Yamaguchi E, Komura N, Ando H, Hoshino A, Yokota Y, Suzuki KGN. Uptake of small extracellular vesicles by recipient cells is facilitated by paracrine adhesion signaling. Nat Commun 2025; 16:2419. [PMID: 40075063 PMCID: PMC11903687 DOI: 10.1038/s41467-025-57617-9] [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: 04/16/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Small extracellular vesicles (sEVs) play crucial roles in intercellular communication. However, the internalization of individual sEVs by recipient cells has not been directly observed. Here, we examined these mechanisms using state-of-the-art imaging techniques. Single-molecule imaging shows that tumor-derived sEVs can be classified into several subtypes. Simultaneous single-sEV particle tracking and observation of super-resolution movies of membrane invaginations in living cells reveal that all sEV subtypes are internalized via clathrin-independent endocytosis mediated by galectin-3 and lysosome-associated membrane protein-2C, while some subtypes that recruited raft markers are internalized through caveolae. Integrin β1 and talin-1 accumulate in recipient cell plasma membranes beneath all sEV subtypes. Paracrine, but not autocrine, sEV binding triggers Ca2+ mobilization induced by the activation of Src family kinases and phospholipase Cγ. Subsequent Ca2+-induced activation of calcineurin-dynamin promotes sEV internalization, leading to the recycling pathway. Thus, we clarified the detailed mechanisms of sEV internalization driven by paracrine adhesion signaling.
Collapse
Affiliation(s)
- Koichiro M Hirosawa
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
| | - Yusuke Sato
- Department of Chemistry, Graduate School of Science, Tohoku University, Sendai, 980-8578, Japan
| | - Rinshi S Kasai
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, 104-0045, Japan
| | - Eriko Yamaguchi
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
| | - Naoko Komura
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
| | - Hiromune Ando
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, 606-8501, Japan
- Innovation Research Center for Quantum Medicine. Graduate School of Medicine, Gifu University, Gifu, 501-1193, Japan
| | - Ayuko Hoshino
- Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, 153-8904, Japan
- Inamori Research Institute for Science, Inamori Foundation, Kyoto, 600-8411, Japan
| | - Yasunari Yokota
- Department of Electrical, Electronics and Computer Engineering, Faculty of Engineering, Gifu University, Gifu, 501-1193, Japan
| | - Kenichi G N Suzuki
- Institute for Glyco-core Research (iGCORE), Gifu University, Gifu, 501-1193, Japan.
- Division of Advanced Bioimaging, National Cancer Center Research Institute (NCCRI), Tokyo, 104-0045, Japan.
- Institute for Integrated Cell-Material Sciences, Kyoto University, Kyoto, 606-8501, Japan.
- Innovation Research Center for Quantum Medicine. Graduate School of Medicine, Gifu University, Gifu, 501-1193, Japan.
| |
Collapse
|
18
|
Ten A, Yudintceva N, Samochernykh K, Combs SE, Jha HC, Gao H, Shevtsov M. Post-Secretion Processes and Modification of Extracellular Vesicles. Cells 2025; 14:408. [PMID: 40136657 PMCID: PMC11940929 DOI: 10.3390/cells14060408] [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: 12/20/2024] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Extracellular vesicles (EVs) are an important mediator of intercellular communication and the regulation of processes occurring in cells and tissues. The processes of EVs secretion by cells into the extracellular space (ECS) leads to their interaction with its participants. The ECS is a dynamic structure that also takes direct part in many processes of intercellular communication and regulation. Changes in the ECS can also be associated with pathological processes, such as increased acidity during the development of solid tumors, changes in the composition and nature of the organization of the extracellular matrix (ECM) during fibroblast activation, an increase in the content of soluble molecules during necrosis, and other processes. The interaction of these two systems, the EVs and the ESC, leads to structural and functional alteration in both participants. In the current review, we will focus on these alterations in the EVs which we termed post-secretory modification and processes (PSMPs) of EVs. PSPMs can have a significant effect on the immediate cellular environment and on the spread of the pathological process in the body as a whole. Thus, it can be assumed that PSPMs are one of the important stages in the regulation of intercellular communication, which has significant differences in the norm and in pathology.
Collapse
Affiliation(s)
- Artem Ten
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia; (A.T.); (N.Y.)
| | - Natalia Yudintceva
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia; (A.T.); (N.Y.)
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia;
| | - Konstantin Samochernykh
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia;
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany;
| | - Hem Chandra Jha
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453552, India;
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China;
| | - Maxim Shevtsov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia; (A.T.); (N.Y.)
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia;
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum Rechts der Isar, Ismaninger Str. 22, 81675 Munich, Germany;
| |
Collapse
|
19
|
López RR, Ben El Khyat CZ, Chen Y, Tsering T, Dickinson K, Bustamante P, Erzingatzian A, Bartolomucci A, Ferrier ST, Douanne N, Mounier C, Stiharu I, Nerguizian V, Burnier JV. A synthetic model of bioinspired liposomes to study cancer-cell derived extracellular vesicles and their uptake by recipient cells. Sci Rep 2025; 15:8430. [PMID: 40069225 PMCID: PMC11897354 DOI: 10.1038/s41598-025-91873-5] [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: 12/03/2024] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Extracellular vesicles (EVs) are secreted by most cell types and play a central role in cell-cell communication. These naturally occurring nanoparticles have been particularly implicated in cancer, but EV heterogeneity and lengthy isolation methods with low yield make them difficult to study. To circumvent the challenges in EV research, we aimed to develop a unique synthetic model by engineering bioinspired liposomes to study EV properties and their impact on cellular uptake. We produced EV-like liposomes mimicking the physicochemical properties as cancer EVs. First, using a panel of cancer and non-cancer cell lines, small EVs were isolated by ultracentrifugation and characterized by dynamic light scattering (DLS) and nanoparticle tracking analysis (NTA). Cancer EVs ranged in mean size from 107.9 to 161 nm by NTA, hydrodynamic diameter from 152 to 355 nm by DLS, with a zeta potential ranging from - 25 to -6 mV. EV markers TSG101 and CD81 were positive on all EVs. Using a microfluidics bottom-up approach, liposomes were produced using the nanoprecipitation method adapted to micromixers developed by our group. A library of liposome formulations was created that mimicked the ranges of size (90-222 nm) and zeta potential (anionic [-47 mV] to neutral [-1 mV]) at a production throughput of up to 41 mL/h and yielding a concentration of 1 × 1012 particles per mL. EV size and zeta potential were reproduced by controlling the flow conditions and lipid composition set by a statistical model based on the response surface methodology. The model was fairly accurate with an R-squared > 70% for both parameters between the targeted EV and the obtained liposomes. Finally, the internalization of fluorescently labeled EV-like liposomes was assessed by confocal microscopy and flow cytometry, and correlated with decreasing liposome size and less negative zeta potential, providing insights into the effects of key EV physicochemical properties. Our data demonstrated that liposomes can be used as a powerful synthetic model of EVs. By mimicking cancer cell-derived EV properties, the effects on cellular internalization can be assessed individually and in combination. Taken together, we present a novel system that can accelerate research on the effects of EVs in cancer models.
Collapse
Grants
- 312831, 344929, 306252, 330312, 330509 Fonds de Recherche du Québec - Santé
- 312831, 344929, 306252, 330312, 330509 Fonds de Recherche du Québec - Santé
- 312831, 344929, 306252, 330312, 330509 Fonds de Recherche du Québec - Santé
- 190179 Canadian Institutes for Health Research
- 190179 Canadian Institutes for Health Research
- 177808 National Sciences and Engineering Research Council of Canada (NSERC)
- NFRFE-2019-01587 Government of Canada's New Frontiers in Research Fund (NFRF)
- Government of Canada’s New Frontiers in Research Fund (NFRF)
Collapse
Affiliation(s)
- Rubén R López
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
- Department of Electrical Engineering, École de Technologie supérieure, 1100 Notre Dame West, Montreal, QC, H3C 1K3, Canada
| | - Chaymaa Zouggari Ben El Khyat
- Department of Electrical Engineering, École de Technologie supérieure, 1100 Notre Dame West, Montreal, QC, H3C 1K3, Canada
| | - Yunxi Chen
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Thupten Tsering
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Kyle Dickinson
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
| | - Prisca Bustamante
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Armen Erzingatzian
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
| | - Alexandra Bartolomucci
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Sarah Tadhg Ferrier
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Noélie Douanne
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada
- Department of Pathology, McGill University, Quebec, Canada
| | - Catherine Mounier
- Department of biological sciences, Université du Québec à Montréal, 141 avenue du président Kennedy, Montreal, QC, H2X 1Y4, Canada
- Department of Mechanical, Industrial and Aerospace Engineering, Concordia University, 1455 de Maisonneuve Blvd. West, Montreal, QC, H3G 1M8, Canada
| | - Ion Stiharu
- Gerald Bronfman Department of Oncology, McGill University, 5100 de Maisonneuve Blvd. West, Montreal, QC, H4A 3T2, Canada
| | - Vahé Nerguizian
- Department of Electrical Engineering, École de Technologie supérieure, 1100 Notre Dame West, Montreal, QC, H3C 1K3, Canada
| | - Julia V Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, 1001 Decarie Blvd, Montreal, QC, H4A 3J1, Canada.
- Department of Pathology, McGill University, Quebec, Canada.
| |
Collapse
|
20
|
Yang Y, Deng C, Aldali F, Huang Y, Luo H, Liu Y, Huang D, Cao X, Zhou Q, Xu J, Li Y, Chen H. Therapeutic Approaches and Potential Mechanisms of Small Extracellular Vesicles in Treating Vascular Dementia. Cells 2025; 14:409. [PMID: 40136659 PMCID: PMC11941715 DOI: 10.3390/cells14060409] [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: 01/12/2025] [Revised: 02/26/2025] [Accepted: 03/10/2025] [Indexed: 03/27/2025] Open
Abstract
Small extracellular vesicles (sEVs), including exosomes as a subtype, with a diameter typically less than 200 nm and originating from the endosomal system, are capable of transporting a diverse array of bioactive molecules, including proteins, nucleic acids, and lipids, thereby facilitating intercellular communication and modulating cellular functions. Vascular dementia (VaD) represents a form of cognitive impairment attributed to cerebrovascular disease, characterized by a complex and multifaceted pathophysiological mechanism. Currently, the therapeutic approach to VaD predominantly emphasizes symptom management, as no specific pharmacological treatment exists to cure the condition. Recent investigations have illuminated the significant role of sEVs in the pathogenesis of vascular dementia. This review seeks to provide a comprehensive analysis of the characteristics and functions of sEVs, with a particular focus on their involvement in vascular dementia and its underlying mechanisms. The objective is to advance the understanding of the interplays between sEVs and vascular dementia, thereby offering novel insights for future research and therapeutic strategies.
Collapse
Affiliation(s)
- Yujie Yang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Chunchu Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Fatima Aldali
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Yunjie Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Hongmei Luo
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Yizhou Liu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Danxia Huang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Xiaojian Cao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Qiuzhi Zhou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Jia Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Yajie Li
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
| | - Hong Chen
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (Y.Y.); (C.D.); (F.A.); (Y.H.); (H.L.); (Y.L.); (D.H.); (X.C.); (Q.Z.); (J.X.); (Y.L.)
- Stem Cell Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan 430030, China
| |
Collapse
|
21
|
Youssef E, Palmer D, Fletcher B, Vaughn R. Exosomes in Precision Oncology and Beyond: From Bench to Bedside in Diagnostics and Therapeutics. Cancers (Basel) 2025; 17:940. [PMID: 40149276 PMCID: PMC11940788 DOI: 10.3390/cancers17060940] [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: 01/13/2025] [Revised: 02/28/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Exosomes have emerged as pivotal players in precision oncology, offering innovative solutions to longstanding challenges such as metastasis, therapeutic resistance, and immune evasion. These nanoscale extracellular vesicles facilitate intercellular communication by transferring bioactive molecules that mirror the biological state of their parent cells, positioning them as transformative tools for cancer diagnostics and therapeutics. Recent advancements in exosome engineering, artificial intelligence (AI)-driven analytics, and isolation technologies are breaking barriers in scalability, reproducibility, and clinical application. Bioengineered exosomes are being leveraged for CRISPR-Cas9 delivery, while AI models are enhancing biomarker discovery and liquid biopsy accuracy. Despite these advancements, key obstacles such as heterogeneity in exosome populations and the lack of standardized isolation protocols persist. This review synthesizes pioneering research on exosome biology, molecular engineering, and clinical translation, emphasizing their dual roles as both mediators of tumor progression and tools for intervention. It also explores emerging areas, including microbiome-exosome interactions and the integration of machine learning in exosome-based precision medicine. By bridging innovation with translational strategies, this work charts a forward-looking path for integrating exosomes into next-generation cancer care, setting it apart as a comprehensive guide to overcoming clinical and technological hurdles in this rapidly evolving field.
Collapse
|
22
|
Karabay AZ, Barar J, Hekmatshoar Y, Rahbar Saadat Y. Multifaceted Therapeutic Potential of Plant-Derived Exosomes: Immunomodulation, Anticancer, Anti-Aging, Anti-Melanogenesis, Detoxification, and Drug Delivery. Biomolecules 2025; 15:394. [PMID: 40149930 PMCID: PMC11940522 DOI: 10.3390/biom15030394] [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: 02/06/2025] [Revised: 03/03/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Most eukaryotic and prokaryotic cells have the potential to secrete a group of structures/membrane-bound organelles, collectively referred to as extracellular vesicles (EVs), which offer several advantages to producer/receiver cells. This review provides an overview of EVs from plant sources with emphasis on their health-promoting potential and possible use as therapeutic agents. This review highlights the essential biological effects of plant-derived extracellular vesicles, including immune modulation, anticancer activities, protection against chemical toxicity and pathogens, as well as anti-aging, anti-melanogenesis, and anti-arthritic effects, along with ongoing clinical studies. Evidence revealed that plant-derived EVs' contents exert their beneficial properties through regulating important signaling pathways by transferring miRNAs and other components. Taken all together, the data proposed that plant-derived EVs can be utilized as nutritional compounds and therapeutic agents, such as drug carriers. However, this emerging research area requires further in vitro/in vivo studies and clinical trials to determine the exact underlying mechanisms of EVs' positive health effects in treating various diseases.
Collapse
Affiliation(s)
- Arzu Zeynep Karabay
- Department of Biochemistry, Faculty of Pharmacy, Ankara University, 06560 Ankara, Türkiye;
| | - Jaleh Barar
- Department of Pharmaceutical Sciences, College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA;
| | - Yalda Hekmatshoar
- Department of Medical Biology, Faculty of Medicine, Altinbas University, 34217 Istanbul, Türkiye;
| | - Yalda Rahbar Saadat
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran
| |
Collapse
|
23
|
Mo W, Peng Y, Zheng Y, Zhao S, Deng L, Fan X. Extracellular vesicle-mediated bidirectional communication between the liver and other organs: mechanistic exploration and prospects for clinical applications. J Nanobiotechnology 2025; 23:190. [PMID: 40055724 PMCID: PMC11889855 DOI: 10.1186/s12951-025-03259-4] [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: 11/26/2024] [Accepted: 02/20/2025] [Indexed: 03/17/2025] Open
Abstract
The liver, functioning as an endocrine organ, secretes a variety of substances that influence the activities of other body organs. Conversely, molecules generated by organs such as bone, the gut, and adipose tissue can also impact liver function. Accumulating evidence suggests bidirectional communication between the liver and other organs. However, research on how extracellular vesicles (EVs), which transport active molecular mediators, contribute to this interorgan communication is still in its nascent stages. EVs are capable of transporting functional molecules, including lipids, nucleic acids, and proteins, thereby affecting recipient cells across different organs at the biological level. This review examines the role of EVs in facilitating bidirectional communication between the liver and other organs such as bone, the cardiovascular system, the gut, the pancreas, the brain, the lungs, the kidneys, and adipose tissue. It explores their potential in disease treatment and highlights the challenges in understanding EV-mediated interorgan interactions. The contribution of mediator-carrying EVs to two-way communication between the liver and other organs remains an area of ongoing investigation. Future research will provide a more comprehensive theoretical foundation to clarify the precise mechanisms governing communication between the liver and other organs, pinpoint medical targets, and expand the application of EVs within the realm of precision medicine.
Collapse
Affiliation(s)
- Wenhui Mo
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yunke Peng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yanyi Zheng
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shenglan Zhao
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liling Deng
- Department of Endocrinology and Metabolism, Chongqing Emergency Medical Centre, Chongqing University Central Hospital, Chongqing, 400014, China.
| | - Xiaoli Fan
- Department of Gastroenterology and Hepatology and Laboratory of Gastrointestinal Cancer and Liver Disease, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
24
|
Song LL, Tang YP, Qu YQ, Yun YX, Zhang RL, Wang CR, Wong VKW, Wang HM, Liu MH, Qu LQ, Wu JH, Lo HH, Law BYK. Exosomal delivery of rapamycin modulates blood-brain barrier penetration and VEGF axis in glioblastoma. J Control Release 2025; 381:113605. [PMID: 40058500 DOI: 10.1016/j.jconrel.2025.113605] [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: 06/21/2024] [Revised: 01/04/2025] [Accepted: 03/03/2025] [Indexed: 03/17/2025]
Abstract
Exosomes (Exos), nanosized membranous vesicles (30-160 nm), have been validated as an effective drug delivery system capable of traversing biological barriers. Mesenchymal stem cells (MSCs), due to their near-limitless self-renewal capabilities, provide a plentiful source of exosomes for clinical applications. In this study, we utilized an exosome-encapsulated rapamycin (Exo-Rapa) delivery strategy, which permits the use of smaller drug dosages to achieve effects typically seen with higher dosages, thus enhancing drug efficacy. Moreover, Exos can transport pharmaceuticals across the blood-brain barrier (BBB) to the brain, and further penetrate GL261 cells to exert their effects. Within the tumor microenvironment, Exo-Rapa is released more rapidly and efficiently at the tumor site. The acidic conditions in tumors accelerate the release of Exo-Rapa, a characteristic that may make it a promising targeted therapeutic in future cancer research. Additionally, a series of in vivo experiments have further demonstrated the permeability of Exo-Rapa across the BBB, enabling it to accumulate at tumor sites; it also ameliorates inflammatory responses in Glioblastoma multiforme (GBM) mouse models and enhances anti-tumor activity through the regulation of angiogenesis via the VEGF/VEGFRs axis. Our results indicate that MSC-derived exosomes are a potent therapeutic carrier for GBM, offering an effective strategy for enhancing drug delivery across the BBB and providing a scientific foundation for the use of exosomes in the treatment of GBM and other diseases.
Collapse
Affiliation(s)
- Lin Lin Song
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yong Pei Tang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yuan Qing Qu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Yun Xiao Yun
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Rui Long Zhang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Cai Ren Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Vincent Kam Wai Wong
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China; Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, China
| | - Hui Miao Wang
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Meng Han Liu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Li Qun Qu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Jian Hui Wu
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Hang Hong Lo
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Betty Yuen Kwan Law
- Dr. Neher's Biophysics Laboratory for Innovative Drug Discovery, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China; Faculty of Chinese Medicine, Macau University of Science and Technology, Macau, China; Macau University of Science and Technology Zhuhai MUST Science and Technology Research Institute, China.
| |
Collapse
|
25
|
Li M, Wu L, Si H, Wu Y, Liu Y, Zeng Y, Shen B. Engineered mitochondria in diseases: mechanisms, strategies, and applications. Signal Transduct Target Ther 2025; 10:71. [PMID: 40025039 PMCID: PMC11873319 DOI: 10.1038/s41392-024-02081-y] [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: 06/24/2024] [Revised: 09/30/2024] [Accepted: 11/17/2024] [Indexed: 03/04/2025] Open
Abstract
Mitochondrial diseases represent one of the most prevalent and debilitating categories of hereditary disorders, characterized by significant genetic, biological, and clinical heterogeneity, which has driven the development of the field of engineered mitochondria. With the growing recognition of the pathogenic role of damaged mitochondria in aging, oxidative disorders, inflammatory diseases, and cancer, the application of engineered mitochondria has expanded to those non-hereditary contexts (sometimes referred to as mitochondria-related diseases). Due to their unique non-eukaryotic origins and endosymbiotic relationship, mitochondria are considered highly suitable for gene editing and intercellular transplantation, and remarkable progress has been achieved in two promising therapeutic strategies-mitochondrial gene editing and artificial mitochondrial transfer (collectively referred to as engineered mitochondria in this review) over the past two decades. Here, we provide a comprehensive review of the mechanisms and recent advancements in the development of engineered mitochondria for therapeutic applications, alongside a concise summary of potential clinical implications and supporting evidence from preclinical and clinical studies. Additionally, an emerging and potentially feasible approach involves ex vivo mitochondrial editing, followed by selection and transplantation, which holds the potential to overcome limitations such as reduced in vivo operability and the introduction of allogeneic mitochondrial heterogeneity, thereby broadening the applicability of engineered mitochondria.
Collapse
Affiliation(s)
- Mingyang Li
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Limin Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Haibo Si
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuangang Wu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuan Liu
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yi Zeng
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| | - Bin Shen
- Department of Orthopedics, Orthopedic Research Institute, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
26
|
Athira AP, Sreekanth S, Chandran A, Lahon A. Dual Role of Extracellular Vesicles as Orchestrators of Emerging and Reemerging Virus Infections. Cell Biochem Biophys 2025; 83:159-175. [PMID: 39225901 DOI: 10.1007/s12013-024-01495-3] [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] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Current decade witnessed the emergence and re-emergence of many viruses, which affected public health significantly. Viruses mainly utilize host cell machinery to promote its growth, and spread of these diseases. Numerous factors influence virus-host cell interactions, of which extracellular vesicles play an important role, where they transfer information both locally and distally by enclosing viral and host-derived proteins and RNAs as their cargo. Thus, they play a dual role in mediating virus infections by promoting virus dissemination and evoking immune responses in host organisms. Moreover, it acts as a double-edged sword during these infections. Advances in extracellular vesicles regulating emerging and reemerging virus infections, particularly in the context of SARS-CoV-2, Dengue, Ebola, Zika, Chikungunya, West Nile, and Japanese Encephalitis viruses are discussed in this review.
Collapse
Affiliation(s)
- A P Athira
- Department of Viral Vaccines, Institute of Advanced Virology, Bio 360 Life Science Park, Thiruvananthapuram, Kerala, India
| | - Smrithi Sreekanth
- Department of Viral Vaccines, Institute of Advanced Virology, Bio 360 Life Science Park, Thiruvananthapuram, Kerala, India
| | - Ananthu Chandran
- Department of Viral Vaccines, Institute of Advanced Virology, Bio 360 Life Science Park, Thiruvananthapuram, Kerala, India
| | - Anismrita Lahon
- Department of Viral Vaccines, Institute of Advanced Virology, Bio 360 Life Science Park, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
27
|
Sangubotla R, Gubbiyappa KS, Devarapogu R, Kim J. Modern insights of nanotheranostics in the glioblastoma: An updated review. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167653. [PMID: 39756713 DOI: 10.1016/j.bbadis.2024.167653] [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: 08/29/2024] [Revised: 12/08/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025]
Abstract
Glioblastoma multiforme (GBM) is a highly malignant subtype of glioma, originating from the glial cells that provide support to other neurons in the brain. GBM predominantly impacts the cerebral hemisphere of the brain, with minimal effects on the cerebellum, brain stem, or spinal cord. Individuals diagnosed with GBM commonly encounter a range of symptoms, starting from auditory abnormalities to seizures. Recently, cell membrane-camouflaged nanoparticles (CMCNPs) are evolving as promising theranostic agents that can carry specific biological moieties from their biological origin and effectively target GBM cells. Moreover, exosomes have gained widespread scientific attention as an effective drug delivery approach due to their excellent stability in the bloodstream, high biocompatibility, low immune response, and inherent targeting capabilities. Exosomes derived from specific cell types can transport endogenous signaling molecules that have therapeutic promise for GBM therapy. In this context, researchers are utilizing various techniques to isolate exosomes from liquid biomarkers from patients, such as serum and cerebrospinal fluid (CSF). Proper isolation of exosomes may induce the clinical diagnosis in GBM due to their commercial accessibility and real-time monitoring options. Since exosomes are unable to penetrate the blood-brain barrier (BBB), strategic theranostic methods are ideal. For this, understanding interactions between glioma-specific exosomes in the TME and biomarkers is necessary. The versatile characteristics of NPs and their capacity to cross the BBB enable them to be indispensable against GBM. In this review article, we discussed the recent theranostic applications of nanotechnology by comparing the limitations of existing nanotechnology-based approaches.
Collapse
Affiliation(s)
- Roopkumar Sangubotla
- Department of Chemical and Biological Engineering, Gachon University, 1342 Seongnam Daero, Seongnam-Si, Gyeonggi-Do 13120, Republic of Korea
| | - Kumar Shiva Gubbiyappa
- GITAM School of Pharmacy, GITAM Deemed to be University, Rudraram, Patencheru, Sangareddy Dist, 502329, Telangana, India
| | - Rajakumari Devarapogu
- Department of Zoology, Sri Venkateswara University, Tirupati, Andhra Pradesh 517502, India
| | - Jongsung Kim
- Department of Chemical and Biological Engineering, Gachon University, 1342 Seongnam Daero, Seongnam-Si, Gyeonggi-Do 13120, Republic of Korea.
| |
Collapse
|
28
|
Kostyusheva A, Romano E, Yan N, Lopus M, Zamyatnin AA, Parodi A. Breaking barriers in targeted Therapy: Advancing exosome Isolation, Engineering, and imaging. Adv Drug Deliv Rev 2025; 218:115522. [PMID: 39855273 DOI: 10.1016/j.addr.2025.115522] [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: 07/30/2024] [Revised: 12/23/2024] [Accepted: 01/19/2025] [Indexed: 01/27/2025]
Abstract
Exosomes have emerged as promising tools for targeted drug delivery in biomedical applications and medicine. This review delves into the scientific advancements, challenges, and future prospects specifically associated with these technologies. In this work, we trace the research milestones that led to the discovery and characterization of exosomes and extracellular vesicles, and discuss strategies for optimizing the synthetic yield and the loading of these particles with various therapeutics. In addition, we report the current major issues affecting the field and hampering the clinical translation of these technologies. Highlighting the pivotal role of imaging techniques, we explore how they drive exosome therapy and development by offering insights into biodistribution and cellular trafficking dynamics. Methodologies for vesicle isolation, characterization, loading, and delivery mechanisms are thoroughly examined, alongside strategies aimed at enhancing their therapeutic efficacy. Special emphasis was dedicated to their therapeutic properties, particularly to their ability to deliver biologics into the cytoplasm. Furthermore, we delve into the intricate balance between surface modifications and targeting properties including also transgenic methods aimed at their functionalization and visualization within biological systems. This review underscores the transformative potential of these carriers in targeted drug delivery and identifies crucial areas for further research and clinical translation.
Collapse
Affiliation(s)
- Anastasiya Kostyusheva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119048 Moscow, Russia
| | | | - Neng Yan
- School of Environmental Studies, China University of Geosciences, Wuhan 430074, China
| | - Manu Lopus
- School of Biological Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai Kalina Campus, Vidyanagari, Mumbai 400098, India
| | - Andrey A Zamyatnin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; Department of Biological Chemistry, Sechenov First Moscow State Medical University, Trubetskaya Str. 8-2, 119991 Moscow, Russia
| | - Alessandro Parodi
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia; Scientific Center for Translational Medicine, Sirius University of Science and Technology, 354340, Sirius, Krasnodar Region, Russia.
| |
Collapse
|
29
|
Ivanova A, Chalupska R, Louro AF, Firth M, González-King Garibotti H, Hultin L, Kohl F, Lázaro-Ibáñez E, Lindgren J, Musa G, Oude Blenke E, Silva AM, Szeponik L, Taylor A, Viken I, Wang X, Jennbacken K, Wiseman J, Dekker N. Barcoded Hybrids of Extracellular Vesicles and Lipid Nanoparticles for Multiplexed Analysis of Tissue Distribution. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2407850. [PMID: 39823165 PMCID: PMC11904941 DOI: 10.1002/advs.202407850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/18/2024] [Indexed: 01/19/2025]
Abstract
Targeted delivery of therapeutic agents is a persistent challenge in modern medicine. Recent efforts in this area have highlighted the utility of extracellular vesicles (EVs) as drug carriers, given that they naturally occur in bloodstream and tissues, and can be loaded with a wide range of therapeutic molecules. However, biodistribution and tissue tropism of EVs remain difficult to study systematically. Here, a multiplexed approach is developed for simultaneous tracking of EVs from various cell lines within a single in vivo experiment. EVs are used from 16 different cell lines, and through controlled fusion with lipid nanoparticles (LNPs) carrying single-stranded DNA barcodes, uniquely barcoded hybrid EV particle (hEV) library is generated. These hEVs are combined for a multiplexed in vivo biodistribution profiling in mice, and discovered that HAP1-derived hEVs demonstrated lung tropism, suggesting that these hEVs may be used for targeted drug delivery into lung tissue. To examine this possibility further, it is shown that HAP1 hEV loaded with Cre mRNA displayed functional delivery to the lungs. Overall, the barcoded hEV technology enables rapid profiling of biodistribution across EV cell sources, which is poised to improve throughput and extent of EV studies, while reducing the number of animals required for research.
Collapse
Affiliation(s)
- Alena Ivanova
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Renata Chalupska
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Ana Filipa Louro
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Mike Firth
- Data Sciences and Quantitative Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, CB2 0AA, UK
| | - Hernán González-King Garibotti
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Leif Hultin
- Clinical Pharmacology and Safety Science, Imaging and Data Analytics BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Franziska Kohl
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solnavägen 1, Solna, Stockholm, 171 77, Sweden
| | - Elisa Lázaro-Ibáñez
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Julia Lindgren
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Gentian Musa
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Erik Oude Blenke
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Andreia M Silva
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Louis Szeponik
- Clinical Pharmacology and Safety Science, Imaging and Data Analytics BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Agnes Taylor
- Advanced Drug Delivery, Pharmaceutical Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Ida Viken
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Xiaoqin Wang
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Karin Jennbacken
- Bioscience Cardiovascular, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - John Wiseman
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| | - Niek Dekker
- Discovery Biology, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, Mölndal, 43150, Sweden
| |
Collapse
|
30
|
Wang M, Liu Z, Wang W, Chopp M, Millman M, Li Y, Cepparulo P, Kemper A, Li C, Zhang L, Zhang Y, Zhang ZG. Enhanced Small Extracellular Vesicle Uptake by Activated Interneurons Improves Stroke Recovery in Mice. JOURNAL OF EXTRACELLULAR BIOLOGY 2025; 4:e70036. [PMID: 40134760 PMCID: PMC11934211 DOI: 10.1002/jex2.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/07/2025] [Accepted: 01/24/2025] [Indexed: 03/27/2025]
Abstract
Neuronal circuitry remodelling, which comprises excitatory and inhibitory neurons, is critical for improving neurological outcomes after a stroke. Preclinical studies have shown that small extracellular vesicles (sEVs) have a therapeutic effect on stroke recovery. However, it is highly challenging to use sEVs to specifically target individual neuronal populations to enhance neuronal circuitry remodelling after stroke. In the present study, using a chemogenetic approach to specifically activate peri-infarct cortical interneurons in combination with the administration of sEVs derived from cerebral endothelial cells (CEC-sEVs), we showed that the CEC-sEVs were preferentially taken up by the activated neurons, leading to significant improvement of functional outcome after stroke, which was associated with augmentation of peri-infarct cortical axonal/dendritic outgrowth and of axonal remodelling of the corticospinal tract. The ultrastructural and Western blot analyses revealed that neurons with internalization of CEC-sEVs exhibited significantly reduced numbers of damaged mitochondria and proteins that mediate dysfunctional mitochondria, respectively. Together, these data indicate that the augmented uptake of CEC-sEVs by activated peri-infarct cortical interneurons facilitates neuronal circuitry remodelling and functional recovery after stroke, which has the potential to be a novel therapy for improving stroke recovery.
Collapse
Affiliation(s)
- Mingjin Wang
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Zhongwu Liu
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Weida Wang
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Michael Chopp
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
- Department of PhysicsOakland UniversityRochesterMichiganUSA
| | - Michael Millman
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Yanfeng Li
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | | | - Amy Kemper
- Department of PathologyHenry Ford HospitalDetroitMichiganUSA
| | - Chao Li
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Li Zhang
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | - Yi Zhang
- Department of NeurologyHenry Ford HospitalDetroitMichiganUSA
| | | |
Collapse
|
31
|
Qian L, Chen P, Zhang S, Wang Z, Guo Y, Koutouratsas V, Fleishman JS, Huang C, Zhang S. The uptake of extracellular vesicles: Research progress in cancer drug resistance and beyond. Drug Resist Updat 2025; 79:101209. [PMID: 39893749 DOI: 10.1016/j.drup.2025.101209] [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: 12/11/2024] [Revised: 01/22/2025] [Accepted: 01/26/2025] [Indexed: 02/04/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous vesicles released by donor cells that can be taken up by recipient cells, thus inducing cellular phenotype changes. Since their discovery decades ago, roles of EVs in modulating initiation, growth, survival and metastasis of cancer have been revealed. Recent studies from multifaceted perspectives have further detailed the contribution of EVs to cancer drug resistance; however, the role of EV uptake in conferring drug resistance seems to be overlooked. In this comprehensive review, we update the EV subtypes and approaches for determining EV uptake. The biological basis of EV uptake is systematically summarized. Moreover, we focus on the diverse uptake mechanisms by which EVs carry out the intracellular delivery of functional molecules and drug resistance signaling. Furthermore, we highlight how EV uptake confers drug resistance and identify potential strategies for targeting EV uptake to overcome drug resistance. Finally, we discuss the research gap on the role of EV uptake in promoting drug resistance. This updated knowledge provides a new avenue to overcome cancer drug resistance by targeting EV uptake.
Collapse
Affiliation(s)
- Luomeng Qian
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Pangzhou Chen
- Department of Breast Surgery, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan 528200, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Nankai University, Tianjin 300121, China
| | - Zhenglu Wang
- Department of Pathology, Tianjin Key Laboratory for Organ Transplantation, Tianjin First Centre Hospital, Tianjin 300192, China
| | - Yuan Guo
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Vasili Koutouratsas
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA
| | - Chuanqiang Huang
- Department of Breast Surgery, Sixth Affiliated Hospital, School of Medicine, South China University of Technology, Foshan 528200, China
| | - Sihe Zhang
- Department of Cell Biology, School of Medicine, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
32
|
Mahboob A, Fatma N, Faraz A, Pervez M, Khan MA, Husain A. Advancements in the conservation of the conformational epitope of membrane protein immunogens. Front Immunol 2025; 16:1538871. [PMID: 40093005 PMCID: PMC11906443 DOI: 10.3389/fimmu.2025.1538871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/03/2025] [Indexed: 03/19/2025] Open
Abstract
Generating antibodies targeting native membrane proteins presents various challenges because these proteins are often embedded in the lipid bilayer, possess various extracellular and intracellular domains, and undergo post-translational modifications. These properties of MPs make it challenging to preserve their stable native conformations for immunization or antibody generation outside of the membranes. In addition, MPs are often hydrophobic due to their membrane-spanning regions, making them difficult to solubilize and purify in their native form. Therefore, employing purified MPs for immunogen preparation may result in denaturation or the loss of native structure, rendering them inadequate for producing antibodies recognizing native conformations. Despite these obstacles, various new approaches have emerged to address these problems. We outline recent advancements in designing and preparing immunogens to produce antibodies targeting MPs. Strategies outlined here are relevant for producing antibodies for research, diagnostics, and therapies and designing immunogens for vaccination purposes.
Collapse
Affiliation(s)
- Aisha Mahboob
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Nishat Fatma
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Ahmed Faraz
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Muntaha Pervez
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Mohammad Afeef Khan
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| | - Afzal Husain
- Department of Biochemistry, Faculty of Life Sciences, Aligarh Muslim University, Aligarh, India
| |
Collapse
|
33
|
Chen Y, Douanne N, Wu T, Kaur I, Tsering T, Erzingatzian A, Nadeau A, Juncker D, Nerguizian V, Burnier JV. Leveraging nature's nanocarriers: Translating insights from extracellular vesicles to biomimetic synthetic vesicles for biomedical applications. SCIENCE ADVANCES 2025; 11:eads5249. [PMID: 40009680 PMCID: PMC11864201 DOI: 10.1126/sciadv.ads5249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 01/24/2025] [Indexed: 02/28/2025]
Abstract
Naturally occurring extracellular vesicles (EVs) and synthetic nanoparticles like liposomes have revolutionized precision diagnostics and medicine. EVs excel in biocompatibility and cell targeting, while liposomes offer enhanced drug loading capacity and scalability. The clinical translation of EVs is hindered by challenges including low yield and heterogeneity, whereas liposomes face rapid immune clearance and limited targeting efficiency. To bridge these gaps, biomimetic synthetic vesicles (SVs) have emerged as innovative platforms, combining the advantageous properties of EVs and liposomes. This review emphasizes critical aspects of EV biology, such as mechanisms of EV-cell interaction and source-dependent functionalities in targeting, immune modulation, and tissue regeneration, informing biomimetic SV engineering. We reviewed a broad array of biomimetic SVs, with a focus on lipid bilayered vesicles functionalized with proteins. These include cell-derived nanovesicles, protein-functionalized liposomes, and hybrid vesicles. By addressing current challenges and highlighting opportunities, this review aims to advance biomimetic SVs for transformative biomedical applications.
Collapse
Affiliation(s)
- Yunxi Chen
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Noélie Douanne
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
- Department of Biomedical Engineering and Victor Philippe Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | - Tad Wu
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Ishman Kaur
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- École de technologie supérieure ÉTS, Montreal, QC, Canada
| | - Thupten Tsering
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - Armen Erzingatzian
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Amélie Nadeau
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
| | - David Juncker
- Department of Biomedical Engineering and Victor Philippe Dahdaleh Institute of Genomic Medicine, McGill University, Montreal, QC, Canada
| | | | - Julia V. Burnier
- Cancer Research Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
- Department of Pathology, McGill University, Montreal, QC, Canada
- Gerald Bronfman Department of Oncology, McGill University, Montreal, QC, Canada
| |
Collapse
|
34
|
Kang S, Jeon S, Baek H, Hwang S, Kim S, Youn SH, Kim JW, Jun SH, Kang NG. Lactobacillus-derived artificial extracellular vesicles for skin rejuvenation and prevention of photo-aging. Biomater Sci 2025. [PMID: 40013489 DOI: 10.1039/d4bm01644k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Extracellular vesicles (EVs) are small membrane-bound sacs released by cells that play crucial roles in intercellular communication. They transport biomolecules between cells and have both diagnostic and therapeutic potential. Artificial EVs, designed to mimic natural EVs, have been developed using various methods. In this study, Lactobacillus plantarum was used to create Lactobacillus-derived artificial EVs (LAEs) for skin rejuvenation and anti-aging. LAEs demonstrated monodispersity and effectively improved adverse gene expression and wound healing in fibroblasts. They also modulated aging-related genes and improved skin conditions in humans. Their simplicity, promptness, and lack of animal-derived sources make LAEs a promising alternative to natural EVs. LAEs have the potential to overcome the technical limitations of artificial EVs and advance EVs or exosome-based technologies for comprehensive skin rejuvenation.
Collapse
Affiliation(s)
- Seongsu Kang
- LG Household and Health Care R&D Center, Seoul 07795, Republic of Korea.
| | - Saetbyeol Jeon
- School of Chemical Engineering, Sungkyunkwan University, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Hwira Baek
- School of Chemical Engineering, Sungkyunkwan University, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Sunghwan Hwang
- LG Household and Health Care R&D Center, Seoul 07795, Republic of Korea.
| | - Seulgi Kim
- School of Chemical Engineering, Sungkyunkwan University, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Sung Hun Youn
- LG Household and Health Care R&D Center, Seoul 07795, Republic of Korea.
| | - Jin Woong Kim
- School of Chemical Engineering, Sungkyunkwan University, Seobu-ro, Jangan-gu, Suwon, Gyeonggi-do 16419, Republic of Korea.
| | - Seung-Hyun Jun
- LG Household and Health Care R&D Center, Seoul 07795, Republic of Korea.
| | - Nae-Gyu Kang
- LG Household and Health Care R&D Center, Seoul 07795, Republic of Korea.
| |
Collapse
|
35
|
Attar FA, Irani S, Oloomi M, Bolhassani A, Geranpayeh L, Atyabi F. Doxorubicin loaded exosomes inhibit cancer-associated fibroblasts growth: in vitro and in vivo study. Cancer Cell Int 2025; 25:72. [PMID: 40016747 PMCID: PMC11869484 DOI: 10.1186/s12935-025-03689-y] [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: 09/22/2024] [Accepted: 02/12/2025] [Indexed: 03/01/2025] Open
Abstract
Cancer-associated fibroblast cells (CAFs) play a key role in the breast cancer (BC) microenvironment that induces resistance to chemotherapy. Adipose mesenchymal stem cells (ADMSCs) derived exosomes were utilized to deliver the doxorubicin (Dox) to BC cell lines (MDA-MB-231, MCF-7) and CAFs in both mono and co-culture systems. Immunocytochemistry (ICC) for VIMENTIN and flow cytometry for the CD45, CD34, CD73, and CD90 markers were used to confirm the phenotypic characteristics of CAFs and MSC cells. Dox was loaded into ADMSCs-derived exosomes (Exo-Dox) through sonication and its loading wasa confirmed by transmission electron microscope (TEM). Compared to free Dox, Exo-Dox showed a higher efficiency in inducing apoptosis and inhibiting growth and migration in co-culture cells with CAFs (P < 0.05). The up-regulation of H19 and UCA1 lncRNAs, associated with chemoresistance, was confirmed using real-time PCR in CAF-derived breast cancer patients, CAF-derived exosomes, and exosome-derived patient serums. H19 and UCA1 expression levels were significantly down-regulated in MDA-MB-231, MCF-7, and co-cultures of MDA-MB-231 and MCF-7 cells with CAFs that received Exo-Dox treatment. In vivo results indicated that ADMSCs-derived exosomes (MSC-Exos) can accumulate at the tumor site. Exo-Dox suppressed cancer cell growth and significantly decreased tumor size compared to PBS (p < 0.01). The findings confirmed the growth inhibition effects of Exo-Dox n in CAFs, BC cells, and tumor-bearing mice.
Collapse
Affiliation(s)
- Fatemeh Akhavan Attar
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shiva Irani
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Mana Oloomi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Azam Bolhassani
- Department of Hepatitis and AIDS, Pasteur Institute of Iran, Tehran, Iran
| | | | - Fatemeh Atyabi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Zhang G, Huang X, Liu S, Xu Y, Wang N, Yang C, Zhu Z. Demystifying EV heterogeneity: emerging microfluidic technologies for isolation and multiplexed profiling of extracellular vesicles. LAB ON A CHIP 2025; 25:1228-1255. [PMID: 39775292 DOI: 10.1039/d4lc00777h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Extracellular vesicles (EVs) are heterogeneous lipid containers carrying complex molecular cargoes, including proteins, nucleic acids, glycans, etc. These vesicles are closely associated with specific physiological characteristics, which makes them invaluable in the detection and monitoring of various diseases. However, traditional isolation methods are often labour-intensive, inefficient, and time-consuming. In addition, single biomarker analyses are no longer accurate enough to meet diagnostic needs. Routine isolation and molecular analysis of high-purity EVs in clinical applications is even more challenging. In this review, we discuss a promising solution, microfluidic-based techniques, that combine efficient isolation and multiplex detection of EVs, to further demystify EV heterogeneity. These microfluidic-based EV multiplexing platforms will hopefully facilitate development of liquid biopsies and offer promising opportunities for personalised therapy.
Collapse
Affiliation(s)
- Guihua Zhang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Xiaodan Huang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Sinong Liu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Yiling Xu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Nan Wang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, School of Medicine, Shanghai Jiao tong University, Shanghai 200127, China
| | - Zhi Zhu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China.
| |
Collapse
|
37
|
Pikwong F, Kamsarn J, Jarisarapurin W, Baipaywad P, Park H, Kumphune S. Cardiac Cell Membrane-Coated Nanoparticles as a Potential Targeted Delivery System for Cardiac Therapy. Biomimetics (Basel) 2025; 10:141. [PMID: 40136795 PMCID: PMC11940174 DOI: 10.3390/biomimetics10030141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/05/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Cardiomyopathies, a cause of heart failure, are a predominant cause of death globally and may lead to discernible myocardial abnormalities. Several therapeutic agents were discovered, developed, investigated, and evaluated to save patients' lives and improve their quality of life. The effective administration of drugs improves therapeutic outcomes while reducing side effects. Nanoparticles (NPs) have been utilised for the delivery of therapeutic agents and demonstrate promise in reducing myocardial ischaemia/reperfusion injury. However, significant limitations of NPs include non-specific targeting and immunogenicity. To improve cardiac targeting and biocompatibility, surface modifications using a cardiac cell membrane (cCM) coating on the surface of NPs have been hypothesised. Here, cCMs were isolated from the human ventricular cell line (AC16), and mesoporous silica nanoparticles (MSNs) were synthesised and then coated with cCMs. The cardiac cell membrane-coated mesoporous silica nanoparticles (cCMCMSNs) did not significantly alter the encapsulation efficiency or the release profile of the loaded drug (Rhodamine B) in comparison to MSN. Moreover, cCMCMSNs demonstrated a significantly enhanced distribution of RhB specifically to cardiac cells, compared to other cell types, without causing cytotoxicity. To evaluate immune escape, cCMCMSNs were exposed to activated macrophages, demonstrating that cCMCMSNs were phagocytosed to a lesser extent than MSN. This study demonstrated the synthesis of cardiac cell membranes coated on the surface of nanoparticles as nanomedicine technologies that enhance selective drug delivery to cardiac cells, potentially offering an alternate method for drug administration in cardiovascular diseases.
Collapse
Affiliation(s)
- Faprathan Pikwong
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (F.P.); (J.K.); (W.J.); (P.B.)
- Biomedical Engineering and Innovation Research Centre, Chiang Mai University, Mueang Chiang Mai District, Chiang Mai 50200, Thailand
| | - Jiraporn Kamsarn
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (F.P.); (J.K.); (W.J.); (P.B.)
- Biomedical Engineering and Innovation Research Centre, Chiang Mai University, Mueang Chiang Mai District, Chiang Mai 50200, Thailand
| | - Wattanased Jarisarapurin
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (F.P.); (J.K.); (W.J.); (P.B.)
- Biomedical Engineering and Innovation Research Centre, Chiang Mai University, Mueang Chiang Mai District, Chiang Mai 50200, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Phornsawat Baipaywad
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (F.P.); (J.K.); (W.J.); (P.B.)
- Biomedical Engineering and Innovation Research Centre, Chiang Mai University, Mueang Chiang Mai District, Chiang Mai 50200, Thailand
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul 06974, Republic of Korea;
| | - Sarawut Kumphune
- Biomedical Engineering Institute, Chiang Mai University, Chiang Mai 50200, Thailand; (F.P.); (J.K.); (W.J.); (P.B.)
- Biomedical Engineering and Innovation Research Centre, Chiang Mai University, Mueang Chiang Mai District, Chiang Mai 50200, Thailand
| |
Collapse
|
38
|
Chen H, Han Z, Wang S, Zhu M, Wang L, Lin Y, Wang X, Zhang Y, Wang W, Li M, Liu X, Mann S, Huang X. Droplet-supported liquid-liquid lateral phase separation as a step to floating protein heterostructures. Nat Commun 2025; 16:1897. [PMID: 39988593 PMCID: PMC11847946 DOI: 10.1038/s41467-025-57141-w] [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: 10/05/2024] [Accepted: 02/12/2025] [Indexed: 02/25/2025] Open
Abstract
Liquid-liquid phase separation plays an important role in many natural and technological processes. Herein, we implement lateral microphase separation at the surface of oil micro-droplets suspended in water to prepare a range of discrete floating protein/polymer continuous two-dimensional (2D) heterostructures with variable interfacial domain structures and dynamics. We show that gel-like domains of bovine serum albumin (BSA) co-exist with fluid-like polyvinyl alcohol (PVA) regions at the oil droplet surface to produce floating heterostructures comprising a 2D phase-separated protein mesh or an array of discrete mobile protein rafts depending on the conditions employed. Enzymes are embedded in the discontinuous BSA domains to produce droplet-supported microphase-separated 2D reaction scaffolds that can be tuned for interfacial catalysis. Taken together, our work has general implications for the structural and functional augmentation of oil droplet interfaces and contributes to the surface engineering and functionality of droplet-based micro-reactors.
Collapse
Affiliation(s)
- Haixu Chen
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
- State Key Laboratory of Urban Water Resource and Environments, Harbin Institute of Technology, Harbin, China
| | - Zhengbin Han
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin, China
| | - Shengliang Wang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Mei Zhu
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Lei Wang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Youping Lin
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Xiaoliang Wang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Yide Zhang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Wei Wang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
- State Key Laboratory of Urban Water Resource and Environments, Harbin Institute of Technology, Harbin, China
| | - Mei Li
- Max Planck Bristol Centre for Minimal Biology, Centre for Protolife Research and Centre for Organized Matter Chemistry, School of Chemistry, University of Bristol, Bristol, UK
| | - Xiaoman Liu
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China
| | - Stephen Mann
- Max Planck Bristol Centre for Minimal Biology, Centre for Protolife Research and Centre for Organized Matter Chemistry, School of Chemistry, University of Bristol, Bristol, UK.
| | - Xin Huang
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, China.
| |
Collapse
|
39
|
Gong X, Liu S, Xia B, Wan Y, Zhang S, Zhang B, Wang Z, Chen J, Xiao F, Liang XJ, Yang Y. Oral delivery of therapeutic proteins by engineered bacterial type zero secretion system. Nat Commun 2025; 16:1862. [PMID: 39984501 PMCID: PMC11845744 DOI: 10.1038/s41467-025-57153-6] [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: 05/21/2024] [Accepted: 02/11/2025] [Indexed: 02/23/2025] Open
Abstract
Genetically engineered commensal bacteria are promising living drugs, however, their therapeutic molecules are frequently confined to their colonization sites. Herein, we report an oral protein delivery technology utilizing an engineered bacterial type zero secretion system (T0SS) via outer membrane vesicles (OMVs). We find that OMVs produced in situ by Escherichia coli Nissle 1917 (EcN) can penetrate the intact gut epithelial barrier to enter the circulation and that epithelial transcytosis involves pinocytosis and dynamin-dependent pathways. EcN is engineered to endogenously load various enzymes into OMVs, and the secreted enzyme-loaded OMVs are able to stably catalyze diverse detoxification reactions against digestive fluid and even enter the circulation. Using hyperuricemic mice and uricase delivery as a demonstration, we demonstrate that the therapeutic efficacy of our engineered EcN with a modified T0SS outperforms that with a direct protein secretion apparatus. The enzyme-loaded OMVs also effectively detoxify human serum samples, highlighting the potential for the clinical treatment of metabolic disorders.
Collapse
Affiliation(s)
- Xu Gong
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Shan Liu
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Bozhang Xia
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China
| | - Yichen Wan
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Shuyi Zhang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, P. R. China
- Center for Synthetic and Systems Biology, School of Life Sciences, Tsinghua University, Beijing, P. R. China
| | - Baoyan Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Zehao Wang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Junge Chen
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China
| | - Fei Xiao
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, P. R. China.
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, Beijing, P. R. China.
| | - Yun Yang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Medical Science and Engineering, Beihang University, Beijing, P. R. China.
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, P. R. China.
| |
Collapse
|
40
|
Zhao ZH, Huang B, Fang ZH, Qi WH, He J, Wang LB, Zhang Y, Li X. Engineered extracellular vesicles for TGF-β encapsulation as a therapeutic strategy against LPS-induced systemic inflammation. Int Immunopharmacol 2025; 148:114109. [PMID: 39862634 DOI: 10.1016/j.intimp.2025.114109] [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: 03/01/2024] [Revised: 12/29/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
Inflammation underlies a wide variety of physiological and pathological processes, the Lipopolysaccharide (LPS)-induced inflammation model is widely recognized as a classical inflammatory paradigm, while Transforming growth factor-β (TGF-β) serves as a potent immunosuppressant capable of inhibiting immune responses and mitigating inflammation. However, its in vivo instability and the high cost associated with purification have imposed limitations on its clinical application. Therefore, we propose a therapeutic strategy for genetically modifying extracellular vesicles (HEVs) derived from HEK-293 T cells to incorporate TGF-β which holds potential for mitigating LPS-induced inflammation. In this study, we engineered a TGF-β lentivirus that specific incorporates TGF-β into HEVs and efficiently produces highly expressed TGF-β HEVs (HEVTs) through infection of HEK293 cells. Our data demonstrated that, compared to the LPS group, HEVTs internalized by immune cells significantly regulated pro-inflammatory cytokine expression in RAW 264.7 cells, such as IL-1β (p < 01), TNF-α (p < 001). Moreover, HEVTs were found to effectively reach the lesion area, compared to the LPS group, resulting in a remarkable inhibition in the activation of macrophages (p < 0.0001), dendritic cells (p < 0.0001), and neutrophils (p < 0.0001) in the peripheral immune system as well as microglia in the central nervous system of LPS-induced inflammation model mice. The utilization of this endogenous loading technique may present a promising strategy for the protein-based pharmacotherapy of inflammatory disorders.
Collapse
Affiliation(s)
- Zhuo-Hua Zhao
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Bo Huang
- The Laboratory Animal Center, Shanxi Provincial People's Hospital, 030012, China
| | - Zi-Han Fang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Wen-Hui Qi
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jin He
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China
| | - Li-Bin Wang
- Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen Nanshan Hospital, Shenzhen, Guangdong 518052, China
| | - Yuan Zhang
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| | - Xing Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry (Shaanxi Normal University), The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi 710119, China.
| |
Collapse
|
41
|
Rolland TJ, Hudson ER, Graser LA, Zahra S, Cucinotta D, Sonkawade SD, Sharma UC, Weil BR. Mitochondrial DNA-Mediated Immune Activation After Resuscitation from Cardiac Arrest. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.02.14.25322318. [PMID: 40034769 PMCID: PMC11875248 DOI: 10.1101/2025.02.14.25322318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Background Post-cardiac arrest syndrome (PCAS) is characterized by a robust inflammatory response that contributes to significant morbidity and mortality among patients resuscitated from sudden cardiac arrest (SCA). Mitochondrial DNA (mtDNA), with its bacterial-like genomic motifs, has been implicated as a damage-associated molecular pattern in other inflammatory contexts, but its role as a pro-inflammatory stimulus in PCAS has not been studied. Accordingly, the present study was designed to determine if PCAS is characterized by a rise in circulating mtDNA and, if so, whether mtDNA is selectively released, how it activates immune cells, and if targeting mtDNA-sensing pathways attenuates leukocyte activation. Methods Plasma mtDNA and nuclear DNA (nucDNA) levels were measured in peripheral blood samples collected ∼4-hours post-ROSC from swine with PCAS (n=8) and patients hospitalized after resuscitation from out-of-hospital cardiac arrest (OHCA; n= 57). Additionally, in vitro studies were performed where porcine peripheral blood mononuclear cells (PBMCs) were treated with mtDNA or extracellular vesicles (EVs) isolated from post-ROSC plasma. Pharmacological inhibitors were utilized to inhibit toll-like receptor 9 (TLR9)- and cyclic GMP-AMP synthase (cGAS)-mediated mtDNA sensing. Results A significant ∼250-fold elevation in circulating mtDNA was observed shortly after ROSC in swine despite negligible changes in circulating nucDNA, suggesting selective release of mtDNA in PCAS. This finding was corroborated in human OHCA survivors, in which circulating mtDNA was similarly elevated during the early post-ROSC period. Circulating mtDNA was largely encapsulated within EVs in swine and humans, suggesting a conserved mechanism of release across species. In vitro studies demonstrated that PBMC internalization of mtDNA-containing-EVs was required for immune activation and promoted development of a pro-inflammatory leukocyte phenotype characterized by altered surface marker expression and increased release of TNFα, IL-1β, and IL-6. Disrupting EVs or degrading enclosed DNA attenuated these responses, which were partially restored upon reintroduction of mtDNA. Pharmacological blockade of TLR9 or cGAS pathways significantly reduced mtDNA-induced inflammation, providing insight regarding signaling pathways that may be targeted to modulate mtDNA-mediated immune activation in PCAS. Conclusions These novel findings demonstrate that brief whole-body ischemia and reperfusion in the context of resuscitation from SCA triggers selective mtDNA release, primarily within EVs, that acts as a potent driver of immune activation in PCAS. By linking EV-encapsulated mtDNA to TLR9 and cGAS activation, this study provides a foundation for the development of novel therapeutic interventions aimed at limiting mtDNA release or disrupting its downstream sensing pathways to enhance survival and improve outcomes after SCA. Clinical Perspective What is new?: Our study reveals that circulating mitochondrial DNA (mtDNA), primarily encapsulated in extracellular vesicles (EV), is selectively released into the bloodstream after resuscitation from sudden cardiac arrest.EV-encapsulated mtDNA triggers immune cell activation, evidenced by phenotypic shifts toward inflammatory dendritic cells and macrophages, as well as increased pro-inflammatory cytokine secretion.Pharmacological inhibition of TLR9 and cGAS pathways significantly attenuates the mtDNA-induced inflammatory response, pointing to novel therapeutic avenues for modulating post-resuscitation immune activation in patients with post-cardiac arrest syndrome (PCAS).What are the clinical implications?: Identification of mtDNA as a key driver of sterile inflammation in PCAS highlights a potential target for interventions aimed at reducing multi-organ damage and improving neurological outcomes.Therapeutic strategies to block mtDNA release or downstream signaling (e.g., TLR9/cGAS inhibition) may limit harmful pro-inflammatory cascades and bolster long-term survival following resuscitation from cardiac arrest.Early clinical screening for elevated EV-encapsulated mtDNA could help refine prognostic evaluations, complement current biomarkers, and guide personalized therapy to lessen the inflammatory burden of PCAS.
Collapse
|
42
|
Zemanek T, Danisovic L, Nicodemou A. Exosomes and solid cancer therapy: where are we now? Med Oncol 2025; 42:77. [PMID: 39961904 PMCID: PMC11832697 DOI: 10.1007/s12032-025-02626-3] [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: 12/21/2024] [Accepted: 02/11/2025] [Indexed: 02/20/2025]
Abstract
Cancer immunotherapy has revolutionized oncology, offering new hope for patients with previously incurable cancers. However, solid tumors remain a significant challenge due to immune evasion, therapeutic resistance, and the immunosuppressive tumor microenvironment. Exosomes, a specialized subset of extracellular vesicles, have emerged as promising tools in cancer therapy owing to their unique role in intercellular communication and immune modulation. These vesicles transport antigens, major histocompatibility complex (MHC) molecules, and immune-modulatory cargo, positioning them as potential platforms for cancer vaccines, drug delivery systems, and combinatorial therapies. Advances in engineered exosomes have improved drug bioavailability, tumor targeting, and immune stimulation, showcasing their potential in personalized medicine. This review highlights their multifaceted role in the tumor microenvironment, and their mechanisms of action in solid cancer therapy. Additionally, we discuss emerging strategies to overcome clinical and technical hurdles, paving the way for novel and effective cancer treatments.
Collapse
Affiliation(s)
- Tomas Zemanek
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia
- GAMMA - ZA s.r.o., Trencin, Slovakia
| | - Lubos Danisovic
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Andreas Nicodemou
- Institute of Medical Biology, Genetics and Clinical Genetics, Faculty of Medicine, Comenius University, Bratislava, Slovakia.
- GAMMA - ZA s.r.o., Trencin, Slovakia.
| |
Collapse
|
43
|
Pachane BC, Rodriguez BV, Shirk EN, Gololobova O, Carlson B, Queen SE, Erickson LD, Selistre-de-Araujo HS, Witwer KW. An ex vivo and in vitro investigation of extracellular vesicle interactions with B cells of Macaca nemestrina and humans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.12.637883. [PMID: 39990430 PMCID: PMC11844526 DOI: 10.1101/2025.02.12.637883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Extracellular vesicles may modify recipient cell behavior through multiple mechanisms, including interacting with the cell surface or internal membrane components and delivering luminal cargo to the cytoplasm. Here, we use a previously established ex vivo approach to investigate the cellular fate of EVs spiked into whole blood samples from nonhuman primate (NHP) and human donors and contrast these findings with results from in vitro assays. We report that EVs are internalized by NHP and human B cells while also associating to some degree with other PBMCs. EVs exhibit greater association with B cells in ex vivo whole blood compared to isolated B cells, suggesting that blood components may promote EV interactions or that cell isolation factors may inhibit this association. Cellular uptake of EVs involves clathrin-dependent endocytosis and may be aided by other pathways, including direct EV-cell membrane fusion. Overall, our data suggest that EV association, including uptake, by B cells occurs in at least two primate species. These findings highlight the potential to develop new strategies to either enhance or inhibit EV tropism toward B cells.
Collapse
|
44
|
Abbasi R, Alamdari-Mahd G, Maleki-Kakelar H, Momen-Mesgin R, Ahmadi M, Sharafkhani M, Rezaie J. Recent advances in the application of engineered exosomes from mesenchymal stem cells for regenerative medicine. Eur J Pharmacol 2025; 989:177236. [PMID: 39753159 DOI: 10.1016/j.ejphar.2024.177236] [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: 10/25/2024] [Revised: 12/14/2024] [Accepted: 12/23/2024] [Indexed: 01/12/2025]
Abstract
Exosomes, cell-derived vesicles produced by cells, are fascinating and drawing growing interest in biomedical exploration due to their exceptional properties. There is intriguing evidence that exosomes are involved in major biological processes, including diseases and regeneration. Exosomes from mesenchymal stem cells (MSCs) have shown promising outcomes in regenerative medicine. Numerous studies suggest that exosomes have several advantages over conventional synthetic nanocarriers, opening novel frontiers for innovative drug delivery. Regenerative medicine has demonstrated the profound therapeutic outcomes of engineered or loaded exosomes from MSCs. Different methods are being used to modify or/load exosomes. These exosomes can improve cell signaling pathways for bone and cartilage diseases, liver diseases, nerve tissues, kidney diseases, skin tissue, and cardiovascular diseases. Despite extensive research, clinical translation of these exosomes remains a challenge. The optimization of cargo loading methods, efficiency, physiological stability, and the isolation and characterization of exosomes present some challenges. The upcoming examination should include the development of large-scale, quality-controllable production approaches, the modification of drug loading approaches, and numerous in vivo investigations and clinical trials. Here, we provided an informative overview of the extracellular vesicles and modification/loading methods of exosomes. We discuss the last exosome research on regeneration disorders, highlighting the therapeutic applications of MSCs-derived exosomes. We also highlight future directions and challenges, underscoring the significance of addressing the main questions in the field.
Collapse
Affiliation(s)
- Reza Abbasi
- Department of Biology, Urmia University, Urmia, Iran
| | - Ghazal Alamdari-Mahd
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Hadi Maleki-Kakelar
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | | | - Mahdi Ahmadi
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohaddeseh Sharafkhani
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
45
|
Kuang L, Wu L, Li Y. Extracellular vesicles in tumor immunity: mechanisms and novel insights. Mol Cancer 2025; 24:45. [PMID: 39953480 PMCID: PMC11829561 DOI: 10.1186/s12943-025-02233-w] [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: 09/30/2024] [Accepted: 01/14/2025] [Indexed: 02/17/2025] Open
Abstract
Extracellular vesicles (EVs), nanoscale vesicles secreted by cells, have attracted considerable attention in recent years due to their role in tumor immunomodulation. These vesicles facilitate intercellular communication by transporting proteins, nucleic acids, and other biologically active substances, and they exhibit a dual role in tumor development and immune evasion mechanisms. Specifically, EVs can assist tumor cells in evading immune surveillance and attack by impairing immune cell function or modulating immunosuppressive pathways, thereby promoting tumor progression and metastasis. Conversely, they can also transport and release immunomodulatory factors that stimulate the activation and regulation of the immune system, enhancing the body's capacity to combat malignant diseases. This dual functionality of EVs presents promising avenues and targets for tumor immunotherapy. By examining the biological characteristics of EVs and their influence on tumor immunity, novel therapeutic strategies can be developed to improve the efficacy and relevance of cancer treatment. This review delineates the complex role of EVs in tumor immunomodulation and explores their potential implications for cancer therapeutic approaches, aiming to establish a theoretical foundation and provide practical insights for the advancement of future EVs-based cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Liwen Kuang
- School of Medicine, Chongqing University, Chongqing, China
| | - Lei Wu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yongsheng Li
- School of Medicine, Chongqing University, Chongqing, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
46
|
Chen J, Madhiyan M, Moor KJ, Chen H, Shuai D. Kinetics and Mechanisms of Solar UVB Disinfection of Vesicle-Cloaked Murine Norovirus Clusters and Free Noroviruses. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025; 59:2461-2472. [PMID: 39893675 DOI: 10.1021/acs.est.4c12583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Human norovirus, a major global cause of gastroenteritis, forms vesicle-cloaked virus clusters (known as viral vesicles), showing increased infectivity and persistence in aquatic environments. We investigated UVB disinfection, a key mechanism of solar disinfection commonly employed in developing countries, targeting murine norovirus vesicles and free murine noroviruses as surrogates for human noroviruses. At low viral concentrations of 109 gene copies per liter, viral infectivity loss as quantified by the integrated cell culture-reverse transcription-quantitative polymerase chain reaction (ICC-RT-qPCR) indicated that vesicles were 1.51 to 1.73 times more resistant to disinfection compared to free viruses. Virus inactivation was primarily due to protein damage as quantified by bicinchoninic acid and Western blot assays, and the damage of virus binding to host cells as quantified by RT-qPCR. Molecular simulations predicted that the oxidation of a tyrosine residue in the viral protein 1 prohibited binding. UVB irradiation of viral/vesicle proteins resulted in 1O2 formation as quantified by time-resolved phosphorescence, and for the first time, endogenous 1O2 was confirmed to contribute to virus inactivation by UVB. Our study recognizes the limitation of UVB disinfection of viral vesicles particularly in solar wastewater treatment and advocates for enhanced disinfection strategies to protect public health.
Collapse
Affiliation(s)
- Jiahao Chen
- Department of Civil and Environmental Engineering, The George Washington University, Washington, District of Columbia 20052, United States
| | - Monika Madhiyan
- Utah Water Research Laboratory, Department of Civil and Environmental Engineering, Utah State University, Logan, Utah 84322, United States
| | - Kyle J Moor
- Utah Water Research Laboratory, Department of Civil and Environmental Engineering, Utah State University, Logan, Utah 84322, United States
| | - Hanning Chen
- Texas Advanced Computing Center, The University of Texas at Austin, Austin, Texas 78758, United States
| | - Danmeng Shuai
- Department of Civil and Environmental Engineering, The George Washington University, Washington, District of Columbia 20052, United States
| |
Collapse
|
47
|
Mohamed AH, Abaza T, Youssef YA, Rady M, Fahmy SA, Kamel R, Hamdi N, Efthimiado E, Braoudaki M, Youness RA. Extracellular vesicles: from intracellular trafficking molecules to fully fortified delivery vehicles for cancer therapeutics. NANOSCALE ADVANCES 2025; 7:934-962. [PMID: 39823046 PMCID: PMC11733735 DOI: 10.1039/d4na00393d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 12/22/2024] [Indexed: 01/19/2025]
Abstract
Extracellular vesicles (EVs) are emerging as viable tools in cancer treatment due to their ability to carry a wide range of theranostic activities. This review summarizes different forms of EVs such as exosomes, microvesicles, apoptotic bodies, and oncosomes. It also sheds the light onto isolation methodologies, characterization techniques and therapeutic applications of all discussed EVs. Evidence indicates that EVs are particularly effective in delivering chemotherapeutic medications, and immunomodulatory agents. However, the advancement of EV-based therapies into clinical practice is hindered by challenges including EVs heterogeneity, cargo loading efficiency, and in vivo stability. Overall, EVs have the potential to change cancer therapeutic paradigms. Continued research and development activities are critical for improving EV-based medications and increasing their therapeutic impact.
Collapse
Affiliation(s)
- Adham H Mohamed
- Department of Chemistry, Faculty of Science, Cairo University 12613 Giza Egypt
| | - Tasneem Abaza
- Biotechnology and Biomolecular Chemistry Program, Faculty of Science, Cairo University 12613 Giza Egypt
- Université Paris-Saclay, Université d'Evry Val D'Essonne 91000 Évry-Courcouronnes Île-de-France France
| | - Yomna A Youssef
- Department of Physiology, Faculty of Physical Therapy, German International University (GIU) 11835 Cairo Egypt
- Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| | - Mona Rady
- Microbiology, Immunology and Biotechnology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC) 11835 Cairo Egypt
- Faculty of Biotechnology, German International University New Administrative Capital 11835 Cairo Egypt
| | - Sherif Ashraf Fahmy
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg Robert-Koch-Str. 4 35037 Marburg Germany
| | - Rabab Kamel
- Pharmaceutical Technology Department, National Research Centre 12622 Cairo Egypt
| | - Nabila Hamdi
- Pharmacology and Toxicology Department, Faculty of Pharmacy and Biotechnology, German University in Cairo (GUC) 11835 Cairo Egypt
| | - Eleni Efthimiado
- Inorganic Chemistry Laboratory, Chemistry Department, National and Kapodistrian University of Athens Athens Greece
| | - Maria Braoudaki
- Department of Clinical, Pharmaceutical, and Biological Science, School of Life and Medical Sciences, University of Hertfordshire Hatfield AL10 9AB UK
| | - Rana A Youness
- Molecular Biology and Biochemistry Department, Faculty of Biotechnology, German International University (GIU) 11835 Cairo Egypt
| |
Collapse
|
48
|
Finan JM, Guo Y, Bartlett AQ, Reyer M, Hawthorne K, Haerr M, Halamish H, Lamikanra O, Calvert V, Chen C, Xia Z, Petricoin EF, Sears RC, Byrne KT, Brody JR. Pancreatic cancer-intrinsic HuR regulates the pro-tumorigenic properties of extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.08.637077. [PMID: 39975239 PMCID: PMC11839136 DOI: 10.1101/2025.02.08.637077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) tumors contain chaotic vasculature that limits immune surveillance and promotes early events in the metastatic cascade. However, current antiangiogenic therapies have failed in PDAC, and thus, it remains important to uncover mechanisms by which cancer cells signal to endothelial cells to increase angiogenesis. Our lab has shown that the tumor-intrinsic RNA-binding protein HuR ( ELAVL1 ) plays an important role re-shaping the tumor microenvironment (TME) by regulating the stability and translation of cytokine encoding transcripts. Herein, we demonstrate that PDAC-intrinsic HuR influences endothelial cell function in the TME via extracellular vesicle (EV) signaling, an underexplored signaling axis in tumor progression. We found that HuR knockout (KO) tumors have impaired growth in an immunocompetent mouse model, and that administering purified wildtype (WT) EVs can increase tumor growth. Further, we observed that PDAC EVs contain HuR-dependent mRNA and protein cargoes relating to endothelial cell function and angiogenesis. Treatment of endothelial cells with HuR WT EVs strongly increased the expression of genes involved in barrier function and endothelial cell development, and directly increased their migratory and tube forming functions. In an immunocompetent orthotopic mouse model of PDAC, we showed that HuR increases endothelial cell presence and sprouting, while decreasing ICAM-1 expression. Importantly, we found utilizing a genetic EV reporter, that decreased ICAM-1 within WT tumors occurs in endothelial cells that have imported PDAC EVs, suggesting that this signaling axis is directly modulating endothelial cell behavior in vivo . Collectively, our data reveal a new role of HuR in EV signaling to endothelial cells, promoting angiogenesis while restricting endothelial cell leukocyte trafficking behavior.
Collapse
|
49
|
Talatapeh SP, Rezaie J, Nejati V. Extracellular Vesicle-based Delivery of Paclitaxel to Lung Cancer Cells: Uptake, Anticancer Effects, Autophagy and Mitophagy Pathways. Arch Med Res 2025; 56:103194. [PMID: 39922153 DOI: 10.1016/j.arcmed.2025.103194] [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: 09/08/2024] [Revised: 01/10/2025] [Accepted: 01/28/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Due to their unique properties, extracellular vesicles (EVs) are promising nanocarriers for exogenous drug delivery. AIM We prepared a drug delivery system based on large EVs (LEVs) containing paclitaxel (PTX) (LEVs-PTX) to investigate anticancer effects on lung cancer cells with a focus on autophagy. METHODS LEVs-PTX were isolated from lung cancer cells by ultracentrifugation and characterized using different techniques. Rhodamine B dye (Rh B) was used to label LEVs-PTX for cell tracking. MTT assay was performed to investigate the cellular toxicity of PTX and LEVs-PTX for 24 h and 48 h. The uptake of LEVs-PTX was monitored by immunofluorescence microscopy in breast and lung cancer cells. A colorimetric assay was performed to evaluate apoptosis, while Western blotting assays were used to investigate autophagy proteins. Real-time PCR was used to measure mitophagy genes. RESULTS Characterization techniques showed that LEVs were isolated and loaded with PTX. Rh B labeled LEVs, which was confirmed by a fluorescence spectrophotometer. Immunofluorescence microscopy showed that the lung and breast cancer cells had captured LEVs. Cell viability was decreased in LEVs-PTX cells which coincided with an increase in caspase-3 activity in LEVs-PTX cells. The Beclin-1 protein level and LC3 II/I ratio decreased, while the P62 protein level was increased in LEVs-PTX cells. The mitophagy genes such as Pink-1 and Parkin were upregulated in LEVs-PTX cells. CONCLUSION The data show that LEVs-PTX induced apoptosis, which inhibited the autophagy pathway and increased mitophagy markers, suggesting damage to cell organelles through intracellular delivery of PTX.
Collapse
Affiliation(s)
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia, Iran.
| | - Vahid Nejati
- Department of Biology, Urmia University, Urmia, Iran
| |
Collapse
|
50
|
Crooke PS, Tossberg JT, Aune TM. Increased unedited Alu RNA patterns found in cortex extracellular vesicles in Alzheimer's disease resemble hippocampus vasculature Alu RNA editing patterns but not cortex Alu RNA editing patterns. J Alzheimers Dis 2025; 103:1216-1225. [PMID: 39865681 DOI: 10.1177/13872877241313054] [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] [Indexed: 01/28/2025]
Abstract
BACKGROUND Endogenous Alu RNAs form double-stranded RNAs recognized by double-stranded RNA sensors and activate IRF and NF-kB transcriptional paths and innate immunity. Deamination of adenosines to inosines by the ADAR family of enzymes, a process termed A-to-I editing, disrupts double-stranded RNA structure and prevents innate immune activation. Innate immune activation is observed in Alzheimer's disease, the most common form of dementia. We have previously reported loss of A-to-I editing in hippocampus vasculature, but no change in cortex or cortex vasculature, associated with Alzheimer's disease. OBJECTIVE Here, we investigated the status of Alu RNA A-to-I editing in cortex extracellular vesicles in Alzheimer's disease. METHODS We used existing RNA-seq data sets and the SPRINT software package to determine levels of Alu RNA A-to-I editing in cortex extracellular vesicles in Alzheimer's disease and control groups and compared these editing profiles to those found in both total cortex and hippocampus vasculature. RESULTS We find substantial loss of Alu A-to-I editing in cortex extracellular vesicles in Alzheimer's disease. By measuring editing patterns on a gene-by-gene basis, we determined that editing patterns in cortex extracellular vesicles resemble editing patterns in hippocampus vasculature rather than total cortex. CONCLUSIONS We conclude that hippocampus vasculature unedited Alu RNAs are packaged in extracellular vesicles, travel to the cortex, deliver their cargo and stimulate innate immunity and alter other basic biological processes contributing to Alzheimer's disease progression.
Collapse
Affiliation(s)
- Philip S Crooke
- Department of Mathematics, Vanderbilt University, Nashville, TN, USA
| | - John T Tossberg
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas M Aune
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|