1
|
Chopra P, Fatima A, Mohapatra S, Murugaiyan K, Vemuganti GK, Rengan AK, Watson SL, Singh V, Basu S, Singh S. Extracellular vesicles in dry eye disease and Sjögren's syndrome: A systematic review on their diagnostic and therapeutic role. Surv Ophthalmol 2025; 70:499-515. [PMID: 39818361 DOI: 10.1016/j.survophthal.2025.01.003] [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: 05/20/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 01/18/2025]
Abstract
Extracellular vesicles (EVs), defined as membrane-bound vesicles released from all cells, are being explored for their diagnostic and therapeutic role in dry eye disease (DED). We systematically shortlisted 32 articles on the role of EVs in diagnosing and treating DED. We cover the progress in the last 2 decades on the classification and isolation of EVs and their role in DED. The diagnostic predictability of exosomes was evaluated in Sjögren syndrome (SS) patients' tears, plasma, and saliva, where upregulation of inflammatory proteins was reported uniformly across studies. Also, we evaluate the therapeutic effects of MSC-derived EVs in in vitro and in vivo studies of SS and DED mouse models. A significant response occurs at a functional level with improved tear production and saliva flow rate and at a cellular level with reduced lymphocyte infiltration, improved corneal structural integrity, decreased epithelial cell apoptosis, and dampening of the inflammatory cytokine response. The proposed mechanisms of EV action include PD-L1, PRDM, NLRP-3, and Nf-kb pathways, and an increase in M2 macrophage phenotype. Current use of exosomes in DED is limited due to their cumbersome isolation techniqus. Further research on human subjects is needed, in addition to optimizing exosome isolation and delivery methods.
Collapse
Affiliation(s)
- Prakshi Chopra
- Sydney Eye Hospital, Sydney, Australia; The University of Sydney, Australia
| | - Asra Fatima
- School of Medical Sciences, University of Hyderabad, India
| | - Sonali Mohapatra
- Brien Holden Centre for Eye Research (BHERC), L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Kavipriya Murugaiyan
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, India
| | | | - Aravind Kumar Rengan
- Department of Biomedical Engineering, Indian Institute of Technology, Hyderabad, India
| | | | - Vivek Singh
- Brien Holden Centre for Eye Research (BHERC), L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Sayan Basu
- Brien Holden Centre for Eye Research (BHERC), L V Prasad Eye Institute, Hyderabad, Telangana, India; Shantilal Shanghvi Cornea Institute, L V Prasad Eye Institute, Hyderabad, Telangana, India
| | - Swati Singh
- Centre for Ocular Regeneration (CORE), L V Prasad Eye Institute, Hyderabad, Telangana, India; Prof. Krothapalli Ravindranath Ophthalmic Research Biorepository, LV Prasad Eye Institute, Hyderabad, Telangana, India.
| |
Collapse
|
2
|
Zhang Y, Yue NN, Chen LY, Tian CM, Yao J, Wang LS, Liang YJ, Wei DR, Ma HL, Li DF. Exosomal biomarkers: A novel frontier in the diagnosis of gastrointestinal cancers. World J Gastrointest Oncol 2025; 17:103591. [DOI: 10.4251/wjgo.v17.i4.103591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/24/2025] [Accepted: 02/25/2025] [Indexed: 03/25/2025] Open
Abstract
Gastrointestinal (GI) cancers, which predominantly manifest in the stomach, colorectum, liver, esophagus, and pancreas, accounting for approximately 35% of global cancer-related mortality. The advent of liquid biopsy has introduced a pivotal diagnostic modality for the early identification of premalignant GI lesions and incipient cancers. This non-invasive technique not only facilitates prompt therapeutic intervention, but also serves as a critical adjunct in prognosticating the likelihood of tumor recurrence. The wealth of circulating exosomes present in body fluids is often enriched with proteins, lipids, microRNAs, and other RNAs derived from tumor cells. These specific cargo components are reflective of processes involved in GI tumorigenesis, tumor progression, and response to treatment. As such, they represent a group of promising biomarkers for aiding in the diagnosis of GI cancer. In this review, we delivered an exhaustive overview of the composition of exosomes and the pathways for cargo sorting within these vesicles. We laid out some of the clinical evidence that supported the utilization of exosomes as diagnostic biomarkers for GI cancers and discussed their potential for clinical application. Furthermore, we addressed the challenges encountered when harnessing exosomes as diagnostic and predictive instruments in the realm of GI cancers.
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
- Department of Medical Administration, Huizhou Institute for Occupational Health, Huizhou 516000, Guangdong Province, China
| | - Ning-Ning Yue
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University), Shenzhen 518000, Guangdong Province, China
| | - Li-Yu Chen
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Cheng-Mei Tian
- Department of Emergency, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People’s Hospital (Jinan University of Second Clinical Medical Sciences), Shenzhen 518000, Guangdong Province, China
| | - Li-Sheng Wang
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Yu-Jie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen 518000, Guangdong Province, China
| | - Dao-Ru Wei
- Department of Rehabilitation, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| | - Hua-Lin Ma
- Department of Nephrology, The Second Clinical Medical College, Jinan University, Shenzhen 518020, Guangdong Province, China
| | - De-Feng Li
- Department of Gastroenterology, Shenzhen People’s Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518000, Guangdong Province, China
| |
Collapse
|
3
|
Chansaenroj J, Kornsuthisopon C, Chansaenroj A, Samaranayake LP, Fan Y, Osathanon T. Potential of Dental Pulp Stem Cell Exosomes: Unveiling miRNA-Driven Regenerative Mechanisms. Int Dent J 2025; 75:415-425. [PMID: 39368923 DOI: 10.1016/j.identj.2024.08.019] [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: 07/11/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 10/07/2024] Open
Abstract
Human dental pulp stem cells (hDPSCs) have emerged as a promising resource in regenerative medicine due to their unique ability to secrete exosomes containing a diverse array of bioactive molecules, particularly microRNAs (miRNAs). These exosomes appear to be essential for stimulating regenerative mechanisms, especially those associated with stem cell pluripotency and tissue repair. However, several challenges such as cargo specificity and delivery efficiency need to be addressed to maximise the therapeutic potential of hDPSC-derived exosomes and miRNA-based therapies. This narrative review explores hDPSCs' potential in regenerative medicine by examining their role in tissue engineering, secretome composition, exosome function, exosomal miRNA in diverse models, and miRNA profiling. Therefore, it is imperative to sustain ongoing research on miRNA to advance clinical applications in the field of regenerative medicine and dentistry. A comprehensive understanding of the specific miRNA composition within hDPSC-derived exosomes is essential to elucidate their mechanistic roles in diverse disease states and to inform the development of innovative therapeutic strategies. These findings hold significant potential for the development of innovative regenerative therapies and emphasises the importance of establishing a strong connection between translational research discoveries and clinical applications. hDPSC-derived exosomes and miRNA-based therapies play a crucial role in immune modulation, regenerative dentistry, and tissue repair.
Collapse
Affiliation(s)
- Jira Chansaenroj
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chatvadee Kornsuthisopon
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| | - Ajjima Chansaenroj
- Department of Animal Husbandry, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Lakshman P Samaranayake
- Office of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Yi Fan
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Thanaphum Osathanon
- Center of Excellence for Dental Stem Cell Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
4
|
An C, Zhao Y, Guo L, Zhang Z, Yan C, Zhang S, Zhang Y, Shao F, Qi Y, wang X, Wang H, Zhang L. Innovative approaches to boost mesenchymal stem cells efficacy in myocardial infarction therapy. Mater Today Bio 2025; 31:101476. [PMID: 39896290 PMCID: PMC11787032 DOI: 10.1016/j.mtbio.2025.101476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/25/2024] [Accepted: 01/08/2025] [Indexed: 02/04/2025] Open
Abstract
Stem cell-based therapy has emerged as a promising approach for heart repair, potentially regenerating damaged heart tissue and improving outcomes for patients with heart disease. However, the efficacy of stem cell-based therapies remains limited by several challenges, including poor cell survival, low retention rates, poor integration, and limited functional outcomes. This article reviews current enhancement strategies to optimize mesenchymal stem cell therapy for cardiac repair. Key approaches include optimizing cell delivery methods, enhancing cell engraftment, promoting cell functions through genetic and molecular modifications, enhancing the paracrine effects of stem cells, and leveraging biomaterials and tissue engineering techniques. By focusing on these enhancement techniques, the paper highlights innovative approaches that can potentially transform stem cell therapy into a more viable and effective treatment option for cardiac repair. The ongoing research and technological advancements continue to push the boundaries, hoping to make stem cell therapy a mainstream treatment for heart disease.
Collapse
Affiliation(s)
- Chuanfeng An
- Ophthalmology and Transformational Innovation Research Center, Faculty of Medicine of Dalian University of Technology&Dalian Third People's Hospital, Dalian, 116033, PR China
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116033, PR China
| | - Yuan Zhao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Liaoning, Dalian, 116024, PR China
| | - Lipeng Guo
- Ophthalmology and Transformational Innovation Research Center, Faculty of Medicine of Dalian University of Technology&Dalian Third People's Hospital, Dalian, 116033, PR China
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116033, PR China
| | - Zhijian Zhang
- Department of Ophthalmology, Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, PR China
| | - Chunxiao Yan
- Department of Ophthalmology, Third People's Hospital of Dalian, Dalian Medical University, Dalian, 116033, PR China
| | - Shiying Zhang
- School of Dentistry, Shenzhen University, Shenzhen, 518060, PR China
| | - Yujie Zhang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Liaoning, Dalian, 116024, PR China
| | - Fei Shao
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Liaoning, Dalian, 116024, PR China
| | - Yuanyuan Qi
- Ophthalmology and Transformational Innovation Research Center, Faculty of Medicine of Dalian University of Technology&Dalian Third People's Hospital, Dalian, 116033, PR China
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116033, PR China
| | - Xun wang
- Ophthalmology and Transformational Innovation Research Center, Faculty of Medicine of Dalian University of Technology&Dalian Third People's Hospital, Dalian, 116033, PR China
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116033, PR China
| | - Huanan Wang
- MOE Key Laboratory of Bio-Intelligent Manufacturing, Dalian Key Laboratory of Artificial Organ and Regenerative Medicine, School of Bioengineering, Dalian University of Technology, Liaoning, Dalian, 116024, PR China
| | - Lijun Zhang
- Ophthalmology and Transformational Innovation Research Center, Faculty of Medicine of Dalian University of Technology&Dalian Third People's Hospital, Dalian, 116033, PR China
- Third People's Hospital of Dalian, Dalian Eye Hospital, Dalian, 116033, PR China
| |
Collapse
|
5
|
Jangam TC, Desai SA, Patel VP, Pagare NB, Raut ND. Exosomes as Therapeutic and Diagnostic Tools: Advances, Challenges, and Future Directions. Cell Biochem Biophys 2025:10.1007/s12013-025-01730-5. [PMID: 40122928 DOI: 10.1007/s12013-025-01730-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2025] [Indexed: 03/25/2025]
Abstract
Exosomes are tiny extracellular vesicles that are essential for intercellular communication and have shown great promise in the detection and treatment of disease. They are especially useful in the treatment of cancer, cardiovascular conditions, and neurological diseases because of their capacity to transport bioactive substances including proteins, lipids, and nucleic acids. Because of their low immunogenicity, ability to traverse biological barriers, and biocompatibility, exosome-based medicines have benefits over conventional treatments. Large-scale production, standardization of separation methods, possible immunological reactions, and worries about unforeseen biological effects are some of the obstacles that still need to be overcome. Furthermore, there are major barriers to the clinical use of exosomes due to their complex cargo sorting mechanisms and heterogeneity. Future studies should concentrate on enhancing separation and purification procedures, optimizing exosome engineering techniques, and creating plans to reduce immune system modifications. This review examines the most recent developments in exosome-based diagnostics and treatments, identifies current issues, and suggests ways to improve their clinical translation in the future.
Collapse
Affiliation(s)
- Tejas C Jangam
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Sharav A Desai
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India.
| | - Vipul P Patel
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Nishant B Pagare
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| | - Nikita D Raut
- Department of Pharmaceutical Biotechnology, Sanjivani College of Pharmaceutical Education & Research, Savitribai Phule Pune University, Kopargaon, Maharashtra, India
| |
Collapse
|
6
|
Rai S, Ray SK, Kanwar JR, Mukherjee S. Exosome-based therapeutics: Advancing drug delivery for neurodegenerative diseases. Mol Cell Neurosci 2025; 133:104004. [PMID: 40122271 DOI: 10.1016/j.mcn.2025.104004] [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: 01/24/2025] [Revised: 02/23/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025] Open
Abstract
Neurodegenerative disorders include Parkinson's disease, spinal cord injury, multiple sclerosis and Alzheimer's disease, cause gradual neuronal loss, protein misfolding, and accumulation, resulting in severe cognitive and movement deficits. Despite substantial study, therapeutic interventions are hampered by the blood-brain barrier, which prevents medication distribution to the central nervous system. Traditional pharmaceutical methods, such as small compounds, peptides, and inhibitors, have shown minimal effectiveness in addressing this obstacle. Exosomes are nanoscale membrane-bound vesicles that are primarily engaged in intercellular communication. They have the inherent capacity to cross the blood-brain barrier, which allows them to be used as medication delivery vehicles for brain illness therapy. Exosomes may be derived from a variety of cells like microglia, astrocytes identified according to origin, increasing their flexibility as drug delivery vehicles. Advanced engineering approaches optimise exosomes for tailored distribution across the blood-brain barrier, paving the path for novel neurodegenerative disease treatments. This review discusses the promise of exosome-based drug delivery, focussing on their composition, biogenesis, engineering, and applications in treating central nervous system illnesses, eventually overcoming the unmet hurdles of crossing the blood-brain barrier.
Collapse
Affiliation(s)
- Sakshi Rai
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh 462020, India
| | - Suman Kumar Ray
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University) Kalinganagar, Bhubaneswar 751003, Odisha, India
| | - Jagat R Kanwar
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh 462020, India
| | - Sukhes Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh 462020, India.
| |
Collapse
|
7
|
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
|
8
|
Wang F, Feng J, Jin A, Shao Y, Shen M, Ma J, Lei L, Liu L. Extracellular Vesicles for Disease Treatment. Int J Nanomedicine 2025; 20:3303-3337. [PMID: 40125438 PMCID: PMC11928757 DOI: 10.2147/ijn.s506456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
Traditional drug therapies suffer from problems such as easy drug degradation, side effects, and treatment resistance. Traditional disease diagnosis also suffers from high error rates and late diagnosis. Extracellular vesicles (EVs) are nanoscale spherical lipid bilayer vesicles secreted by cells that carry various biologically active components and are integral to intercellular communication. EVs can be found in different body fluids and may reflect the state of the parental cells, making them ideal noninvasive biomarkers for disease-specific diagnosis. The multifaceted characteristics of EVs render them optimal candidates for drug delivery vehicles, with evidence suggesting their efficacy in the treatment of various ailments. However, poor stability and easy degradation of natural EVs have affected their applications. To solve the problems of poor stability and easy degradation of natural EVs, they can be engineered and modified to obtain more stable and multifunctional EVs. In this study, we review the shortcomings of traditional drug delivery methods and describe how to modify EVs to form engineered EVs to improve their utilization. An innovative stimulus-responsive drug delivery system for EVs has also been proposed. We also summarize the current applications and research status of EVs in the diagnosis and treatment of different systemic diseases, and look forward to future research directions, providing research ideas for scholars.
Collapse
Affiliation(s)
- Fangyan Wang
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Jiayin Feng
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Anqi Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Yunyuan Shao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Mengen Shen
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Jiaqi Ma
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, People’s Republic of China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, People’s Republic of China
| |
Collapse
|
9
|
Ljungström M, Oltra E. Methods for Extracellular Vesicle Isolation: Relevance for Encapsulated miRNAs in Disease Diagnosis and Treatment. Genes (Basel) 2025; 16:330. [PMID: 40149481 PMCID: PMC11942051 DOI: 10.3390/genes16030330] [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: 01/31/2025] [Revised: 02/27/2025] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Extracellular vesicles (EVs) are nanovesicles that facilitate intercellular communication by carrying essential biomolecules under physiological and pathological conditions including microRNAs (miRNAs). They are found in various body fluids, such as blood, urine, and saliva, and their levels fluctuate with disease progression, making them valuable diagnostic tools. However, isolating EVs is challenging due to their small size and biological complexity. Here, we summarize the principles behind the most common EV isolation methods including ultracentrifugation, precipitation, immunoaffinity, sorting, ultrafiltration, size exclusion chromatography, and microfluidics while highlighting protocol strengths and weaknesses. We also review the main strategies to identify and quantify circulating miRNAs with a particular focus on EV-encapsulated miRNAs. Since these miRNAs hold special clinical interest derived from their superior stability and therapeutic potential, the information provided here should provide valuable guidance for future research initiatives in the promising field of disease diagnostic and treatment based on EV-encapsulated miRNAs.
Collapse
Affiliation(s)
- Maria Ljungström
- Escuela de Doctorado, School of Health Sciences, Catholic University of Valencia, 46001 Valencia, Spain;
| | - Elisa Oltra
- Department of Pathology, School of Health Sciences, Catholic University of Valencia, 46001 Valencia, Spain
| |
Collapse
|
10
|
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
|
11
|
Hernandez R, Garcia-Rodriguez NS, Arriaga MA, Perez R, Bala AA, Leandro AC, Diego VP, Almeida M, Parsons JG, Manusov EG, Galan JA. The hepatocellular model of fatty liver disease: from current imaging diagnostics to innovative proteomics technologies. Front Med (Lausanne) 2025; 12:1513598. [PMID: 40109726 PMCID: PMC11919916 DOI: 10.3389/fmed.2025.1513598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/06/2025] [Indexed: 03/22/2025] Open
Abstract
Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) is a prevalent chronic liver condition characterized by lipid accumulation and inflammation, often progressing to severe liver damage. We aim to review the pathophysiology, diagnostics, and clinical care of MASLD, and review highlights of advances in proteomic technologies. Recent advances in proteomics technologies have improved the identification of novel biomarkers and therapeutic targets, offering insight into the molecular mechanisms underlying MASLD progression. We focus on the application of mass spectrometry-based proteomics including single cell proteomics, proteogenomics, extracellular vesicle (EV-omics), and exposomics for biomarker discovery, emphasizing the potential of blood-based panels for noninvasive diagnosis and personalized medicine. Future research directions are presented to develop targeted therapies and improve clinical outcomes for MASLD patients.
Collapse
Affiliation(s)
- Renee Hernandez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Natasha S Garcia-Rodriguez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Marco A Arriaga
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Ricardo Perez
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Auwal A Bala
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Ana C Leandro
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Vince P Diego
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Marcio Almeida
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Jason G Parsons
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Eron G Manusov
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| | - Jacob A Galan
- Division of Human Genetics, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
- South Texas Diabetes and Obesity Institute, School of Medicine, The University of Texas Rio Grande Valley, Brownsville, TX, United States
| |
Collapse
|
12
|
Fu Y, Yang Q, Xu N, Zhang X. MiRNA affects the advancement of breast cancer by modulating the immune system's response. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167759. [PMID: 40037267 DOI: 10.1016/j.bbadis.2025.167759] [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: 10/29/2024] [Revised: 02/05/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
Breast cancer (BC), which is the most common tumor in women, has greatly endangered women's lives and health. Currently, patients with BC receive comprehensive treatments, including surgery, chemotherapy, radiotherapy, endocrine therapy, and targeted therapy. According to the latest research, the development of BC is closely related to the inflammatory immune response, and the immunogenicity of BC has steadily been recognized. As such, immunotherapy is one of the promising and anticipated forms of treatment for BC. The potential values of miRNA in the diagnosis and prognosis of BC have been established, and aberrant expression of associated miRNA can either facilitate or inhibit progression of BC. In the tumor immune microenvironment (TME), miRNAs are considered to be an essential molecular mechanism by which tumor cells interact with immunocytes and immunologic factors. Aberrant expression of miRNAs results in reprogramming of tumor cells actively, which may suppress the generation and activation of immunocytes and immunologic factors, avoid tumor cells apoptosis, and ultimately result in uncontrolled proliferation and deterioration. Therefore, through activating and regulating the immunocytes related to tumors and associated immunologic factors, miRNA can contribute to the advancement of BC. In this review, we assessed the function of miRNA and associated immune system components in regulating the advancement of BC, as well as the potential and viability of using miRNA in immunotherapy for BC.
Collapse
Affiliation(s)
- Yeqin Fu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang 310022, China
| | - Qiuhui Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), 310006, China
| | - Ning Xu
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China; School of Medicine, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Xiping Zhang
- Zhejiang cancer hospital, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
13
|
Zhang Q, Liu Q, Long K, Zhou K, Yang Z, Ge A, Hu J, Peng C, Wang W, Wang H, Li B. Visual and fluorescence dual mode platform for sensitive and accurate screening of breast carcinoma. Biosens Bioelectron 2025; 271:117047. [PMID: 39705784 DOI: 10.1016/j.bios.2024.117047] [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: 10/15/2024] [Revised: 12/03/2024] [Accepted: 12/06/2024] [Indexed: 12/23/2024]
Abstract
Compared to single-mode detection, dual-mode sensing strategies have garnered increasing attention from researchers due to their superior detection accuracy and reliability. Exosomes, as non-invasive biomarkers, hold significant potential for disease diagnosis. However, sensitive and precise detection of exosomes still presents considerable technical challenges. Inspired by the advantages of dual-mode detection, we developed a visual and fluorescence dual-mode platform (VFDMP) based on an aptamer strategy for exosome detection using enzyme-free nucleic acid amplification and nanomaterial-assisted cation exchange reactions (CERs). The Aptamer-ssDNA complexes capture tumor-derived exosomes, releasing abundant single-stranded DNA (ssDNAs), which then triggers the catalytic hairpin assembly (CHA) cycle, leading to the release of Ag+. The introduced CdTe quantum dots (QDs) act as signal reporters, interacting with Ag+ through CERs, and switching both fluorescence and visual signals from "on" to "off" to achieve exosome detection. Based on this innovative sensing principle, the developed FL/visual dual-mode aptasensor demonstrated excellent sensitivity and accuracy, achieving a low detection limit of 1.1 particles/μL by fluorometer, while exosome concentrations as low as 300 particles/mL could be visually distinguished by naked eye. Furthermore, this dual-mode platform can directly detect exosomes in clinical human serum samples, with only a small volume (10 μL) required. It can accurately differentiate between healthy individuals and breast cancer patients, as well as identify cancer stages (Stage II and Stage III) and subtypes (triple-negative, luminal B, and HER2+). These results suggest that the developed dual-mode detection strategy holds great promise as a sensitive, accurate method for biomarker analysis in clinical samples.
Collapse
Affiliation(s)
- Qiongdan Zhang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China
| | - Qingyi Liu
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China
| | - Kang Long
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China
| | - Kang Zhou
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China
| | - Zheng Yang
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Anqi Ge
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Jinhui Hu
- The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Caiyun Peng
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China; Science and Technology Innovation Center, Hunan University of Chinese Medicine, Changsha, China
| | - Wei Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China.
| | - Huizhen Wang
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China.
| | - Bin Li
- TCM and Ethnomedicine Innovation & Development International Laboratory, School of Pharmacy, Hunan·University of Chinese Medicine, Changsha, China.
| |
Collapse
|
14
|
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
|
15
|
Lee SH. The role of extracellular vesicles in embryo development: implications for reproductive health and therapeutic potential. Reprod Fertil Dev 2025; 37:RD24151. [PMID: 40153376 DOI: 10.1071/rd24151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 03/11/2025] [Indexed: 03/30/2025] Open
Abstract
Extracellular vesicles (EVs) contain various biological molecules, such as proteins, lipids, and diverse nucleic acids, which alter various physiological and pathological processes in recipient cells. This review focuses on the current understanding of the biological characteristics of EVs on embryo development and their potential therapeutic value in treating reproductive disorders. EVs play a crucial role in early embryo development, from fertilization to the pre-implantation stage, gastrulation, cell differentiation, and organogenesis. During the pre-implantation period, EVs interact with maternal reproductive tissue and promote implantation receptivity. In gastrulation, EVs regulate cell differentiation, contributing to tissue formation and maintenance. Abnormal bioactive molecules in EVs are closely related to developmental disorders. Thus, EVs have the potential to serve as biomarkers. Moreover, EVs can serve as therapeutic agents, delivering genetic material for targeted tissue/organs. The findings of this review highlight the potential role of EVs in intercellular signaling during embryo development. This can help advance assisted reproductive technologies and therapies to overcome infertility issues and developmental disorders.
Collapse
Affiliation(s)
- Seok Hee Lee
- Center for Reproductive Sciences, Department of Obstetrics and Gynecology, University of California San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
16
|
Ramu D, Kim E. Exosomal Lipids in Cancer Progression and Metastasis. Cancer Med 2025; 14:e70687. [PMID: 40111100 PMCID: PMC11924287 DOI: 10.1002/cam4.70687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 01/26/2025] [Accepted: 01/29/2025] [Indexed: 03/22/2025] Open
Abstract
BACKGROUND Metastasis is the primary cause of cancer mortality. It is responsible for 90% of all cancer-related deaths. Intercellular communication is a crucial feature underlying cancer metastasis and progression. Cancerous tumors secrete membrane-derived small extracellular vesicles (30-150 nm) into their extracellular milieu. These tiny organelles, known as exosomes, facilitate intercellular communication by transferring bioactive molecules. These exosomes harbor different cargos, such as proteins, nucleic acids, and lipids, that mediate multifaceted functions in various oncogenic processes. Of note, the amount of lipids in exosomes is multifold higher than that of other cargos. Most studies have investigated the role of exosomes' protein and nucleic acid content in various oncogenic processes, while the role of lipid cargo in cancer pathophysiology remains largely obscure. MATERIALS AND METHODS We conducted an extensive literature review on the role of exosomes and lipids in cancer progression, specifically addressing the topic of exosomal lipids and their involvement in cancer metastasis and progression. CONCLUSIONS This review aims to shed light on the lipid contents of exosomes in cancer metastasis. In this context, the role of exosomal lipids in signaling pathways, immunomodulation, and energy production for cancer cell survival provides insights into overcoming cancer progression and metastasis.
Collapse
Affiliation(s)
- Dandugudumula Ramu
- Division of ABB Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| | - Eunjoo Kim
- Division of ABB Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu, Republic of Korea
| |
Collapse
|
17
|
Yi S, Jung E, Kim H, Choi J, Kim S, Lim E, Kim K, Kang T, Jung J. Harnessing Lactobacillus reuteri-Derived Extracellular Vesicles for Multifaceted Cancer Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2406094. [PMID: 39422169 PMCID: PMC11899519 DOI: 10.1002/smll.202406094] [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: 07/19/2024] [Revised: 10/09/2024] [Indexed: 10/19/2024]
Abstract
Extracellular vesicles (EVs) have emerged as valuable biological materials for treating intractable diseases. Extensive studies are conducted on EVs derived from various cellular sources. In this study, EVs derived from Lactobacillus reuteri (L. reuteri), a probiotic, exhibit remarkable cancer therapeutic efficacy when administered orally is reported. These L. reuteri-derived EVs (REVs) demonstrate stability in the gastrointestinal tract and exert significant anti-tumor effects. Using A549 cells and murine models, we confirmed that REVs mediate their therapeutic effects by modulating apoptotic signaling pathways. Furthermore, the combination of REV with drugs enhances tumor ablation and induces immunogenic cell death. In a mouse model, oral administration of REVs encapsulating indocyanine green followed by photothermal therapy led to complete tumor elimination within 32 days. REVs represent a promising biological therapeutic platform for cancer treatment, either independently or in combination with other therapies, depending on the treatment objectives.
Collapse
Grants
- KGM5472413 Korea Research Institute of Bioscience and Biotechnology
- National NanoFab Center
- RS-2024-00401639 Ministry of Agriculture, Food and Rural Affairs
- 2021003370003 Ministry of Environment
- RS-2022-00154853 Ministry of Trade, Industry and Energy
- RS-2024-00403563 Ministry of Trade, Industry and Energy
- RS-2024-00432382 Ministry of Trade, Industry and Energy
- 2021M3H4A1A02051048 Ministry of Science and ICT, South Korea
- 2023R1A2C2005185 Ministry of Science and ICT, South Korea
- 2021M3E5E3080844 Ministry of Science and ICT, South Korea
- 2022R1C1C1008815 Ministry of Science and ICT, South Korea
- RS-2024-00348576 Ministry of Science and ICT, South Korea
- RS-2024-00438316 Ministry of Science and ICT, South Korea
- RS-2024-00459749 Ministry of Science and ICT, South Korea
- Korea Research Institute of Bioscience and Biotechnology
- National NanoFab Center
- Ministry of Agriculture, Food and Rural Affairs
- Ministry of Environment
- Ministry of Trade, Industry and Energy
- Ministry of Science and ICT, South Korea
Collapse
Affiliation(s)
- Soyeon Yi
- Bionanotechnology Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)125 Gwahak‐ro, Yuseong‐guDaejeon34141Republic of Korea
| | - Eunkyeong Jung
- Bionanotechnology Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)125 Gwahak‐ro, Yuseong‐guDaejeon34141Republic of Korea
| | - Hyeran Kim
- Bionanotechnology Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)125 Gwahak‐ro, Yuseong‐guDaejeon34141Republic of Korea
| | - Jinsol Choi
- Bionanotechnology Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)125 Gwahak‐ro, Yuseong‐guDaejeon34141Republic of Korea
- College of PharmacyKorea UniversitySejong30019Republic of Korea
| | - Suhyeon Kim
- Bionanotechnology Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)125 Gwahak‐ro, Yuseong‐guDaejeon34141Republic of Korea
- Department of Fundamental Pharmaceutical SciencesGraduate SchoolKyung Hee University26 Kyungheedae‐Ro, Dongdaemun‐GuSeoul02447Republic of Korea
| | - Eun‐Kyung Lim
- Bionanotechnology Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)125 Gwahak‐ro, Yuseong‐guDaejeon34141Republic of Korea
- Department of NanobiotechnologyKRIBB School of BiotechnologyUST217 Gajeong‐ro, Yuseong‐guDaejeon34113Republic of Korea
- School of PharmacySungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwon16419Republic of Korea
| | - Kwang‐Sun Kim
- Department of Chemistry and Chemistry Institute for Functional MaterialsPusan National University2 Busandaehak‐ro, Geumjeon‐guBusan46241Republic of Korea
| | - Taejoon Kang
- Bionanotechnology Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)125 Gwahak‐ro, Yuseong‐guDaejeon34141Republic of Korea
- School of PharmacySungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwon16419Republic of Korea
| | - Juyeon Jung
- Bionanotechnology Research CenterKorea Research Institute of Bioscience and Biotechnology (KRIBB)125 Gwahak‐ro, Yuseong‐guDaejeon34141Republic of Korea
- Department of NanobiotechnologyKRIBB School of BiotechnologyUST217 Gajeong‐ro, Yuseong‐guDaejeon34113Republic of Korea
- School of PharmacySungkyunkwan University2066 Seobu‐ro, Jangan‐guSuwon16419Republic of Korea
| |
Collapse
|
18
|
Xu Y, Zhang R, Du X, Huang Y, Gao Y, Wen Y, Qiao D, Sun N, Liu Z. Identification of aberrant plasma vesicles containing AAK1 and CCDC18-AS1 in adolescents with major depressive disorder and preliminary exploration of treatment efficacy. Genomics 2025; 117:110993. [PMID: 39798887 DOI: 10.1016/j.ygeno.2025.110993] [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: 06/24/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
BACKGROUND Major depressive disorder (MDD) during adolescence significantly jeopardizes both mental and physical health. However, the etiology underlying MDD in adolescents remains unclear. METHODS A total of 74 adolescents with MDD and 40 health controls (HCs) who underwent comprehensive clinical and cognitive assessments were enrolled. Differential expression analysis was conducted on plasma extracellular vesicles (EVs) carrying long non-coding RNAs (lncRNAs) and messenger RNAs (mRNAs) by microarray analysis. Two possible lncRNA-miR-mRNA networks were established and candidate regulatory axes were generated using the StarBase, miRDB, and TargetScan bioinformatics databases. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to validate the candidate molecules and signaling axes in a clinical cohort. RESULTS A total of 3752 dysregulated lncRNAs and 1789 dysfunctional mRNAs were identified. Two candidate regulatory axes (AC156455.1/miR-126-5p/AAK1 and CCDC18-AS1/miR-6835-5p/CCND2) with potential connections with MDD were selected. The candidate molecules exhibit differential expression patterns among adolescents with MDD and HCs, as well as before and after treatment with sertraline in adolescents with MDD. Furthermore, AAK1, CCDC18-AS1, and miR-6835-5p expressions exhibited significant differences between the response and non-response groups. Baseline expression of CCDC18-AS1, miR-6835-5p, and CCND2 could predict the therapeutic effect of sertraline, which may be associated with reducing suicidal ideation and improving cognitive function. CONCLUSION Our study may provide insights into the understanding of the underlying pathological mechanisms in adolescents with MDD.
Collapse
Affiliation(s)
- Yifan Xu
- Department of Psychiatry, First Hospital /First Clinical Medical College of Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Rong Zhang
- Department of Psychiatry, First Hospital /First Clinical Medical College of Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xinzhe Du
- Department of Psychiatry, First Hospital /First Clinical Medical College of Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yangxi Huang
- Department of Psychiatry, First Hospital /First Clinical Medical College of Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yao Gao
- Department of Psychiatry, First Hospital /First Clinical Medical College of Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yujiao Wen
- Department of Psychiatry, First Hospital /First Clinical Medical College of Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Dan Qiao
- Department of Psychiatry, First Hospital /First Clinical Medical College of Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Ning Sun
- Department of Psychiatry, First Hospital /First Clinical Medical College of Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhifen Liu
- Department of Psychiatry, First Hospital /First Clinical Medical College of Shanxi Medical University, Taiyuan, China; Shanxi Key Laboratory of Artificial Intelligence Assisted Diagnosis and Treatment for Mental Disorder, First Hospital of Shanxi Medical University, Taiyuan, China..
| |
Collapse
|
19
|
Xu CX, Huang W, Shi XJ, Du Y, Liang JQ, Fang X, Chen HY, Cheng Y. Dysregulation of Serum Exosomal Lipid Metabolism in Schizophrenia: A Biomarker Perspective. Mol Neurobiol 2025; 62:3556-3567. [PMID: 39312067 DOI: 10.1007/s12035-024-04477-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 09/01/2024] [Indexed: 02/04/2025]
Abstract
Exosomes, crucial extracellular vesicles, have emerged as potential biomarkers for neurological conditions, including schizophrenia (SCZ). However, the exploration of exosomal lipids in the context of SCZ remains scarce, necessitating in-depth investigation. Leveraging ultra-performance liquid chromatography-tandem mass spectrometry (UPLC-MS/MS), this study aimed to characterize the lipidomic profile of serum exosomes from SCZ patients, assessing their potential as novel biomarkers for SCZ diagnosis through absolute quantitative lipidomics. Our comprehensive lipidomic analysis unveiled 39 serum exosomal lipids that were differentially expressed between SCZ patients (n = 20) and healthy controls (HC, n = 20). These findings revealed a profound dysregulation in lipid metabolism pathways, notably in sphingolipid metabolism, glycerophospholipid metabolism, and linoleic acid metabolism. Among these, seven exosomal lipids stood out for their diagnostic potential, exhibiting remarkable ability to differentiate SCZ patients from HCs with an unparalleled classification performance, evidenced by an area under the curve (AUC) of 0.94 (95% CI, 0.82-1.00). These lipids included specific ceramides and phosphoethanolamines, pointing to a distinct lipid metabolic fingerprint associated with SCZ. Furthermore, bioinformatic analyses reinforced the pivotal involvement of these lipids in SCZ-related lipid metabolic processes, suggesting their integral role in the disorder's pathophysiology. This study significantly advances our understanding of SCZ by pinpointing dysregulated exosomal lipid metabolism as a key factor in its pathology. The identified serum exosome-derived lipids emerge as compelling biomarkers for SCZ diagnosis, offering a promising avenue towards the development of objective and reliable diagnostic tools.
Collapse
Affiliation(s)
- Chen-Xi Xu
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, No. 27, South Street of Zhongguancun, Haidian District, Beijing, 100081, China
| | - Wei Huang
- The Third People's Hospital of Foshan, Foshan, Guangdong, China
| | - Xiao-Jie Shi
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China
| | - Yang Du
- Henan Mental Hospital, the Second Affiliated Hospital of Xinxiang Medical University, Xinxiang, China
| | - Jia-Quan Liang
- The Third People's Hospital of Foshan, Foshan, Guangdong, China
| | - Xuan Fang
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, No. 27, South Street of Zhongguancun, Haidian District, Beijing, 100081, China
| | - He-Yuan Chen
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, No. 27, South Street of Zhongguancun, Haidian District, Beijing, 100081, China
| | - Yong Cheng
- Center on Translational Neuroscience, College of Life and Environmental Sciences, Minzu University of China, No. 27, South Street of Zhongguancun, Haidian District, Beijing, 100081, China.
- Key Laboratory of Ethnomedicine of Ministry of Education, School of Pharmacy, Minzu University of China, Beijing, China.
- Institute of National Security, Minzu University of China, Beijing, China.
| |
Collapse
|
20
|
Maria MKM, Abdel Moniem EM, Hanafy AK, Farag DBE, Radwan IA, Abbass MMS, El Moshy S, Rady D, Dörfer CE, Fawzy El-Sayed KM. Age-Related Oral and Para-Oral Tissue Disorders: The Evolving Therapeutic and Diagnostic Potential of Exosomes. Dent J (Basel) 2025; 13:106. [PMID: 40136734 PMCID: PMC11941486 DOI: 10.3390/dj13030106] [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: 01/02/2025] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
This review highlights the key molecular and cellular mechanisms contributing to aging, such as DNA damage, mitochondrial dysfunction, telomere shortening, protein dysfunction, and defective autophagy. These biological mechanisms are involved in various oral health conditions prevalent in the elderly, including periodontal disease, oral cancer, xerostomia, dental caries, and temporomandibular joint disorders. Exosomes generated by mesenchymal stem cells possess substantial therapeutic potential. These exosomes are nanosized extracellular vesicles derived from cells and are involved in essential intercellular communication and tissue homeostasis. The exosome-based therapies proved superior to traditional cell-based approaches, due to lower immunogenicity, ease of storage, and avoidance of complications associated with cell transplantation. Furthermore, the diagnostic potential of exosomes as non-invasive biomarkers for aging processes and age-related oral diseases offers insights into disease diagnosis, staging, and monitoring. Among the challenges and future perspectives of translating exosome research from preclinical studies to clinical applications is the need for standardized procedures to fully harness the therapeutic and diagnostic capabilities of exosomes.
Collapse
Affiliation(s)
- Mohamed Khaled Mohamed Maria
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
| | | | - Ahmed Khaled Hanafy
- Oral Biology Department, Faculty of Dentistry, Egyptian Russian University, Badr City 11829, Egypt;
| | - Dina B. E. Farag
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
| | - Israa Ahmed Radwan
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 12588, Egypt
| | - Marwa M. S. Abbass
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 12588, Egypt
| | - Sara El Moshy
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 12588, Egypt
| | - Dina Rady
- Oral Biology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt; (M.K.M.M.); (D.B.E.F.); (I.A.R.); (M.M.S.A.); (S.E.M.); (D.R.)
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 12588, Egypt
| | - Christof E. Dörfer
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24118 Kiel, Germany;
| | - Karim M. Fawzy El-Sayed
- Stem Cells and Tissue Engineering Research Group, Faculty of Dentistry, Cairo University, Cairo 12588, Egypt
- Clinic for Conservative Dentistry and Periodontology, School of Dental Medicine, Christian Albrechts University, 24118 Kiel, Germany;
- Oral Medicine and Periodontology Department, Faculty of Dentistry, Cairo University, Cairo 12613, Egypt
| |
Collapse
|
21
|
Díaz Reyes M, Gatti S, Delgado Ocaña S, Ortega HH, Banchio C. Neuroprotective effect of NSCs-derived extracellular vesicles in Parkinson's disease models. Sci Rep 2025; 15:6092. [PMID: 39971975 PMCID: PMC11839983 DOI: 10.1038/s41598-025-87238-7] [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: 09/02/2024] [Accepted: 01/17/2025] [Indexed: 02/21/2025] Open
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by both motor and non-motor symptoms, caused by the degeneration and loss of dopaminergic neurons in the substantia nigra. Current therapies are limited to symptom management, unable to prevent neuronal loss or halt the progression of the disease. A significant limitation to more effective treatments is the difficulty of crossing the blood-brain barrier (BBB). Extracellular vesicles (EVs) communication plays a crucial role in several physiological processes within the nervous system. Notably, EVs have the unique ability to cross the BBB, making them a highly promising vehicle for delivering therapeutic agents directly to the brain. Given the rising prevalence of PD, the need for therapies that prevent neuronal death and promote cell survival is urgent. This study explores the potential of neural stem cell-derived extracellular vesicles (NSC-EVs) using two in vitro models of PD. Our findings demonstrate that NSC-EVs significantly enhance the survival of dopaminergic neurons by reducing apoptosis and showing strong neuroprotective effects. Notably, the natural extracellular vesicles used in this study are enriched with Catalase, a potent scavenger protein with antioxidant properties. This natural enrichment further strengthens their neuroprotective capacity, enabling them to mitigate oxidative stress and protect vulnerable neurons. The use of such naturally enriched extracellular vesicles represents a promising approach for developing innovative therapies to effectively combat Parkinson's disease.
Collapse
Affiliation(s)
- Mercyleidi Díaz Reyes
- Laboratorio de Biología Molecular y Celular de Lípidos, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET) Ocampo y Esmeralda, Predio CONICET and Departamento de Ciencias Biológicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000, Rosario, Argentina
| | - Sabrina Gatti
- Laboratorio de Biología Molecular y Celular de Lípidos, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET) Ocampo y Esmeralda, Predio CONICET and Departamento de Ciencias Biológicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000, Rosario, Argentina
| | - Susana Delgado Ocaña
- Laboratorio de Biología Molecular y Celular de Lípidos, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET) Ocampo y Esmeralda, Predio CONICET and Departamento de Ciencias Biológicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000, Rosario, Argentina
| | - Hugo H Ortega
- Centro de Medicina Comparada, ICiVet-Litoral, Universidad Nacional del Litoral-CONICET, 3080, Esperanza, Santa Fe, Argentina
| | - Claudia Banchio
- Laboratorio de Biología Molecular y Celular de Lípidos, Instituto de Biología Molecular y Celular de Rosario (IBR-CONICET) Ocampo y Esmeralda, Predio CONICET and Departamento de Ciencias Biológicas, Facultad de Ciencias Bioquímicas y Farmacéuticas, Universidad Nacional de Rosario, 2000, Rosario, Argentina.
| |
Collapse
|
22
|
Kim SY. Extracellular vesicles offer enticing opportunities to target and treat lung inflammation. Thorax 2025; 80:127-128. [PMID: 39900491 DOI: 10.1136/thorax-2024-222696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2025] [Indexed: 02/05/2025]
Affiliation(s)
- Sally Yunsun Kim
- Institute of Pharmaceutical Science, King's College London, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
23
|
Margolis LB, Sadovsky Y. When Extracellular Vesicles Go Viral: A Bird's Eye View. Pathog Immun 2025; 10:140-158. [PMID: 40017586 PMCID: PMC11867185 DOI: 10.20411/pai.v10i1.787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 01/22/2025] [Indexed: 03/01/2025] Open
Abstract
The science of extracellular vesicles (EVs) is a rapidly growing field that spans multiple aspects of normal physiology and pathophysiology. EVs play a critical role in most basic biological processes of cell-cell communications under normal conditions and in disease. EVs have "gone viral" not only in terms of research popularity, but also in our realization that they exhibit an elaborate crosstalk with viruses, particularly with the enveloped ones, which are also extracellular vesicles that are released by cells as a part of their virulence cycle yet are replicative. Here, we highlight some of the complexities underlying EV-virus crosstalk and pathways and provide our insights on key challenges from the viewpoint of EV biology.
Collapse
Affiliation(s)
- Leonid B. Margolis
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia
| | - Yoel Sadovsky
- Magee-Womens Research Institute, Department of OBGYN and Reproductive Sciences, Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
24
|
Pacilio S, Lombardi S, Costa R, Paris F, Petrocelli G, Marrazzo P, Cenacchi G, Alviano F. Role of Perinatal Stem Cell Secretome as Potential Therapy for Muscular Dystrophies. Biomedicines 2025; 13:458. [PMID: 40002871 PMCID: PMC11852414 DOI: 10.3390/biomedicines13020458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Inflammation mechanisms play a critical role in muscle homeostasis, and in Muscular Dystrophies (MDs), the myofiber damage triggers chronic inflammation which significantly controls the disease progression. Immunomodulatory strategies able to target inflammatory pathways and mitigate the immune-mediated damage in MDs may provide new therapeutic options. Owing to its capacity of influencing the immune response and enhancing tissue repair, stem cells' secretome has been proposed as an adjunct or standalone treatment for MDs. In this review study, we discuss the challenging points related to the inflammation condition characterizing MD pathology and provide a concise summary of the literature supporting the potential of perinatal stem cells in targeting and modulating the MD inflammation.
Collapse
Affiliation(s)
- Serafina Pacilio
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Sara Lombardi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Roberta Costa
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Francesca Paris
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Giovannamaria Petrocelli
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Pasquale Marrazzo
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Giovanna Cenacchi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Francesco Alviano
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| |
Collapse
|
25
|
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
|
26
|
Abbad L, Esteve E, Chatziantoniou C. Advances and challenges in kidney fibrosis therapeutics. Nat Rev Nephrol 2025:10.1038/s41581-025-00934-5. [PMID: 39934355 DOI: 10.1038/s41581-025-00934-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2025] [Indexed: 02/13/2025]
Abstract
Chronic kidney disease (CKD) is a major global health burden that affects more than 10% of the adult population. Current treatments, including dialysis and transplantation, are costly and not curative. Kidney fibrosis, defined as an abnormal accumulation of extracellular matrix in the kidney parenchyma, is a common outcome in CKD, regardless of disease aetiology, and is a major cause of loss of kidney function and kidney failure. For this reason, research efforts have focused on identifying mediators of kidney fibrosis to inform the development of effective anti-fibrotic treatments. Given the prominent role of the transforming growth factor-β (TGFβ) family in fibrosis, efforts have focused on inhibiting TGFβ signalling. Despite hopes raised by the efficacy of this approach in preclinical models, translation into clinical practice has not met expectations. Antihypertensive and antidiabetic drugs slow the decline in kidney function and could slow fibrosis but, owing to the lack of technologies for in vivo renal imaging, their anti-fibrotic effect cannot be truly assessed at present. The emergence of new drugs targeting pro-fibrotic signalling, or enabling cell repair and cell metabolic reprogramming, combined with better stratification of people with CKD and the arrival of nanotechnologies for kidney-specific drug delivery, open up new perspectives for the treatment of this major public health challenge.
Collapse
Affiliation(s)
- Lilia Abbad
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Emmanuel Esteve
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France
| | - Christos Chatziantoniou
- INSERM UMR S 1155, Common and Rare Kidney Diseases, Tenon Hospital, Faculty of Medicine, Sorbonne University, Paris, France.
| |
Collapse
|
27
|
Raizada G, Guillouzouic J, Rouleau A, Lesniewska E, Le Ferrec E, Elie-Caille C, Boireau W. NanoBioAnalytical (NBA) Platform to Decipher Extracellular Vesicles Secreted by Microvascular Endothelial Cells Under Benzo[a]pyrene Exposure. BIOSENSORS 2025; 15:103. [PMID: 39997005 PMCID: PMC11852858 DOI: 10.3390/bios15020103] [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: 12/22/2024] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025]
Abstract
Recent advances in the clinical extracellular vesicles (EVs) field highlight their potential as biomarkers for diverse diseases and therapeutic applications. This study provides an in-depth characterization of 10k EVs from human microvascular endothelial cells (HMEC-1) exposed to benzo[a]pyrene (B[a]P), a polycyclic aromatic hydrocarbon found in food and smoke. Given EVs' complexity, with numerous surface and cargo proteins, phenotyping remains challenging. Here, we introduce a multiplex biosensor, in µarray format, for profiling EVs from distinct cellular conditions, employing a multimodal approach that combines surface plasmon resonance imaging (SPRi) and in situ atomic force microscopy (AFM) to decipher EVs' biochemical and biophysical properties. SPRi experiments showed notable EV capture differences on ligands such as Anti-CD36, Anti-CD81, and Anti-ApoA between treated and control conditions, likely due to B[a]P exposure. A complementary AFM study and statistical analyses revealed size differences between EVs from treated and control samples, with ligands like Annexin-V, Anti-CD36, and Anti-VEGFR1 emerging as ligands specific to potential cytotoxicity biomarkers. Our findings suggest that B[a]P exposure may increase EV size and alter marker expression, indicating phenotypic shifts in EVs under cytotoxic stress. The original combination of SPRi and AFM reveals valuable data on the phenotypical and morphological heterogeneities of EV subsets linked to cytotoxic stresses and highlights the potential of EVs as specific toxicological markers.
Collapse
Affiliation(s)
- Geetika Raizada
- Université Marie et Louis Pasteur, CNRS, Institut FEMTO-ST, 25030 Besançon, France
| | - Joan Guillouzouic
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, 35000 Rennes, France
| | - Alain Rouleau
- Université Marie et Louis Pasteur, CNRS, Institut FEMTO-ST, 25030 Besançon, France
| | - Eric Lesniewska
- ICB UMR 6303 CNRS, University Bourgogne Europe, 21078 Dijon, France
| | - Eric Le Ferrec
- Univ Rennes, Inserm, EHESP, Irset (Institut de Recherche en Santé Environnement et Travail), UMR_S 1085, 35000 Rennes, France
| | - Céline Elie-Caille
- Université Marie et Louis Pasteur, CNRS, Institut FEMTO-ST, 25030 Besançon, France
| | - Wilfrid Boireau
- Université Marie et Louis Pasteur, CNRS, Institut FEMTO-ST, 25030 Besançon, France
| |
Collapse
|
28
|
Mangena V, Chanoch-Myers R, Sartore R, Paulsen B, Gritsch S, Weisman H, Hara T, Breakefield XO, Breyne K, Regev A, Chung K, Arlotta P, Tirosh I, Suvà ML. Glioblastoma Cortical Organoids Recapitulate Cell-State Heterogeneity and Intercellular Transfer. Cancer Discov 2025; 15:299-315. [PMID: 39373549 PMCID: PMC11803396 DOI: 10.1158/2159-8290.cd-23-1336] [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: 11/10/2023] [Revised: 08/27/2024] [Accepted: 10/03/2024] [Indexed: 10/08/2024]
Abstract
Glioblastoma (GBM) is characterized by heterogeneous malignant cells that are functionally integrated within the neuroglial microenvironment. In this study, we model this ecosystem by growing GBM into long-term cultured human cortical organoids that contain the major neuroglial cell types found in the cerebral cortex. Single-cell RNA sequencing analysis suggests that, compared with matched gliomasphere models, GBM cortical organoids more faithfully recapitulate the diversity and expression programs of malignant cell states found in patient tumors. Additionally, we observe widespread transfer of GBM transcripts and GFP to nonmalignant cells in the organoids. Mechanistically, this transfer involves extracellular vesicles and is biased toward defined GBM cell states and astroglia cell types. These results extend previous GBM organoid modeling efforts and suggest widespread intercellular transfer in the GBM neuroglial microenvironment. Significance: Models that recapitulate intercellular communications in GBM are limited. In this study, we leverage GBM cortical organoids to characterize widespread mRNA and GFP transfer from malignant to nonmalignant cells in the GBM neuroglial microenvironment. This transfer involves extracellular vesicles, may contribute to reprogramming the microenvironment, and may extend to other cancer types. See related commentary by Shakya et al., p. 261.
Collapse
Affiliation(s)
- Vamsi Mangena
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Rony Chanoch-Myers
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Rafaela Sartore
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Harvard University, Cambridge, Massachusetts
| | - Bruna Paulsen
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Harvard University, Cambridge, Massachusetts
| | - Simon Gritsch
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Hannah Weisman
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Toshiro Hara
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| | - Xandra O. Breakefield
- Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Department of Radiology, Center for Molecular Imaging Research, Massachusetts General Hospital and Program in Neuroscience, Boston, Massachusetts
| | - Koen Breyne
- Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Aviv Regev
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Genentech, South San Francisco, California
| | - Kwanghun Chung
- MIT Department of Chemical Engineering, Cambridge, Massachusetts
- Picower Institute for Learning and Memory, Cambridge, Massachusetts
| | - Paola Arlotta
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
- Harvard University, Cambridge, Massachusetts
| | - Itay Tirosh
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Mario L. Suvà
- Department of Pathology and Krantz Family Center for Cancer Research, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
- Broad Institute of Harvard and MIT, Cambridge, Massachusetts
| |
Collapse
|
29
|
Kraft A, Kirschner MB, Orlowski V, Ronner M, Bodmer C, Boeva V, Opitz I, Meerang M. Exploring RNA cargo in extracellular vesicles for pleural mesothelioma detection. BMC Cancer 2025; 25:212. [PMID: 39920655 PMCID: PMC11804012 DOI: 10.1186/s12885-025-13617-y] [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: 11/29/2024] [Accepted: 01/30/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Pleural Mesothelioma (PM) is a highly aggressive cancer, for which effective early detection remains a challenge due to limited screening options and low sensitivity of biomarkers discovered so far. While extracellular vesicles (EVs) have emerged as promising candidates for blood-based biomarkers, their role in PM has not been studied yet. In this study, we characterized the transcriptomic profile of EVs secreted by PM primary cells and explored their potential as a biomarker source for PM detection. METHODS We collected cell culture supernatant from early-passage PM cell cultures derived from the pleural effusion of 4 PM patients. EVs were isolated from the supernatant using Qiagen exoEasy Maxi kit. RNA isolation from EVs was done using the mirVana PARIS kit. Finally, single-end RNA sequencing was done with Illumina Novaseq 6000. RESULTS We identified a range of RNA species expressed in EVs secreted by PM cells, including protein-coding RNA (80%), long non-coding RNA (13%), pseudogenes (4.5%), and short non-coding RNA (1.6%). We detected a subset of genes associated with the previously identified epithelioid (32 genes) and sarcomatoid molecular components (36 genes) in PM-EVs. To investigate whether these markers could serve as biomarkers for PM detection in blood, we compared the RNA content of PM-EVs with the cargo of EVs isolated from the plasma of healthy donors (publicly available data). Majority of upregulated genes in PM-EVs were protein-coding and long non-coding RNAs. Interestingly, 25 of them were the sarcomatoid and epithelioid marker genes. Finally, functional analysis revealed that the PM-EV RNA cargo was associated with Epithelial-Mesenchymal transition, glycolysis, and hypoxia. CONCLUSIONS This is the first study to characterize the transcriptomic profile of EVs secreted by PM primary cell cultures, demonstrating their potential as biomarker source for early detection. Further investigation of the functional role of PM-EVs will provide new insights into disease biology and therapeutic avenues.
Collapse
Affiliation(s)
- Agnieszka Kraft
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- Institute for Machine Learning, Department of Computer Science, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Michaela B Kirschner
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Vanessa Orlowski
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Manuel Ronner
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Caroline Bodmer
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Valentina Boeva
- Institute for Machine Learning, Department of Computer Science, ETH Zurich, Zurich, Switzerland
- Swiss Institute of Bioinformatics (SIB), Zurich, Switzerland
- ETH AI Center, ETH Zurich, Zurich, Switzerland
- UMR 8104, UMR-S1016, Cochin InstituteCNRSParis Descartes University, Inserm U1016, 75014, Paris, France
| | - Isabelle Opitz
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland
- University of Zurich, Zurich, Switzerland
| | - Mayura Meerang
- Department of Thoracic Surgery, University Hospital Zurich, Raemistrasse 100, 8091, Zurich, Switzerland.
- University of Zurich, Zurich, Switzerland.
| |
Collapse
|
30
|
Peppicelli S, Calorini L, Bianchini F, Papucci L, Magnelli L, Andreucci E. Acidity and hypoxia of tumor microenvironment, a positive interplay in extracellular vesicle release by tumor cells. Cell Oncol (Dordr) 2025; 48:27-41. [PMID: 39023664 PMCID: PMC11850579 DOI: 10.1007/s13402-024-00969-z] [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] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
The complex and continuously evolving features of the tumor microenvironment, varying between tumor histotypes, are characterized by the presence of host cells and tumor cells embedded in a milieu shaped by hypoxia and low pH, resulting from the frequent imbalance between vascularity and tumor cell proliferation. These microenvironmental metabolic stressors play a crucial role in remodeling host cells and tumor cells, contributing to the stimulation of cancer cell heterogeneity, clonal evolution, and multidrug resistance, ultimately leading to progression and metastasis. The extracellular vesicles (EVs), membrane-enclosed structures released into the extracellular milieu by tumor/host cells, are now recognized as critical drivers in the complex intercellular communication between tumor cells and the local cellular components in a hypoxic/acidic microenvironment. Understanding the intricate molecular mechanisms governing the interactions between tumor and host cells within a hypoxic and acidic microenvironment, triggered by the release of EVs, could pave the way for innovative strategies to disrupt the complex interplay of cancer cells with their microenvironment. This approach may contribute to the development of an efficient and safe therapeutic strategy to combat cancer progression. Therefore, we review the major findings on the release of EVs in a hypoxic/acidic tumor microenvironment to appreciate their role in tumor progression toward metastatic disease.
Collapse
Affiliation(s)
- Silvia Peppicelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy.
| | - Lido Calorini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Francesca Bianchini
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Laura Papucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Lucia Magnelli
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| | - Elena Andreucci
- Department of Experimental and Clinical Biomedical Sciences "Mario Serio", University of Florence, Florence, 50134, Italy
| |
Collapse
|
31
|
Mishra S, Tejesvi MV, Hekkala J, Turunen J, Kandikanti N, Kaisanlahti A, Suokas M, Leppä S, Vihinen P, Kuitunen H, Sunela K, Koivunen J, Jukkola A, Kalashnikov I, Auvinen P, Kääriäinen OS, Peñate Medina T, Peñate Medina O, Saarnio J, Meriläinen S, Rautio T, Aro R, Häivälä R, Suojanen J, Laine M, Erawijattari PP, Lahti L, Karihtala P, Ruuska TS, Reunanen J. Gut microbiome-derived bacterial extracellular vesicles in patients with solid tumours. J Adv Res 2025; 68:375-386. [PMID: 38458256 PMCID: PMC11785572 DOI: 10.1016/j.jare.2024.03.003] [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: 12/21/2023] [Revised: 02/17/2024] [Accepted: 03/06/2024] [Indexed: 03/10/2024] Open
Abstract
INTRODUCTION Gut microbiome-derived nanoparticles, known as bacterial extracellular vesicles (bEVs), have garnered interest as promising tools for studying the link between the gut microbiome and human health. The diverse composition of bEVs, including their proteins, mRNAs, metabolites, and lipids, makes them useful for investigating diseases such as cancer. However, conventional approaches for studying gut microbiome composition alone may not be accurate in deciphering host-gut microbiome communication. In clinical microbiome research, there is a gap in the knowledge on the role of bEVs in solid tumor patients. OBJECTIVES Analyzing the functionality of bEVs using (meta)genomics and proteomics could highlight the unique aspects of host-gut microbiome interactions in solid tumor patients. Therefore, we performed a comparative analysis of the proteome and microbiota composition of gut microbiome-derived bEVs isolated from patients with solid tumors and healthy controls. METHODS After isolating bEVs from the feces of solid tumor patients and healthy controls, we performed spectrometry analysis of their proteomes and next-generation sequencing (NGS) of the 16S gene. We also investigated the gut microbiomes of feces from patients and controls using 16S sequencing and used machine learning to classify the samples into patients and controls based on their bEVs and fecal microbiomes. RESULTS Solid tumor patients showed decreased microbiota richness and diversity in both the bEVs and feces. However, the bEV proteomes were more diverse in patients than in the controls and were enriched with proteins associated with the metabolism of amino acids and carbohydrates, nucleotide binding, and oxidoreductase activity. Metadata classification of samples was more accurate using fecal bEVs (100%) compared with fecal samples (93%). CONCLUSION Our findings suggest that bEVs are unique functional entities. There is a need to explore bEVs together with conventional gut microbiome analysis in functional cancer research to decipher the potential of bEVs as cancer diagnostic or therapeutic biomarkers.
Collapse
Affiliation(s)
- Surbhi Mishra
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland.
| | - Mysore Vishakantegowda Tejesvi
- Biocenter Oulu, University of Oulu, Oulu, Finland; Ecology and Genetics, Faculty of Science, University of Oulu, Oulu, Finland
| | - Jenni Hekkala
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Jenni Turunen
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland; Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Niyati Kandikanti
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Anna Kaisanlahti
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Marko Suokas
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Sirpa Leppä
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, University of Helsinki, Helsinki, Finland
| | - Pia Vihinen
- FICAN West Cancer Centre and Department of Oncology, Turku University Hospital and University of Turku, 20521 Turku, Finland
| | - Hanne Kuitunen
- Department of Oncology, Oulu University Hospital, Oulu, Finland
| | | | - Jussi Koivunen
- Department of Medical Oncology and Radiotherapy and Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Arja Jukkola
- Tampere Cancer Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ilja Kalashnikov
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, University of Helsinki, Helsinki, Finland; Research Program Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Päivi Auvinen
- Cancer Center, Kuopio University Hospital, Northern Savonia Healthcare Municipality, Kuopio, Finland
| | - Okko-Sakari Kääriäinen
- Cancer Center, Kuopio University Hospital, Northern Savonia Healthcare Municipality, Kuopio, Finland
| | - T Peñate Medina
- Section Biomedical Imaging, Department of Radiology and Neuroradiology and Institute for Experimental Cancer Research, Kiel University, 24105 Kiel, Germany
| | - O Peñate Medina
- Section Biomedical Imaging, Department of Radiology and Neuroradiology and Institute for Experimental Cancer Research, Kiel University, 24105 Kiel, Germany; Lonza Netherlands B.V., 6167 RB Geleen, the Netherlands
| | - Juha Saarnio
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Sanna Meriläinen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Tero Rautio
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Raila Aro
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Reetta Häivälä
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Juho Suojanen
- Päijät-Häme Joint Authority for Health and Wellbeing, Department of Oral and Maxillofacial Surgery, Lahti Central Hospital, 15850 Lahti, Finland; Cleft Palate and Craniofacial Centre, Department of Plastic Surgery, Helsinki University Hospital, 00029 Helsinki, Finland; Clinicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Mikael Laine
- Department of Abdominal Surgery, Porvoo Hospital, Hospital District of Helsinki and Uusimaa, Porvoo, Finland
| | | | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Peeter Karihtala
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, University of Helsinki, Helsinki, Finland; Department of Oncology, Oulu University Hospital, Oulu, Finland
| | - Terhi S Ruuska
- Biocenter Oulu, University of Oulu, Oulu, Finland; Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland; Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| | - Justus Reunanen
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
32
|
Li Q, Liu Q, Li S, Zuo X, Zhou H, Gao Z, Xia B. Golgi-derived extracellular vesicle production induced by SARS-CoV-2 envelope protein. Apoptosis 2025; 30:197-209. [PMID: 39580578 DOI: 10.1007/s10495-024-02035-3] [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] [Accepted: 10/21/2024] [Indexed: 11/25/2024]
Abstract
Extracellular vesicles facilitate cell-to-cell communication, and some enveloped viruses utilize these vesicles as carriers to mediate viral transmission. SARS-CoV-2 envelope protein (2-E) forms a cation channel and overexpression of 2-E led to the generation of a distinct type of large extracellular vesicles (2-E-EVs). Although 2-E-EVs have been demonstrated to facilitate viral transmission in a receptor-independent way, the characteristics and biogenesis mechanism remain enigmatic. Via lipidomics and proteomic analysis, we found 2-E-EVs are distinct from endosome-derived exosomes. 2-E-EVs are notably enriched in Golgi apparatus components, aligning with the observed fragmentation in Golgi morphology. Through live cell imaging, we established a connection between 2-E-EVs formation, Golgi fragmentation, and channel activity, emphasizing the role of 2-E-EVs as ion channel-induced extracellular vesicles. Our work highlights 2-E-EVs as distinctive Golgi-derived vesicles, contributing to a deeper understanding of 2-E channel-mediated virus-host dynamics, with implications for therapeutic strategies and drug delivery.
Collapse
Affiliation(s)
- Qiguang Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China
| | - Qian Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Shuangqu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China
| | - Xiaoli Zuo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China
| | - Hu Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China.
| | - Zhaobing Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China.
- Zhongshan Institute of Drug Discovery, Institution for Drug Discovery Innovation, Chinese Academy of Science, Zhongshan, 528400, China.
| | - Bingqing Xia
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing, 100049, China.
| |
Collapse
|
33
|
Li H, Wang L, Cheng H, Zhang Q, Wang S, Zhong W, He C, Wei Q. Unlocking the Potential of Extracellular Vesicles in Cardiovascular Disease. J Cell Mol Med 2025; 29:e70407. [PMID: 39910696 PMCID: PMC11798870 DOI: 10.1111/jcmm.70407] [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: 07/08/2024] [Revised: 12/02/2024] [Accepted: 01/24/2025] [Indexed: 02/07/2025] Open
Abstract
Extracellular vesicles (EVs) are micro-nanoscale biological particles encapsulated by phospholipid bilayers, which regulate cell migration, angiogenesis and tumour cell growth by transmitting various biomolecules such as nucleic acids and proteins. EVs are composed of exosomes, microparticles and apoptotic bodies. Its benefits pass through biofilms and are not degraded by various enzymes, so it can be used as a biomarker in potential diseases and has attracted much attention from researchers. Current studies have found that EVs are involved in the development of various cardiovascular diseases (CVD), such as heart failure and myocardial ischemia-reperfusion injury. In addition, stem cell-derived EVs play an important role in the diagnosis and treatment of a variety of CVD. In this review, we present the biological features of EVs, the role of EVs in various CVD, and the challenges they encounter in the treatment of CVD.
Collapse
Affiliation(s)
- Hanbin Li
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Lu Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Hongxin Cheng
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Qing Zhang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Shiqi Wang
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Wen Zhong
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Chengqi He
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| | - Quan Wei
- Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China HospitalSichuan UniversityChengduChina
- Key Laboratory of Rehabilitation Medicine in Sichuan Province, West China HospitalSichuan UniversityChengduChina
| |
Collapse
|
34
|
Weeks KL, Bernardo BC. Adeno-Associated Viruses as Gene Delivery Tools for Diabetic Heart Disease and Failure: Key Considerations for Clinicians and Preclinical Researchers. Heart Lung Circ 2025; 34:118-124. [PMID: 39818494 DOI: 10.1016/j.hlc.2024.11.021] [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: 09/19/2024] [Revised: 11/13/2024] [Accepted: 11/21/2024] [Indexed: 01/18/2025]
Abstract
Diabetes is becoming more common worldwide, and people with diabetes are twice as likely to experience heart problems compared to those without diabetes. These cardiovascular complications are the foremost cause of mortality among people with diabetes. A specific form of heart failure known as "diabetic cardiomyopathy" can develop in individuals with diabetes. There are no treatments specifically approved for diabetic cardiomyopathy. Ongoing research is exploring innovative treatments, including the development of gene therapy techniques (e.g., adeno-associated viral vectors) designed to target specific molecular pathways affected in the disease. Here, we discuss the progress, challenges, and experimental considerations of gene therapy for the diabetic heart.
Collapse
Affiliation(s)
- Kate L Weeks
- Department of Anatomy & Physiology, University of Melbourne, Vic, Australia; Baker Department of Cardiometabolic Health, University of Melbourne, Vic, Australia; Central Clinical School, Monash University, Melbourne, Vic, Australia.
| | - Bianca C Bernardo
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Burwood, Vic, Australia; Department of Paediatrics, University of Melbourne, Parkville, Vic, Australia.
| |
Collapse
|
35
|
Habibi A, Ruf W, Schurgers L. Protease-activated receptors in vascular smooth muscle cells: a bridge between thrombo-inflammation and vascular remodelling. Cell Commun Signal 2025; 23:57. [PMID: 39891111 PMCID: PMC11786455 DOI: 10.1186/s12964-025-02066-6] [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: 12/10/2024] [Accepted: 01/25/2025] [Indexed: 02/03/2025] Open
Abstract
Coagulation factors are responsible for blood clot formation yet have also non-canonical functions as signalling molecules. In this context, they can activate protease-activated receptors (PARs) ubiquitously expressed in the vasculature. During vascular repair, vascular smooth muscle cells (VSMCs) will switch from a contractile to a synthetic reparative phenotype. During prolonged vascular stress, VSMCs acquire a pathological phenotype leading to cardiovascular disease. Activated coagulation factors impact on vessel wall permeability and integrity after vascular injury with a key role for PAR activation on endothelial cells. The activation of PARs on VSMCs supports vessel wall repair following injury. Prolonged PAR activation, however, results in pathological vascular remodelling. Therefore, understanding the mechanisms of PAR activation on VSMCs is key to propel our understanding of the molecular and cellular mechanisms to develop novel therapeutic strategies to resolve vascular remodelling.In this review, we discuss recent advances on the role of PAR signalling on VSMCs and specifically their role in vascular remodelling contributing to cardiovascular disease. Additionally, we discuss current therapeutic strategies targeting PAR signalling - indirectly or directly - in relation to cardiovascular disease.
Collapse
Affiliation(s)
- Anxhela Habibi
- Department of Biochemistry, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands.
- Center for Thrombosis and Hemostasis, Johannes-Gutenberg-University Medical Center Mainz, Mainz, Germany.
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, Johannes-Gutenberg-University Medical Center Mainz, Mainz, Germany
| | - Leon Schurgers
- Department of Biochemistry, CARIM, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
36
|
Orefice NS, Petrillo G, Pignataro C, Mascolo M, De Luca G, Verde S, Pentimalli F, Condorelli G, Quintavalle C. Extracellular vesicles and microRNAs in cancer progression. Adv Clin Chem 2025; 125:23-54. [PMID: 39988407 DOI: 10.1016/bs.acc.2024.11.004] [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: 02/25/2025]
Abstract
Extracellular vesicles (EVs) have emerged as critical mediators of intercellular communication in cancer. These membranous structures, secreted by normal and cancerous cells, carry a cargo of bioactive molecules including microRNAs (miRNAs) that modulate various cellular processes. miRNAs are small non-coding RNAs that play pivotal roles in post-transcriptional gene regulation and have been implicated in cancer initiation, progression, and metastasis. In cancer, tumor-derived EVs transport specific miRNAs to recipient cells, modulating tumorigenesis, growth, angiogenesis, and metastasis. Dysregulation of miRNA expression profiles within EVs contributes to the acquisition of cancer hallmarks that include increased proliferation, survival, and migration. EV miRNAs influence the tumor microenvironment, promoting immune evasion, remodeling the extracellular matrix, and establishing pre-metastatic niches. Understanding the complex interplay between EVs, miRNAs, and cancer holds significant promise for developing novel diagnostic and therapeutic strategies. This chapter provides insights into the role of EV-mediated miRNA signaling in cancer pathogenesis, highlighting its potential as a biomarker for cancer detection, prognosis, and treatment response assessment.
Collapse
Affiliation(s)
- Nicola S Orefice
- Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| | - Gianluca Petrillo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Claudia Pignataro
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Martina Mascolo
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy
| | - Giada De Luca
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy
| | - Sara Verde
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy; Aka biotech S.r.l., Napoli, Italy
| | - Francesca Pentimalli
- Department of Medicine and Surgery, LUM University "Giuseppe DeGennaro", Bari, Italy
| | - Gerolama Condorelli
- Department of Molecular Medicine and Medical Biotechnology, Federico II University of Naples, Naples, Italy; Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy.
| | - Cristina Quintavalle
- Institute of Endotypes in Oncology, Metabolism and Immunology "G. Salvatore" (IEOMI) National Research Council (CNR), Naples, Italy
| |
Collapse
|
37
|
Kasiński D, Szeliski K, Drewa T, Pokrywczyńska M. Extracellular vesicles-a new player in the development of urinary bladder cancer. Ther Adv Med Oncol 2025; 17:17588359241297529. [PMID: 39850919 PMCID: PMC11755519 DOI: 10.1177/17588359241297529] [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: 05/06/2024] [Accepted: 10/18/2024] [Indexed: 01/25/2025] Open
Abstract
Bladder cancer was the 10th most commonly diagnosed cancer worldwide in 2020. Extracellular vesicles (EVs) are nano-sized membranous structures secreted by all types of cells into the extracellular space. EVs can transport proteins, lipids, or nucleic acids to specific target cells. What brings more attention and potential implications is the fact that cancer cells secrete more EVs than non-malignant cells. EVs are widely studied for their role in cancer development. This publication summarizes the impact of EVs secreted by urinary bladder cancer cells on urinary bladder cancer development and metastasis. EVs isolated from urinary bladder cancer cells affect other lower-grade cancer cells or normal cells by inducing different metabolic pathways (transforming growth factor β/Smads pathway; phosphoinositide 3-kinase/Akt pathway) that promote epithelial-mesenchymal transition. The cargo carried by EVs can also induce angiogenesis, another critical element in the development of bladder cancer, and modulate the immune system response in a tumor-beneficial manner. In summary, the transfer of substances produced by tumor cells via EVs to the environment influences many stages of tumor progression. An in-depth understanding of the role EVs play in the development of urinary bladder cancer is crucial for the development of future anticancer therapies.
Collapse
Affiliation(s)
- Damian Kasiński
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Jagiellońska 13/15, 85-067 Bydgoszcz, Poland
| | - Kamil Szeliski
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Tomasz Drewa
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| | - Marta Pokrywczyńska
- Chair of Urology and Andrology, Department of Regenerative Medicine, Collegium Medicum, Nicolaus Copernicus University, Bydgoszcz, Poland
| |
Collapse
|
38
|
Birjandi AA, Sharpe P. The Secretome of the Inductive Tooth Germ Exhibits Signals Required for Tooth Development. Bioengineering (Basel) 2025; 12:96. [PMID: 40001617 PMCID: PMC11851894 DOI: 10.3390/bioengineering12020096] [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: 11/21/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 02/27/2025] Open
Abstract
Teeth develop from reciprocal signaling between inductive and receptive cells. The inductive signals for tooth development are initially in the epithelium of the developing branchial arch, but later shift to the underlying mesenchyme of a developing tooth germ. The inductive signals that are needed for tooth development have not yet been fully identified. Our lab previously provided a basis for bioengineering new teeth by separating the tooth germ's epithelial and mesenchyme cells into a single cell population and recombing them. This approach, however, is not clinically applicable as the cells lose their inductive ability when expanded in vitro. In this study, we investigate whether the secretome and small extracellular vehicles (sEV) derived from inductive tooth germ mesenchyme can contribute to inductive signals required for tooth development. To address this, small extracellular vesicles and secretome were purified from inductive tooth germ mesenchyme and characterized. We investigated the proteome of sEV and proteome of inductive tooth germ mesenchyme and the impact of the culture condition and duration on the proteome. Additionally, we investigated the transcriptomic changes in tooth germ epithelium after treatment with sEV from inductive tooth germ mesenchyme. We show that culture duration of inductive tooth germ mesenchyme has an impact on the proteome of sEV purified from these cells. Similarly, culturing these cells in 2D and 3D environments results in different protein content. Proteome unique to sEV derived from inductive shows an association with multiple signaling pathways related to tooth development. Our RNASeq results show that treatment of tooth germ epithelial cells with small extracellular vesicles derived from inductive tooth germ mesenchyme results in an increased expression of some of the known odontogenic genes. Whilst further analysis is required to harvest the full potential of these sEV, our results suggests that extracellular vehicles contribute to signals required during tooth development, potentially through modulation of cellular metabolism and ECM organization.
Collapse
Affiliation(s)
| | - Paul Sharpe
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
| |
Collapse
|
39
|
Badawy AA, El-Hofey SM, Shaban AM, Orif SE, Uyanıkgil Y, El-Magd MA. Camel milk extracellular vesicles/exosomes: a fascinating frontier in isolation and therapeutic potential. Food Funct 2025; 16:344-365. [PMID: 39714264 DOI: 10.1039/d4fo04331f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Camel milk has a unique composition that sets it apart from other types of animal milk, which has captured the interest of medical and scientific communities. Extracellular vesicles (EVs) mainly contain exosomes (Exos, 30-200 nm) and microvesicles (MVs, 200-1000 nm). Camel milk EVs, particularly Exos, which we named EVs/Exos, have arisen as a fascinating area of scientific inquiry, holding enormous potential for the future of biomedicine due to their anticancer, antibacterial, antidiabetic nephropathy, and immunostimulatory impacts. Camel milk EVs/Exos affect the antioxidant status and oxidative stress differently depending on the target cells. They boosted ROS in cancer cells but improved the antioxidant state in healthy cells. Camel milk EVs/Exos have distinct exosomal lactoferrin and kappa casein mRNAs, which could be responsible for their anticancer and immunomodulatory effects. Due to the high fat content of milk, there is a lack of established protocols for the precise isolation of EVs/Exos from milk, despite the increasing interest in this area of study. This review highlighted the techniques employed for milk EV/Exo isolation and characterization, acknowledging the challenges faced by researchers and the latest advancements in overcoming these hurdles. This review also detailed the potential of camel milk EVs/Exos in therapeutic applications. This comprehensive analysis positions camel milk EVs/Exos at the forefront of scientific inquiry, paving the way for groundbreaking discoveries in the years to come.
Collapse
Affiliation(s)
- Abdelnaser A Badawy
- Biochemistry Department, Faculty of Medicine, Northern Border University, Arar City, Saudi Arabia
| | - Salma M El-Hofey
- Department of Anatomy & Embryology, Faculty of Veterinary Medicine, Kafrelsheikh University, Egypt.
| | - Amira M Shaban
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University 62511, Egypt
| | - Sahar E Orif
- Department of Anatomy & Embryology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
- Department of Stem Cells, Institute of Health Sciences, Ege University, İzmir, Türkiye
| | - Yiğit Uyanıkgil
- Department of Stem Cells, Institute of Health Sciences, Ege University, İzmir, Türkiye
- Cord Blood Cell - Tissue Research and Application Center, Ege University, İzmir, 35100, Turkiye
| | - Mohammed A El-Magd
- Department of Anatomy & Embryology, Faculty of Veterinary Medicine, Kafrelsheikh University, Egypt.
| |
Collapse
|
40
|
Yi J, Kim S, Lim M, Jeong H, Han C, Cho S, Park J. Size-based separation of extracellular vesicles investigating the relationship between Tetraspanins and RNA. Anal Chim Acta 2025; 1335:343421. [PMID: 39643292 DOI: 10.1016/j.aca.2024.343421] [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: 08/23/2024] [Revised: 11/01/2024] [Accepted: 11/12/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Extracellular vesicles (EVs), nano-sized particles released by cells, exhibit inherent heterogeneity, posing challenges for precise classification. This study introduces a method utilizing the coffee ring effect for size-based separation analysis, driven by the outward flow during droplet evaporation with Marangoni flow resulting from a surface tension gradient. RESULTS The controlled separation of nanoparticles based on size was applied to characterize EV and virus-like particle (VLP) samples. Tetraspanin markers exhibited distinct distribution patterns in dried droplets, with CD63-single-positive particles being smaller than those positive for CD9, CD81, or gag. Additionally, using previously developed fluorescence nanoparticle tracking analysis and transmission electron microscopy, single vesicle analysis validated the size disparities in CD-positive particles. RNA analysis through the coffee ring effect and fluorescent NTA revealed differential patterns in EVs and VLPs, providing different insights into RNA packaging. SIGNIFICANCE This multifaceted approach enhances understanding EV heterogeneity, emphasizing the potential influence of cellular origin and biogenesis pathways on particle characteristics.
Collapse
Affiliation(s)
- Johan Yi
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeong-buk, 37673, Republic of Korea
| | - Suyeon Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeong-buk, 37673, Republic of Korea
| | - Minyeob Lim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeong-buk, 37673, Republic of Korea
| | - Hwapyeong Jeong
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeong-buk, 37673, Republic of Korea
| | - Chungmin Han
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeong-buk, 37673, Republic of Korea
| | - Siwoo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeong-buk, 37673, Republic of Korea
| | - Jaesung Park
- Department of Mechanical Engineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeong-buk, 37673, Republic of Korea; School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology, 77 Cheongam-Ro, Nam-Gu, Pohang, Gyeong-buk, 37673, Republic of Korea.
| |
Collapse
|
41
|
Afzal M, Hameed H, Paiva-Santos AC, Saleem M, Hameed A, Ahmad SM. Bioengineered exosomes: Cellular membrane-camouflaged biomimetic nanocarriers for Parkinson's disease management. Eur J Pharmacol 2025; 987:177199. [PMID: 39662659 DOI: 10.1016/j.ejphar.2024.177199] [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: 09/16/2024] [Revised: 11/21/2024] [Accepted: 12/09/2024] [Indexed: 12/13/2024]
Abstract
Parkinson's disease is a prevalent neurological condition that affects around 1% of adults over 60 worldwide. Deep brain stimulation and dopamine replacement therapy are common therapies for Parkinson's disease, yet they are unable to reverse the disease it simply because of the blood brain barrier. The use of bioengineered exosomes to treat Parkinson's disease is being studied because they have the ability to cross the blood-brain barrier. Their natural ability to cross the blood-brain barrier (BBB) and their biocompatibility make them highly suitable for delivering therapeutic agents to manage PD, specifically the role of astrocytes, microglial cells, and alpha-synuclein. It also explores the biogenesis and preparation of these bioengineered exosomes. In comparison to conventional nanocarriers, the modified exosomal-membrane-camouflaged abiotic nanocarriers show improved resilience and compatibility. Improved cellular absorption and targeted delivery of therapeutic payloads, such as medications and enzymes, are being shown in laboratory trials. A viable strategy for treating PD involves combining abiotic nanocarriers with bioengineered exosomal membranes. Despite their promising potential, successful clinical application requires overcoming hurdles related to scalable production, regulatory approval, and long-term safety evaluation. Nevertheless, the innovative use of bioengineered exosomes holds significant promise for advancing PD management and improving patient outcomes through more targeted and effective therapeutic strategies.
Collapse
Affiliation(s)
- Maham Afzal
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Huma Hameed
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal; REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal.
| | - Makkia Saleem
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan.
| | - Anam Hameed
- Department of Human Nutrition and Dietetics, Faculty of Rehabilitation and Allied Health Sciences, Riphah International University, Gulberg III, Lahore, 54000, Pakistan.
| | - Syed Muhammad Ahmad
- Faculty of Pharmaceutical Sciences, University of Central Punjab (UCP), Lahore, 54000, Pakistan.
| |
Collapse
|
42
|
Zhang R, Liao W, Chen X, Wang J, Li J, Chen G, Wu W, Wang X, Zhang Y, Chen Z, Zhu X, Lin Z, Zhu Y, Ma L, Yu H. PKCα regulates the secretion of PDL1-carrying small extracellular vesicles in a p53-dependent manner. Cell Death Dis 2025; 16:19. [PMID: 39809736 PMCID: PMC11733117 DOI: 10.1038/s41419-025-07341-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 12/06/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025]
Abstract
Small extracellular vesicles (sEVs), carrying PD-L1, have been implicated in immune evasion and tumor progression. However, understanding how PD-L1 sEVs are secreted still needs to be improved. We found that the secretion dynamics of PD-L1 sEVs is similar to that of other sEVs. Intracellular calcium and the associated downstream PKC signaling plays pivotal roles in releasing PD-L1 sEVs in non-small cell lung cancer cells (NSCLC). Particularly, we observed that knocking down PKCα has profound impacts on PD-L1 sEVs secretion, especially in the resting state and in the activated state, when induced by an intracellular calcium rise. Furthermore, our study revealed that PKCα regulates PD-L1 expression and PD-L1 sEVs secretion by influencing STAT1 phosphorylation and nuclear translocation in a p53-dependent manner. Notably, p53 can regulate STAT1 phosphorylation and nuclear localization, but it does not affect PKCα expression. This suggests that PKCα plays a significant role in regulating PD-L1 expression. Our findings suggest that targeting PKCα to modulate PD-L1 dynamics in NSCLC may be a promising therapeutic strategy to enhance the efficacy of immunotherapeutic interventions.
Collapse
Grants
- Macau Science and Technology Development Fund, Macau, China, Project code 0062/2021/A2, 002/2023/ALC, 003/2022/ALC & 006/2023/SKL
- Macau Science and Technology Development Fund, Macau, China, Project code 003/2022/ALC
- Macau Science and Technology Development Fund, Macau, China, Project code 0062/2021/A2, 002/2023/ALC & 006/2023/SKL
Collapse
Affiliation(s)
- Ren Zhang
- School of Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
- 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
| | - Weilin Liao
- 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
| | - Xi Chen
- 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
- Guangdong Second Provincial General Hospital, Postdoctoral Research Station of Basic Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Junyi 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
| | - Jiaqi Li
- 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
| | - Geer Chen
- 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
| | - Weiyu 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
| | - Xiaoxuan 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
| | - Yao 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
| | - Ziyu Chen
- 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
| | - Xiaoyu Zhu
- 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
| | - Zicong Lin
- 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
| | - Yizhun Zhu
- School of Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China
| | - Lijuan Ma
- School of Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
- 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.
| | - Haijie Yu
- School of Pharmacy, Faculty of Medicine & State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau, China.
- 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.
| |
Collapse
|
43
|
Dogan AB, Marsh SR, Tschetter RJ, E Beard C, Amin MR, Jane Jourdan L, Gourdie RG. Stabilizing milk-derived extracellular vesicles (mEVs) through lyophilization: a novel trehalose and tryptophan formulation for maintaining structure and Bioactivity during long-term storage. J Biol Eng 2025; 19:4. [PMID: 39806456 PMCID: PMC11727230 DOI: 10.1186/s13036-024-00470-z] [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: 08/26/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Extracellular vesicles (EVs) are widely investigated for their implications in cell-cell signaling, immune modulation, disease pathogenesis, cancer, regenerative medicine, and as a potential drug delivery vector. However, maintaining integrity and bioactivity of EVs between Good Manufacturing Practice separation/filtration and end-user application remains a consistent bottleneck towards commercialization. Milk-derived extracellular vesicles (mEVs), separated from bovine milk, could provide a relatively low-cost, scalable platform for large-scale mEV production; however, the reliance on cold supply chain for storage remains a logistical and financial burden for biologics that are unstable at room temperature. Herein, we aim to characterize and engineer a freeze-dried, mEV formulation that can be stored at room temperature without sacrificing structure/bioactivity and can be reconstituted before delivery. In addition to undertaking established mEV assays of structure and function on our preparations, we introduce a novel, efficient, high throughput assay of mEV bioactivity based on Electric Cell Substrate Impedance Sensing (ECIS) in Human dermal fibroblast monolayers. By adding appropriate excipients, such as trehalose and tryptophan, we describe a protective formulation that preserves mEV bioactivity during long-term, room temperature storage. Our identification of the efficacy of tryptophan as a novel additive to mEV lyophilization solutions could represent a significant advancement in stabilizing small extracellular vesicles outside of cold storage conditions.
Collapse
Affiliation(s)
- Alan B Dogan
- Virginia Tech Carilion School of Medicine, Roanoke, VA, 24016, USA
| | - Spencer R Marsh
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA
| | - Rachel J Tschetter
- Materials Science and Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Claire E Beard
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
| | - Md R Amin
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
- Translational Biology, Medicine, and Health graduate program at Virginia Tech, Roanoke, VA, 24016, USA
| | - L Jane Jourdan
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA
| | - Robert G Gourdie
- Fralin Biomedical Research Institute at Virginia Tech Carilion, Roanoke, VA, 24016, USA.
- Center for Vascular and Heart Research, Virginia Tech, Roanoke, VA, 24016, USA.
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, 24061, USA.
- Department of Emergency Medicine, Virginia Tech Carilion School of Medicine, Virginia Tech, Roanoke, VA, 24016, USA.
- Faculty of Health Science, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
44
|
Silva RO, Haddad M, Counil H, Zaouter C, Patten SA, Fulop T, Ramassamy C. Exploring the potential of plasma and adipose mesenchymal stem cell-derived extracellular vesicles as novel platforms for neuroinflammation therapy. J Control Release 2025; 377:880-898. [PMID: 39617173 DOI: 10.1016/j.jconrel.2024.11.060] [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: 09/10/2024] [Revised: 11/10/2024] [Accepted: 11/22/2024] [Indexed: 12/10/2024]
Abstract
Persistent reactive oxygen species (ROS) and neuroinflammation contribute to the onset and progression of neurodegenerative diseases, underscoring the need for targeted therapeutic strategies to mitigate these effects. Extracellular vesicles (EVs) show promise in drug delivery due to their biocompatibility, ability to cross biological barriers, and specific interactions with cell and tissue receptors. In this study, we demonstrated that human plasma-derived EVs (pEVs) exhibit higher brain-targeting specificity, while adipose-derived mesenchymal stem cells EVs (ADMSC-EVs) offer regenerative and immunomodulatory properties. We further investigated the potential of these EVs as therapeutic carriers for brain-targeted drug delivery, using Donepezil (DNZ) as the model drug. DNZ, a cholinesterase inhibitor commonly used for Alzheimer's disease (AD), also has neuroprotective and anti-inflammatory properties. The size of EVs used ranged from 50 to 300 nm with a surface charge below -30 mV. Both formulations showed rapid cellular internalization, without toxicity, and the ability to cross the blood-brain barrier (BBB) in a zebrafish model. The have analyzed the anti-inflammatory and antioxidant actions of pEVs-DNZ and ADMSC-EVs-DNZ in the presence of lipopolysaccharide (LPS). ADMSC-EVs significantly reduced the inflammatory mediators released by HMC3 microglial cells while treatment with pEVs-DNZ and ADMSC-EVs-DNZ lowered both phagocytic activity and ROS levels in these cells. In vivo experiments using zebrafish larvae revealed that both EV formulations reduced microglial proliferation and exhibited antioxidant effects. Overall, this study highlights the potential of EVs loaded with DNZ as a novel approach for treating neuroinflammation underlying various neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Mohamed Haddad
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
| | - Hermine Counil
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
| | - Charlotte Zaouter
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
| | - Shunmoogum A Patten
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boul des Prairies, Laval, QC H7V 1B7, Canada
| | - Tamas Fulop
- Research Center on Aging, Faculty of Medicine and Health Sciences, University Sherbrooke, Sherbrooke, QC J1H 4N4, Canada
| | - Charles Ramassamy
- INRS, Centre Armand-Frappier Santé Biotechnologie, 531 Boul des Prairies, Laval, QC H7V 1B7, Canada.
| |
Collapse
|
45
|
Hu H, Wang X, Yu H, Wang Z. Extracellular vesicular microRNAs and cardiac hypertrophy. Front Endocrinol (Lausanne) 2025; 15:1444940. [PMID: 39850481 PMCID: PMC11753959 DOI: 10.3389/fendo.2024.1444940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
Cardiac hypertrophy is an adaptive response to pressure or volume overload such as hypertension and ischemic heart diseases. Sustained cardiac hypertrophy eventually leads to heart failure. The pathophysiological alterations of hypertrophy are complex, involving both cellular and molecular systems. Understanding the molecular events that inhibit or repress cardiac hypertrophy may help identify novel therapeutic strategies. Increasing evidence has indicated that extracellular vesicle (EV)-derived microRNAs (miRNAs) play a significant role in the development and progression of cardiac hypertrophy. In this review, we briefly review recent advancements in EV research, especially on biogenesis, cargoes and its role in cardiac hypertrophy. We then describe the latest findings regarding EV-derived miRNAs, highlighting their functions and regulatory mechanisms in cardiac hypertrophy. Finally, the potential role of EV-derived miRNAs as targets in the diagnosis and treatment of cardiac hypertrophy will be discussed.
Collapse
Affiliation(s)
- Hai Hu
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- School of Basic Medicine, Baotou Medical College, Baotou, China
| | - Xiulian Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| | - Hui Yu
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
- School of Basic Medicine, Baotou Medical College, Baotou, China
| | - Zhanli Wang
- Inner Mongolia Key Laboratory of Disease-Related Biomarkers, The Second Affiliated Hospital, Baotou Medical College, Baotou, China
| |
Collapse
|
46
|
Su N, Zhang J, Liu W, Zheng H, Li M, Zhao J, Gao M, Zhang X. Specific isolation and quantification of PD-L1 positive tumor derived exosomes for accurate breast cancer discrimination via aptamer-functionalized magnetic composites and SERS immunoassay. Talanta 2025; 281:126956. [PMID: 39332044 DOI: 10.1016/j.talanta.2024.126956] [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: 08/09/2024] [Revised: 09/17/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
PD-L1 positive tumor derived exosomes (TEXsPD-L1) play a significant role in disease progression, tumor metastasis and cancer immunotherapy. However, the overlap of PD-L1 between TEXs and non-tumor derived exosomes (non-TEXs) restricts the specific isolation and quantification of TEXPD-L1 from clinical samples. Herein, a new aptamer-functionalized and hydrophilic immunomagnetic substrate was designed by decorating generation 5 polyamidoamine dendrimers (G5 PAMAM), zwitterionic trimethylamine N-oxide (TMAO) and EpCAM (Epithelial cell adhesion molecule) aptamers on magnetic cores sequentially (Fe3O4@PAMAM@TMAO@Aptamer, named as FPTA) for rapid target and efficient capture of TEXs. The FPTA substrate gathered excellent characters of strong magnetic responsiveness of Fe3O4, abundant affinity sites of PAMAM, strong hydrophilicity of TMAO and enhanced affinity properties of EpCAM aptamers. Because of these advantages, FPTA can isolate TEXs quickly within 30min with high capture efficiency of 90.5 % ± 3.0 % and low nonspecific absorption of 8.2 % ± 2.0 % for non-TEXs. Furthermore, PD-L1 (Programmed cell death-ligand 1) positive TEXs (TEXsPD-L1) from the captured TEXs were recognized and quantitatively analyzed by utilizing SERS (surface-enhanced Raman spectroscopy) reporter molecules 4-NTP (4-Nitrothiophenol) on PD-L1 aptamers-functionalized gold immunoaffinity probe. The signal of TEXsPD-L1 was converted to SERS signal of 4-NTP at 1344 cm-1 which exhibited a linear correlation to concentration of TEXsPD-L1(R2 = 0.9905). With these merits, this strategy was further applied to clinical plasma samples from breast cancer (BC) patients and healthy controls (HC), exhibited an excellent diagnosis accuracy with area under curve (AUC) of receiver operating characteristic (ROC) curve reaching 0.988. All these results demonstrate that the FPTA immunomagnetic substrate combined with SERS immunoaffinity probe may become a generic tool for specific isolation and quantitative analysis of PD-L1 positive tumor-derived exosomes in clinics.
Collapse
Affiliation(s)
- Ning Su
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Jin Zhang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Wei Liu
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Haoyang Zheng
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Mengran Li
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| | - Jiandong Zhao
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Mingxia Gao
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China.
| | - Xiangmin Zhang
- Department of Chemistry and Institutes of Biomedical Sciences, Fudan University, Shanghai, 200433, China
| |
Collapse
|
47
|
Selvaraj S, Weerasinghe L. The Role of Nanotechnology in Understanding the Pathophysiology of Traumatic Brain Injury. Cent Nerv Syst Agents Med Chem 2025; 25:20-38. [PMID: 38676493 DOI: 10.2174/0118715249291999240418112531] [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: 11/09/2023] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/29/2024]
Abstract
Recently, traumatic brain injury (TBI) has been a growing disorder due to frequent brain dysfunction. The Glasgow Coma Scale expresses TBI as classified as having mild, moderate, or severe brain effects, according to the effects on the brain. Brain receptors undergo various modifications in their pathology through chemical synaptic pathways, leading to depression, Alzheimer's, and Parkinson's disease. These brain disorders can be controlled using central receptors such as dopamine, glutamate, and γ-aminobutyric acid, which are clearly explained in this review. Furthermore, there are many complications in TBI's clinical trials and diagnostics, leading to insignificant treatment, causing permanent neuro-damage, physical disability, and even death. Bio-screening and conventional molecular-based therapies are inappropriate due to poor preclinical testing and delayed recovery. Hence, modern nanotechnology utilizing nanopulsed laser therapy and advanced nanoparticle insertion will be suitable for TBI's diagnostics and treatment. In recent days, nanotechnology has an important role in TBI control and provides a higher success rate than conventional therapies. This review highlights the pathophysiology of TBI by comprising the drawbacks of conventional techniques and supports suitable modern alternates for treating TBI.
Collapse
Affiliation(s)
- Saranya Selvaraj
- Department of Chemistry, Faculty of Applied sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| | - Laksiri Weerasinghe
- Department of Chemistry, Faculty of Applied sciences, University of Sri Jayewardenepura, Gangodawila, Nugegoda, Sri Lanka
| |
Collapse
|
48
|
Dong JC, Liao Y, Zhou W, Sun MJ, Zhang HY, Li Y, Song ZC. Porphyromonas gingivalis LPS-stimulated BMSC-derived exosome promotes osteoclastogenesis via miR-151-3p/PAFAH1B1. Oral Dis 2025; 31:206-216. [PMID: 38923332 DOI: 10.1111/odi.15031] [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: 11/28/2023] [Revised: 05/06/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024]
Abstract
OBJECTIVES Porphyromonas gingivalis-LPS regulated bone metabolism by triggering dysfunction of osteoblasts directly, and affecting activity of osteoclasts through intracellular communication. Exosome, as the mediator of intercellular communication, was important vesicle to regulate osteogenesis and osteoclastogenesis. This research was designed for investigating the mechanism of BMSCs-EXO in modulating osteoclastic activity under the P. gingivalis-LPS. MATERIALS AND METHODS The cytotoxicity and osteogenic effects of P. gingivalis-LPS on BMSCs was evaluated, and then osteoclastic activity of RAW264.7 co-cultured with exosomes was detected. Besides, Affymetrix miRNA array and luciferase reporter assay were used to identify the target exosomal miRNA signal pathway. RESULTS BMSCs' osteogenic differentiation and proliferation were decreased under 1 and 10 μg/mL P. gingivalis-LPS. Osteoclastic-related genes and proteins levels were promoted by P. gingivalis-LPS-stimulated BMSCs-EXO. Based on the miRNA microarray analysis, exosomal miR-151-3p was lessened in BMExo-LPS group, which facilitated osteoclastic differentiation through miR-151-3p/PAFAH1B1. CONCLUSIONS Porphyromonas gingivalis-LPS could regulated bone metabolism by inhibiting proliferation and osteogenesis of BMSCs directly. Also, P. gingivalis-LPS-stimulated BMSCs-EXO promoted osteoclastogenesis via activating miR-151-3p/PAFAH1B1 signal pathway.
Collapse
Affiliation(s)
- Jia-Chen Dong
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yue Liao
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Wei Zhou
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Meng-Jun Sun
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Huan-Yu Zhang
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| | - Yan Li
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
- Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, China
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, China
| | - Zhong-Chen Song
- Department of Periodontology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, China
| |
Collapse
|
49
|
Liu ZX, Chen G, Yu ZL. Advances in subpopulation separation and detection of extracellular vesicles: for liquid biopsy and downstream research. Theranostics 2025; 15:1135-1155. [PMID: 39776815 PMCID: PMC11700854 DOI: 10.7150/thno.106459] [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: 11/06/2024] [Accepted: 11/27/2024] [Indexed: 01/11/2025] Open
Abstract
Extracellular vesicles (EVs) are carriers of a diverse array of bioactive molecules, making them valuable clinical tools for liquid biopsy in disease diagnosis and prognosis evaluation. These molecules play critical roles in various physiological and pathological conditions, and effective separation of EVs is essential to achieve these objectives. Due to the high heterogeneity of EVs, particularly with regard to their cargo molecules, merely isolating the general EV population is inadequate for liquid biopsy and biological function studies. Therefore, separating EV subpopulations becomes crucial. Traditional separation methods, such as differential ultracentrifugation and size exclusion chromatography, along with burgeoning techniques like classical microfluidic chips and covalent chemistry, often prove time-consuming, yield low purity, and have limited ability to address cargo heterogeneity. Thus, precise separation of EV subpopulations is of utmost importance. Additionally, detecting subpopulation-specific cargo is vital for validating the effectiveness of separation methods and supporting clinical biopsy applications. However, reviews that focus specifically on detection methods for EV subpopulations are limited. This paper provides a comprehensive overview of the methods for separating and detecting EV subpopulations with surface marker heterogeneity, comparing the advantages and limitations of each technique. Furthermore, it discusses challenges and future prospects for these methods in the context of liquid biopsy and downstream research. Collectively, this review aims to offer innovative insights into the separation and detection of EV subpopulations, guiding researchers to avoid common pitfalls and refine their investigative approaches.
Collapse
Affiliation(s)
- Zi-Xiu Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Gang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, China
- Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
50
|
Lihon MV, Tuchscherer NA, Tao WA. Isolation and Identification of Brain Tissue Extracellular Vesicles for Translational Proteomics. Methods Mol Biol 2025; 2884:225-239. [PMID: 39716007 DOI: 10.1007/978-1-0716-4298-6_15] [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: 12/25/2024]
Abstract
Extracellular vesicles (EVs) are small membrane-bound structures that play important roles in intercellular communication and the transfer of biomolecules between cells. EVs have become a topic of interest for research in translational proteomics for disease biomarker discovery due to their ability to reflect changes in the cellular proteome, including diseases affecting the brain. Utilizing the proteome analysis of EVs to its fullest potential requires proper isolation and purity. In this chapter, we describe a detailed method for the isolation and identification of brain tissue EVs for translational proteomics using our in-house chemical affinity magnetic bead-based (non-antibody) method, the EVtrap. We also discuss various methods for quantification, characterization, and functional analysis of isolated brain tissue EVs, including western blotting, and proteomic profiling of post-translational modifications (PTMs) involved in neurodegenerative diseases, such as protein N-terminal acetylation. This protocol provides a valuable resource for studies conducted on brain tissue EVs and their potential as biomarkers and therapeutic targets for neurological diseases.
Collapse
Affiliation(s)
- Michelle V Lihon
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA
| | | | - W Andy Tao
- Department of Biochemistry, Purdue University, West Lafayette, IN, USA.
- Department of Chemistry, Purdue University, West Lafayette, IN, USA.
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, USA.
- Purdue Institute for Cancer Research, Purdue University, West Lafayette, IN, USA.
- Tymora Analytical Operations, West Lafayette, IN, USA.
| |
Collapse
|