1
|
Arthur P, Kandoi S, Kalvala A, Boirie B, Nathani A, Aare M, Bhattacharya S, Kulkarni T, Sun L, Lamba DA, Li Y, Singh M. Cannabidiol-Loaded Retinal Organoid-Derived Extracellular Vesicles Protect Oxidatively Stressed ARPE-19 Cells. Biomedicines 2025; 13:1167. [PMID: 40426994 DOI: 10.3390/biomedicines13051167] [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: 04/01/2025] [Revised: 05/03/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
Background/Objectives: Age-related macular degeneration (AMD) is the third leading cause of irreversible blindness in elderly individuals aged over 50 years old. Oxidative stress plays a crucial role in the etiopathogenesis of multifactorial AMD disease. The phospholipid bilayer EVs derived from the culture-conditioned medium of human induced pluripotent stem cell (hiPSC) differentiated retinal organoids aid in cell-to-cell communication, signaling, and extracellular matrix remodeling. The goal of the current study is to establish and evaluate the encapsulation of a hydrophobic compound, cannabidiol (CBD), into retinal organoid-derived extracellular vesicles (EVs) for potential therapeutic use in AMD. Methods: hiPSC-derived retinal organoid EVs were encapsulated with CBD via sonication (CBD-EVs), and structural features were elucidated using atomic force microscopy, nanoparticle tracking analysis, and small/microRNA (miRNA) sequencing. ARPE-19 cells and oxidative-stressed (H2O2) ARPE-19 cells treated with CBD-EVs were assessed for cytotoxicity, apoptosis (MTT assay), reactive oxygen species (DCFDA), and antioxidant proteins (immunohistochemistry and Western blot). Results: Distinct miRNA cargo were identified in early and late retinal organoid-derived EVs, implicating their roles in retinal development, differentiation, and functionality. The therapeutic effects of CBD-loaded EVs on oxidative-stressed ARPE-19 cells showed greater viability, decreased ROS production, downregulated expression of inflammation- and apoptosis-related proteins, and upregulated expression of antioxidants by Western blot and immunocytochemistry. Conclusions: miRNAs are both prognostic and predictive biomarkers and can be a target for developing therapy since they regulate RPE physiology and diseases. Our findings indicate that CBD-EVs could potentially alleviate the course of AMD by activating the targeted proteins linked to the adenosine monophosphate kinase (AMPK) pathway. Implicating the use of CBD-EVs represents a novel frontline to promote long-term abstinence from drugs and pharmacotherapy development in treating AMD.
Collapse
Affiliation(s)
- Peggy Arthur
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Sangeetha Kandoi
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Anil Kalvala
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Breana Boirie
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Aakash Nathani
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Mounika Aare
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| | - Santanu Bhattacharya
- Department of Biochemistry and Molecular Biology, Mayo College of Medicine and Science, Jacksonville, FL 32224, USA
- Department of Physiology and Biomedical Engineering, Mayo College of Medicine and Science, Jacksonville, FL 32224, USA
| | - Tanmay Kulkarni
- Department of Biochemistry and Molecular Biology, Mayo College of Medicine and Science, Jacksonville, FL 32224, USA
| | - Li Sun
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, FL 32306, USA
| | - Deepak A Lamba
- Department of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
- Immunology and Regenerative Medicine, Genentech, South San Francisco, CA 94080, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Tallahassee, FL 32310, USA
| | - Mandip Singh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL 32307, USA
| |
Collapse
|
2
|
Balic N, Nikolac Perkovic M, Milos T, Vuic B, Kurtovic Kodzoman M, Svob Strac D, Nedic Erjavec G. Extracellular vesicles as a promising tool in neuropsychiatric pharmacotherapy application and monitoring. Prog Neuropsychopharmacol Biol Psychiatry 2025; 139:111393. [PMID: 40340017 DOI: 10.1016/j.pnpbp.2025.111393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 04/23/2025] [Accepted: 04/30/2025] [Indexed: 05/10/2025]
Abstract
This review deals with the application of extracellular vesicles (EVs) in the treatment of various neuropsychiatric disorders, including mood disorders, neurodegeneration, psychosis, neurological insults and injuries, epilepsy and substance use disorders. The main challenges of most neuropsychiatric pharmaceuticals nowadays are how to reach the central nervous system at therapeutic concentration and maintain it long enough and how to avoid undesirable side effects caused by unsatisfying toxicity. Extracellular vesicles, as very important mediators of intercellular communication, can have a variety of therapeutic qualities. They can act neuroprotective, regenerative and anti-inflammatory, but they also have characteristics of a good drug delivery system, including their nano- scale size, biological safety and abilities to cross BBB, to pack drugs within the lipid bilayer, and not to trigger an immunological response. Besides, due to their presence in readily accessible biofluids, they are good candidates for biomarkers of the disease, its progression and therapy response monitoring. Alternations in EVs' cargo profiles can reflect the pathogenesis of neuropsychiatric disorders, but they could also affect the disease outcomes. In the future, EVs could help physicians to tailor treatment strategies for individual patients, however, more extensive studies are needed to standardize isolation, purification and production procedures, increase efficacy of drug loading and limit unwanted effects of innate EVs' content.
Collapse
Affiliation(s)
- Nikola Balic
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | | | - Tina Milos
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | - Barbara Vuic
- Ruder Boskovic Institute, Bijenicka 54, 10000 Zagreb, Croatia.
| | | | | | | |
Collapse
|
3
|
Botto A, De Cesari C, Ndimurwanko N, Finamore F, Greco F, Cappello V, Casieri V, Immordino B, Lionetti V, Gemmi M, Tonazzini I, Giovannetti E, McDonnell LA. Novel PPT+SEC Workflow for High-Sensitivity Extracellular Vesicle Proteomics from Cell Media. J Proteome Res 2025. [PMID: 40315925 DOI: 10.1021/acs.jproteome.5c00082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
Abstract
Size exclusion chromatography (SEC) is a well-established method for the isolation of extracellular vesicles (EVs), but the large elution volumes necessitate a concentration step prior to proteomics analysis. This concentration step can lead to a significant EV loss. Here we report an EV proteomics approach that enables the isolation of EVs into just 80 μL, which is directly compatible with proteomics analysis without the need for a prior concentration. EVs were characterized by transmission electron microscopy, Western blot, and nanoparticle tracking analysis, all of which confirmed the presence of small EVs. Proteomics analysis of the EVs was performed and benchmarked against those isolated by using an automated UHPLC-SEC platform. The novel workflow identified more proteins and more EV markers, including 96 of the 100 top exosomal proteins from the ExoCarta database, compared to 91 identified using EV samples isolated by UHPLC-SEC. When applied to EVs isolated from pancreatic cancer cell lines, the workflow demonstrated higher sensitivity for previously reported EV markers of pancreatic cancer.
Collapse
Affiliation(s)
- Asia Botto
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
- Department of Chemistry and Industrial Chemistry, University of Pisa, 56124 Pisa, Italy
| | - Chiara De Cesari
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
- Nanoscience Institute, National Research Council, @NEST, 56127 Pisa, Italy
| | - Noa Ndimurwanko
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
- Scuola Normale Superiore, 56126 Pisa, Italy
| | - Francesco Finamore
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
| | - Francesco Greco
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
| | - Valentina Cappello
- Center for Materials Interfaces, Istituto Italiano di Tecnologia, 56025 Pontedera, Italy
| | | | - Benoit Immordino
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
- Scuola Superiore Sant'Anna, 56127 Pisa, Italy
| | - Vincenzo Lionetti
- Scuola Superiore Sant'Anna, 56127 Pisa, Italy
- UOSVD Anesthesia and Intensive Care, Fondazione Toscana G. Monasterio, 56124 Pisa, Italy
| | - Mauro Gemmi
- Center for Materials Interfaces, Istituto Italiano di Tecnologia, 56025 Pontedera, Italy
| | - Ilaria Tonazzini
- Nanoscience Institute, National Research Council, @NEST, 56127 Pisa, Italy
| | - Elisa Giovannetti
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
- Department of Medical Oncology, Cancer Center Amsterdam, VU University, 1081 HV Amsterdam, The Netherlands
| | - Liam A McDonnell
- Fondazione Pisana per la Scienza ONLUS, 56017 San Giuliano Terme (PI), Italy
| |
Collapse
|
4
|
Tao K, Tao K, Wang J. The potential mechanisms of extracellular vesicles in transfusion-related adverse reactions: Recent advances. Transfus Clin Biol 2025; 32:205-227. [PMID: 40180029 DOI: 10.1016/j.tracli.2025.03.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 03/15/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
Blood transfusion is an irreplaceable clinical treatment. Blood components are differentiated and stored according to specific guidelines. Storage temperatures and times vary depending on the blood component, but they all release extracellular vesicles (EVs) during storage. Although blood transfusions can be life-saving, they can also cause many adverse transfusion reactions, among which the effects of EVs are of increasing interest to researchers. EVs are submicron particles that vary in size, composition, and surface biomarkers, are encapsulated by a lipid bilayer, and are not capable of self-replication. EVs released by blood cells are important contributors to pathophysiologic states through proinflammatory, coagulant, and immunosuppressive effects, which in turn promote or inhibit the associated disease phenotype. Therefore, this review explores the potential mechanisms of hematopoietic-derived EVs in transfusion-associated adverse reactions and discusses the potential of the latest proteomics tools to be applied to the analysis of EVs in the field of transfusion medicine with a view to reducing the risk of blood transfusion.
Collapse
Affiliation(s)
- Keyi Tao
- Panzhihua University, Panzhihua 617000 Sichuan, China
| | - Keran Tao
- Institute of Medicine and Nursing, Hubei University of Medicine, Shiyan 442000 Hubei, China
| | - Jing Wang
- Southwest Medical University, Luzhou 646000 Sichuan, China; Department of Blood Transfusion, The Affiliated Hospital of Southwest Medical University, Luzhou Sichuan, 646000 China.
| |
Collapse
|
5
|
Hoang VT, Nguyen QT, Phan TTK, Pham TH, Dinh NTH, Anh LPH, Dao LTM, Bui VD, Dao H, Le DS, Ngo ATL, Le Q, Nguyen Thanh L. Tissue Engineering and Regenerative Medicine: Perspectives and Challenges. MedComm (Beijing) 2025; 6:e70192. [PMID: 40290901 PMCID: PMC12022429 DOI: 10.1002/mco2.70192] [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: 07/06/2024] [Revised: 12/30/2024] [Accepted: 03/04/2025] [Indexed: 04/30/2025] Open
Abstract
From the pioneering days of cell therapy to the achievement of bioprinting organs, tissue engineering, and regenerative medicine have seen tremendous technological advancements, offering solutions for restoring damaged tissues and organs. However, only a few products and technologies have received United States Food and Drug Administration approval. This review highlights significant progress in cell therapy, extracellular vesicle-based therapy, and tissue engineering. Hematopoietic stem cell transplantation is a powerful tool for treating many diseases, especially hematological malignancies. Mesenchymal stem cells have been extensively studied. The discovery of induced pluripotent stem cells has revolutionized disease modeling and regenerative applications, paving the way for personalized medicine. Gene therapy represents an innovative approach to the treatment of genetic disorders. Additionally, extracellular vesicle-based therapies have emerged as rising stars, offering promising solutions in diagnostics, cell-free therapeutics, drug delivery, and targeted therapy. Advances in tissue engineering enable complex tissue constructs, further transforming the field. Despite these advancements, many technical, ethical, and regulatory challenges remain. This review addresses the current bottlenecks, emphasizing novel technologies and interdisciplinary research to overcome these hurdles. Standardizing practices and conducting clinical trials will balance innovation and regulation, improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Van T. Hoang
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Quyen Thi Nguyen
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Trang Thi Kieu Phan
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Trang H. Pham
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Nhung Thi Hong Dinh
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Le Phuong Hoang Anh
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Lan Thi Mai Dao
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Van Dat Bui
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- School of Chemical EngineeringCollege of EngineeringSungkyunkwan University (SKKU)SuwonRepublic of Korea
| | - Hong‐Nhung Dao
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Duc Son Le
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Anh Thi Lan Ngo
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Quang‐Duong Le
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| | - Liem Nguyen Thanh
- Vinmec Research Institute of Stem Cell and Gene TechnologyCollege of Health SciencesVinUniversityVinhomes Ocean ParkHanoiVietnam
- Vinmec Health Care SystemHanoiVietnam
| |
Collapse
|
6
|
Meza-Morales W, Ayus-Martinez S, Jimenez-Osorio J, Buendia-Otero M, López L, Suleiman D, Suarez E, Freytes DO, Cunci L, Mora C. Functionalized screen-printed electrodes for non-invasive detection of vascular-endothelial cadherin in extracellular vesicles. RSC Adv 2025; 15:12609-12621. [PMID: 40264865 PMCID: PMC12012609 DOI: 10.1039/d4ra08926j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 04/07/2025] [Indexed: 04/24/2025] Open
Abstract
In this study, we developed a biosensor using a gold screen-printed electrode (Au-SPE) functionalized with mercaptoundecanoic acid (MUA) and an antibody for detecting the vascular-endothelial cadherin (CD144) as a endothelial biomarker protein on extracellular vesicles (EVs) isolated from saliva. The MUA functionalization provides a stable platform for immobilizing the CD144 antibody, ensuring the detection of the target protein. This biosensor combines Au-SPE technology with an immunoassay, offering a rapid, sensitive, and non-invasive method for detection of CD144 carried by EVs. Characterization of saliva-derived EVs using transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA) confirmed their morphology and size, which fell within the expected range of 80-180 nm. NTA indicated a lower concentration of particles in saliva-EVs than in serum-EVs (controls), highlighting the need for sensitive detection of EV cargos in this type of EV. Immunodetection confirmed the presence of CD144 in both saliva and serum-derived EVs, with higher concentrations in serum. Functionalization of Au-SPEs with MUA and CD144 antibodies was confirmed by significant resistance changes, and atomic force microscopy (AFM) was used to verify the preservation of EV morphology and their capturing post-immune adsorption. A calibration curve demonstrated the high sensitivity of the biosensor prototype for detecting CD144-positive EVs, with a limit of detection (LOD) of 0.111 ng mL-1 and a limit of quantification (LOQ) of 0.37 ng mL-1, requiring only 3 μL of EV-sample. This biosensor shows potential as a novel method for detecting and studying endothelial biomarkers associated with cardiovascular disease in EVs isolated from saliva, a capability not currently available with existing tools. Furthermore, it provides a key platform for expanding research to other biomarkers and diseases by monitoring protein cargos in the EVs, enhancing its utility across diverse clinical applications.
Collapse
Affiliation(s)
- William Meza-Morales
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| | - Sahimy Ayus-Martinez
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| | - Jesus Jimenez-Osorio
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| | - Maria Buendia-Otero
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| | - Luis López
- Department of Chemistry, University of Puerto Rico-Rio Piedras 601 Av. Universidad San Juan Puerto Rico USA
| | - David Suleiman
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| | - Edu Suarez
- Department of Biology, University of Puerto Rico-Ponce Av. Santiago de los Caballeros Ponce Puerto Rico USA
| | - Donald O Freytes
- Lampe Joint Department of Biomedical Engineering, The University of North Carolina at Chapel Hill/North Carolina State University 4130 Engineering Building III, Campus Box 7115 Raleigh NC 27695 USA
| | - Lisandro Cunci
- Department of Chemistry, University of Puerto Rico-Rio Piedras 601 Av. Universidad San Juan Puerto Rico USA
| | - Camilo Mora
- Department of Chemical Engineering, University of Puerto Rico-Mayaguez Route 108 Mayaguez Puerto Rico USA
| |
Collapse
|
7
|
Tiwari P, Shukla RP, Yadav K, Sharma M, Bakshi AK, Panwar D, Singh N, Agarwal N, Mugale MN, Mishra PR. YIGSR Functionalized Hybrid Exosomes Spatially Target Dasatinib to Laminin Receptors for Precision Therapy in Breast Cancer. Adv Healthc Mater 2025; 14:e2402673. [PMID: 39962816 DOI: 10.1002/adhm.202402673] [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: 07/21/2024] [Revised: 12/17/2024] [Indexed: 04/08/2025]
Abstract
In this study, YIGSR-functionalized exosomes (Exo) are engineered and hybridized with lipid polymeric nanoparticles (LPNPs) followed by loading of chemotherapy Dasatinib (DST) to spatially target laminin receptors on tumors. Exo derived from differentiated macrophages are engineered with YIGSR targeting peptides.These YIGSR-Exo are subsequently fused with LPNPs membranes using the freeze-thaw method, resulting in fused hybrid YIGSR-Exo, which are then loaded with DST, creating DST-FuNP@YIGSR-Exo and targeted breast cancer (BC), leading to enhanced mitochondrial membrane potential (54.50 ±5.0%), increased reactive oxygen species (59.50 ± 6.0%), and apoptosis (63 ± 6.5%), ultimately inducing cell death. Further, cellular uptake and receptor blocking studies confirm the binding affinity and interaction of DST-FuNP@YIGSR-Exo with laminin receptors, Intravenous pharmacokinetic analysis of DST-FuNP@YIGSR-Exo reveals a significant improvement in AUC0-∞, with a 20.84-fold increase compared to free DST and a 1.61-fold enhancement over DST-FuNP@Exo. This is further supported by in vivo imaging and demonstrated improved tumor localization. A tumor regression study shows a 6.8-fold reduction in tumors. Tumor tissue-specific IHC for the Ki67 proliferative marker is significantly reduced in the targeted formulation. The potential of DST-FuNP@YIGSR-Exo as an effective carrier for delivering chemotherapeutic drugs, paving the path for the advancement of biologically obtained nanocarriers for targeted breast cancer.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Dilip Panwar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Neha Agarwal
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Madhav Nilakanth Mugale
- Division of Toxicology and Experimental Medicine, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, 226031, India
- Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, Uttar Pradesh, 201002, India
| |
Collapse
|
8
|
Gao Y, Xie J, Yang Z, Li M, Yuan H, Li R. Functional tumor-derived exosomes in NSCLC progression and clinical implications. Front Pharmacol 2025; 16:1485661. [PMID: 40176898 PMCID: PMC11962733 DOI: 10.3389/fphar.2025.1485661] [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: 09/20/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancer cases and remains one of the leading causes of cancer-related mortality worldwide. The high mortality rate is primarily driven by delayed diagnosis, rapid metastasis, and frequent recurrence. Tumor-derived exosomes (TEXs) have emerged as critical mediators in NSCLC progression, offering valuable insights into the tumor microenvironment. Exosomes are small membrane vesicles that facilitate intercellular communication and transport bioactive molecules, including proteins, RNAs, and DNAs, thereby reflecting the genetic complexity of tumors. These exosomes play a key role in promoting tumor metastasis, epithelial-mesenchymal transition (EMT), neovascularization, drug resistance, and immune evasion, all of which are pivotal in the development of NSCLC. This review explores the diverse roles of TEXs in NSCLC progression, focusing on their involvement in pre-metastatic niche formation, tissue metastasis, and immune modulation. Specifically, we discuss the roles of exosome-associated RNAs and proteins in NSCLC, and their contribute to tumor growth and metastasis. Furthermore, we explore the potential of TEXs as biomarkers for NSCLC, emphasizing their application in diagnosis, prognosis, and prediction of resistance to targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Yuxin Gao
- Department of Abdominal Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Xie
- Information Technology Center, West China Hospital of Sichuan University, Chengdu, China
- Information Technology Center, West China Sanya Hospital of Sichuan University, Sanya, China
| | - Zhenya Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Mengxi Li
- College of pharmacy, Chengdu Medical College, Chengdu, China
| | - Hongfan Yuan
- Department of Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
9
|
Grossini E, Surico D, Venkatesan S, Ola Pour MM, Aquino CI, Remorgida V. Extracellular Vesicles and Pregnancy-Related Hypertensive Disorders: A Descriptive Review on the Possible Implications "From Bench to Bedside". BIOLOGY 2025; 14:240. [PMID: 40136497 PMCID: PMC11939443 DOI: 10.3390/biology14030240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/08/2025] [Accepted: 02/21/2025] [Indexed: 03/27/2025]
Abstract
Pregnancy involves extracellular vesicles (EVs) through mechanisms that are poorly understood to date. Furthermore, it is not surprising that EVs may also be involved in the pathophysiology of pre-eclampsia (PE) and gestational hypertension, two clinical conditions with high morbidity and mortality, given their capacity to mediate intracellular communications and regulate inflammation and angiogenesis. We searched major online scientific search engines (PubMed, Google Scholar, Scopus, WES, Embase, etc.) using the terms "Preeclampsia", "Pregnancy", "Hypertension", "Pregnancy-related hypertension", "Extracellular vesicles", "Biomarkers", "Gestation" AND "Obstetrics". Finding potential early biomarkers of risk or illness progression would be essential for the optimum care of expectant mothers with the aforementioned conditions. Nevertheless, none of the various screening assays that have been discovered recently have shown high predictive values. The analysis of EVs in the peripheral blood starting from the first trimester of pregnancy may hold great promise for the possible correlation with gestational hypertension problems and represent a marker of the early stages of the disease. EVs use may be a novel therapeutic approach for the management of various illnesses, as well. In order to define EVs' function in the physiopathology of pregnancy-associated hypertension and PE, as well as their potential as early biomarkers and therapeutic tools, we have compiled the most recent data in this review.
Collapse
Affiliation(s)
- Elena Grossini
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Daniela Surico
- Gynecology and Obstetrics Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.S.); (C.I.A.); (V.R.)
| | - Sakthipriyan Venkatesan
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Mohammad Mostafa Ola Pour
- Laboratory of Physiology, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (S.V.); (M.M.O.P.)
| | - Carmen Imma Aquino
- Gynecology and Obstetrics Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.S.); (C.I.A.); (V.R.)
| | - Valentino Remorgida
- Gynecology and Obstetrics Unit, Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy; (D.S.); (C.I.A.); (V.R.)
| |
Collapse
|
10
|
Furioso Ferreira R, Ghaffari MH, Ceciliani F, Fontana M, Caruso D, Audano M, Savoini G, Agazzi A, Mrljak V, Sauerwein H. Untargeted lipidomics reveals unique lipid signatures of extracellular vesicles from porcine colostrum and milk. PLoS One 2025; 20:e0313683. [PMID: 39946395 PMCID: PMC11825007 DOI: 10.1371/journal.pone.0313683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 10/30/2024] [Indexed: 02/16/2025] Open
Abstract
Extracellular vesicles (EV) are membranous vesicles considered as significant players in cell-to-cell communication. Milk provides adequate nutrition, transfers immunity, and promotes neonatal development, and milk EV are suggested to play a crucial role in these processes. Milk samples were obtained on days 0, 7, and 14 after parturition from sows receiving either a standard diet (ω-6:ω-3 = 13:1) or a test diet enriched in ω-3 (ω-6:ω-3 = 4:1). EV were isolated using ultracentrifugation coupled with size exclusion chromatography, and characterized by nanoparticle tracking analysis, transmission electron microscopy, and assessment of EV markers via Western blotting. The lipidome was determined following a liquid chromatography-quadrupole time-of-flight mass spectrometry approach. Here, we show that different stages of lactation (colostrum vs mature milk) have a distinct extracellular vesicle lipidomic profile. The distinct lipid content can be further explored to understand and regulate milk EV functionalities and primordial for enabling their diagnostic and therapeutic potential.
Collapse
Affiliation(s)
- Rafaela Furioso Ferreira
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn, Germany
- Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Morteza H. Ghaffari
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn, Germany
| | - Fabrizio Ceciliani
- Department of Veterinary Medicine and Animal Sciences, Università degli Studi di Milano, Lodi, Italy
| | - Manuela Fontana
- Unitech OMICs, Università degli Studi di Milano, Milano, Italy
| | - Donatella Caruso
- Unitech OMICs, Università degli Studi di Milano, Milano, Italy
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Matteo Audano
- Unitech OMICs, Università degli Studi di Milano, Milano, Italy
| | - Giovanni Savoini
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Alessandro Agazzi
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milano, Italy
| | - Vladimir Mrljak
- Faculty of Veterinary Medicine, University of Zagreb, Zagreb, Croatia
| | - Helga Sauerwein
- Institute of Animal Science, Physiology Unit, University of Bonn, Bonn, Germany
| |
Collapse
|
11
|
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
|
12
|
Goswami V, Sodhi KK, Singh CK. Innovative approaches to asthma treatment: harnessing nanoparticle technology. DISCOVER NANO 2025; 20:21. [PMID: 39922940 PMCID: PMC11807046 DOI: 10.1186/s11671-025-04211-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
In the domain of respiratory illnesses, asthma remains a critical obstacle. The heterogeneous nature of this chronic inflammatory disease poses challenges during its treatment. Glucocorticoid-based combination drug therapy now dominates clinical treatments for asthma; however, glucocorticoid resistance, numerous adverse effects, the incidence of inadequate drug delivery, and other factors need the development of more effective therapies. In recent years, there has been extensive research on nanotechnology in medicine. It has been shown in studies that these drug delivery systems can greatly enhance targeting and bioavailability and decrease the toxicity of medication. Nanoparticle drug delivery systems offer improved therapeutic efficacy compared to conventional administration techniques. Nanotechnology enables advancements in precision medicine, offering benefits for heterogeneous conditions such as asthma. This review will examine the critical factors of asthma to consider when formulating medications, as well as the role of nanomaterials and their mechanisms of action in pulmonary medicine for asthma treatment.
Collapse
Affiliation(s)
- Vanshika Goswami
- Department of Zoology, Hansraj College, University of Delhi, Delhi, 110007, India
| | - Kushneet Kaur Sodhi
- Department of Zoology, Sri Guru Tegh Bahadur Khalsa College, University of Delhi, Delhi, 110007, India
| | | |
Collapse
|
13
|
Kasprzyk-Pochopień J, Kamińska A, Mielczarek P, Piekoszewski W, Klimkowska A, Sładek K, Soja J, Adamek D, Stępień E. Comparison of nanoLC-MALDI-MS/MS with nanoLC-TIMS-MS/MS in the proteomic analysis of extracellular vesicles of bronchoalveolar lavage fluid. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2025; 17:1173-1187. [PMID: 39835386 DOI: 10.1039/d4ay01599a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
The study aims to evaluate and compare two advanced proteomic techniques, nanoLC-MALDI-MS/MS and nanoLC-TIMS-MS/MS, in characterizing extracellular vesicles (EVs) from the bronchoalveolar lavage fluid (BALF) of patients with asthma and idiopathic pulmonary fibrosis (IPF). Pulmonary diseases, driven by pollutants and infections, often necessitate detailed analysis of BALF to identify diagnostic biomarkers and therapeutic targets. EVs, which include exosomes, microvesicles, and apoptotic bodies, are isolated using filtration and ultracentrifugation, and their morphology, concentration, and size distribution are assessed through transmission electron microscopy (TEM) and nanoparticle tracking analysis (NTA). The proteomic profiles of these EVs are then analyzed using the aforementioned techniques, highlighting their unique and common proteins. The study found that nanoLC-TIMS-MS/MS identified significantly more proteins compared to nanoLC-MALDI-MS/MS. Functional analysis via Gene Ontology revealed pathways related to inflammation and cell signaling, underscoring the role of EVs in disease pathophysiology. The findings suggest that EVs in BALF can serve as valuable biomarkers and therapeutic targets in respiratory diseases, providing a foundation for future research and clinical applications.
Collapse
Affiliation(s)
- Joanna Kasprzyk-Pochopień
- Laboratory of High-Resolution Mass Spectrometry, Faculty of Chemistry, Jagiellonian University, Krakow, Poland.
| | - Agnieszka Kamińska
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Przemysław Mielczarek
- Department of Analytical Chemistry and Biochemistry, Faculty of Materials Science and Ceramics, AGH University of Krakow, Krakow, Poland
| | - Wojciech Piekoszewski
- Laboratory of High-Resolution Mass Spectrometry, Faculty of Chemistry, Jagiellonian University, Krakow, Poland.
- Department of Analytical Chemistry, Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | | | - Krzysztof Sładek
- Department of Pulmonology, University Hospital in Krakow, Krakow, Poland
| | - Jerzy Soja
- Department of Pulmonology, University Hospital in Krakow, Krakow, Poland
| | - Dariusz Adamek
- Department of Pathomorphology, Faculty of Medicine Jagiellonian University, Krakow, Poland
| | - Ewa Stępień
- Department of Medical Physics, M. Smoluchowski Institute of Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
- Total-Body Jagiellonian-PET Laboratory, Jagiellonian University, Krakow, Poland
- Center for Theranostics, Jagiellonian University, Krakow, Poland
| |
Collapse
|
14
|
Zarovni N, Mladenović D, Brambilla D, Panico F, Chiari M. Stoichiometric constraints for detection of EV-borne biomarkers in blood. J Extracell Vesicles 2025; 14:e70034. [PMID: 39901737 PMCID: PMC11791308 DOI: 10.1002/jev2.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 12/03/2024] [Accepted: 12/16/2024] [Indexed: 02/05/2025] Open
Abstract
Stochiometric issues, encompassing both the quantity and heterogeneity of extracellular vesicles (EVs) derived from tumour or other tissues in blood, pose important challenges across various stages of biomarker discovery and detection, affecting the integrity of data, introducing losses and artifacts during blood processing, EV purification and analysis. These challenges shape the diagnostic utility of EVs especially within the framework of established and emerging methodologies. By addressing these challenges, we aim to delineate crucial parameters and requirements for tumour-specific EV detection, or more precisely, for tumour identification via EV based assays. Our endeavour involves a comprehensive examination of the layers that mask or confound the traceability of EV markers such as nucleic acids and proteins, and focus on 'low prevalence-low concentration' scenario. Finally, we evaluate the advantages versus limitations of single-particle analysers over more conventional bulk assays, suggesting that the combined use of both to capture and interpret the EV signals, in particular the EV surface displayed proteins, may ultimately provide quantitative information on their absolute abundance and distribution.
Collapse
Affiliation(s)
| | - Danilo Mladenović
- HansaBioMed Life Sciences OÜTallinnEstonia
- School of Natural Sciences and HealthTallinn UniversityTallinnEstonia
| | - Dario Brambilla
- Institute of Chemical Sciences and TechnologyNational Research Council of ItalyMilanItaly
| | - Federica Panico
- Institute of Chemical Sciences and TechnologyNational Research Council of ItalyMilanItaly
| | - Marcella Chiari
- RoseBioMilanItaly
- Institute of Chemical Sciences and TechnologyNational Research Council of ItalyMilanItaly
| |
Collapse
|
15
|
Choi Y, Park JH, Jo A, Lim CW, Park JM, Hwang JW, Lee KS, Kim YS, Lee H, Moon J. Blood-derived APLP1 + extracellular vesicles are potential biomarkers for the early diagnosis of brain diseases. SCIENCE ADVANCES 2025; 11:eado6894. [PMID: 39742488 DOI: 10.1126/sciadv.ado6894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 11/19/2024] [Indexed: 01/03/2025]
Abstract
The early detection of neurodegenerative diseases necessitates the identification of specific brain-derived biomolecules in peripheral blood. In this context, our investigation delineates the role of amyloid precursor-like protein 1 (APLP1)-a protein predominantly localized in oligodendrocytes and neurons-as a previously unidentified biomarker in extracellular vesicles (EVs). Through rigorous analysis, APLP1+ EVs from human sera were unequivocally determined to be of cerebral origin. This assertion was corroborated by distinctive small RNA expression patterns of APLP1+ EVs. The miRNAs' putative targets within these EVs manifested pronounced expression in the brain, fortifying their neurospecific provenance. We subjected our findings to stringent validation using Thy-1 GFP M line mice, transgenic models wherein GFP expression is confined to hippocampal neurons. An amalgamation of these results with an exhaustive data analysis accentuates the potential of APLP1+ EVs as cerebrally originated biomarkers. Synthesizing our findings, APLP1+ EVs are postulated not merely as diagnostic markers but as seminal entities shaping the future trajectory of neurodegenerative disease diagnostics.
Collapse
Affiliation(s)
- Yuri Choi
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Jae Hyun Park
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Ala Jo
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Chul-Woo Lim
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Ji-Min Park
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Jin Woo Hwang
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| | - Kang Soo Lee
- Department of Psychiatry, CHA Bundang Medical Center, CHA University College of Medicine, Gyeonggi-do 13496, Republic of Korea
| | - Young-Sang Kim
- Department of Family Medicine, CHA Bundang Medical Center, CHA University College of Medicine, Gyeonggi-do 13496, Republic of Korea
| | - Hakho Lee
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jisook Moon
- Department of Biotechnology, College of Life Science, CHA University, Gyeonggi-do 13488, Republic of Korea
| |
Collapse
|
16
|
Špilak A, Brachner A, Friedl HP, Klepe A, Nöhammer C, Neuhaus W. Effects of small extracellular vesicles derived from normoxia- and hypoxia-treated prostate cancer cells on the submandibular salivary gland epithelium in vitro. Tissue Barriers 2025; 13:2347062. [PMID: 38721756 PMCID: PMC11875469 DOI: 10.1080/21688370.2024.2347062] [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: 12/28/2023] [Revised: 04/18/2024] [Accepted: 04/18/2024] [Indexed: 03/03/2025] Open
Abstract
Small extracellular vesicles (sEVs) are an important part of intercellular communication. They are phospholipid bilayer particles that carry active biomolecules such as proteins, various nucleic acids, and lipids. In recipient cells, sEVs can alter cellular functions, including cancer development and premetastatic niche formation in distant organs. Moreover, sEVs can carry cancer-specific features, which makes them promising biomarker candidates. However, the interactions of sEVs with biological barriers and consequences thereof, are not clarified yet. The blood-saliva barrier is crucial for preventing the entry of pathogens and (in)organic substances into the bloodstream, as well as molecule filtration from blood to saliva. The effects of brain derived DU145 prostate cancer (PCa) sEVs on a human submandibular salivary gland barrier (SSGB) in vitro were investigated. Small EVs were harvested from normoxic (N, atmospheric O2) or hypoxic (H, 1% O2) conditions, fluorescently labeled with CellTrackerTM Orange and thoroughly characterized. HTB-41 B2 cells were used as SSGB model cultured on 24-well ThinCert® inserts. After model optimization indicating effects of serum and serum-sEVs on barrier properties, PCa sEVs were applied to the basolateral (blood) side in either 10% serum, or serum-free conditions, and barrier integrity was continuously monitored for 40 hours. This study found that H and N PCa sEVs were uptaken by the SSGB in vitro model in similar quantities regardless of the media composition in the basolateral compartment. Permeation of fluorescent PCa sEVs into the apical compartment was not detectable with the applied methods. However, treatment with H and N sEVs under different serum conditions revealed distinct molecular clusters after hierarchical analysis of mRNA data measured by high-throughput qPCR, which were partly reflected at the protein level. For example, serum-reduction dependent decrease of barrier properties was accompanied with the decrease of CDH1 or Claudin-7 expression. Interestingly, the presence of H sEVs significantly increased the number of sEV-sized particles in the apical compartment of the SSGB model compared to basolaterally added N sEVs. This functional effect on the number of particles in the saliva (apical) compartment induced by different sEVs applied in the blood (basolateral) compartment might be a new approach to understand one possible mechanism how differences of salivary EVs might occur which then could be used as biomarker.
Collapse
Affiliation(s)
- Ana Špilak
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, AIT - Austrian Institute of Technology GmbH, Vienna, Austria
| | - Andreas Brachner
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, AIT - Austrian Institute of Technology GmbH, Vienna, Austria
| | - Heinz-Peter Friedl
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, AIT - Austrian Institute of Technology GmbH, Vienna, Austria
| | - Adrián Klepe
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, AIT - Austrian Institute of Technology GmbH, Vienna, Austria
| | - Christa Nöhammer
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, AIT - Austrian Institute of Technology GmbH, Vienna, Austria
| | - Winfried Neuhaus
- Competence Unit Molecular Diagnostics, Center for Health and Bioresources, AIT - Austrian Institute of Technology GmbH, Vienna, Austria
- Faculty of Medicine and Dentistry, Danube Private University, Krems, Austria
| |
Collapse
|
17
|
Butta N, van der Wal DE. Desialylation by neuraminidases in platelets, kiss of death or bittersweet? Curr Opin Hematol 2025; 32:43-51. [PMID: 38529832 DOI: 10.1097/moh.0000000000000815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
PURPOSE OF REVIEW Loss of surface sialic acid by neuraminidases is known as 'desialylation'. Platelets are desialylated in bacterial or viral infections, during storage, senescence, various mutations, platelet auto antibodies, hemostasis and shear stress. In this review the recent literature on the different sialic acid capped glycan structures will be covered as well as platelet desialylation in inherited glycan disorders and induced by external neuraminidases. RECENT FINDINGS Neuraminidases are released from platelet intracellular stores and translocated to the platelet surface. Apart from clearance, loss of surface sialic acid by neuraminidases ('desialylation') affects platelet signaling including ligand binding and their procoagulant function. Platelets are also desialylated in infections, various mutations, presence of platelet auto antibodies. SUMMARY Since platelet desialylation occurs in various healthy and pathological conditions, measuring desialylation might be a new diagnostic tool.
Collapse
Affiliation(s)
- Nora Butta
- Group of Coagulopathies and Haemostasis Disorders, La Paz University Hospital Research Institute (IdiPAZ), Madrid, Spain
| | - Dianne E van der Wal
- Platelets and Thrombosis Research Laboratory, Anzac Research Institute, Concord Repatriation General Hospital, Concord, New South Wales, Australia
| |
Collapse
|
18
|
Ranjit A, Lee CB, Tenora L, Mettu VS, Pal A, Alt J, Slusher BS, Rais R. Pharmacokinetic Evaluation of Neutral Sphinghomyelinase2 (nSMase2) Inhibitor Prodrugs in Mice and Dogs. Pharmaceutics 2024; 17:20. [PMID: 39861669 PMCID: PMC11768932 DOI: 10.3390/pharmaceutics17010020] [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: 12/09/2024] [Revised: 12/22/2024] [Accepted: 12/23/2024] [Indexed: 01/27/2025] Open
Abstract
Background: Extracellular vesicles (EVs) can carry pathological cargo, contributing to disease progression. The enzyme neutral sphingomyelinase 2 (nSMase2) plays a critical role in EV biogenesis, making it a promising therapeutic target. Our lab previously identified a potent and selective inhibitor of nSMase2, named DPTIP (IC50 = 30 nM). Although promising, DPTIP exhibits poor pharmacokinetics (PKs) with a low oral bioavailability (%F < 5), and a short half-life (t1/2 ≤ 0.5 h). To address these limitations, we previously developed DPTIP prodrugs by masking its phenolic hydroxyl group, demonstrating improved plasma exposure in mice. Recognizing that species-specific metabolic differences can influence prodrug PK, we expanded our studies to evaluate selected prodrugs in both mice and dogs. Methods: The scaleup of selected prodrugs was completed and two additional valine- ester based prodrugs were synthesized. Mice were dosed prodrugs via peroral route (10 mg/kg equivalent). For dog studies DPTIP was dosed via intravenous (1 mg/kg) or peroral route (2 mg/kg) and prodrugs were given peroral at a dose 2 mg/kg DPTIP equivalent. Plasma samples were collected at predetermined points and analyzed using developed LC/MS-MS methods. Results: In mice, several of the tested prodrugs showed similar or improved plasma exposures compared to DPTIP. However, in dog studies, the double valine ester prodrug 9, showed significant improvement with an almost two-fold increase in DPTIP plasma exposure (AUC0-t = 1352 vs. 701 pmol·h/mL), enhancing oral bioavailability from 8.9% to 17.3%. Conclusions: These findings identify prodrug 9 as a promising candidate for further evaluation and underscore the critical role of species-specific differences in prodrug PKs.
Collapse
Affiliation(s)
- Arina Ranjit
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Chae Bin Lee
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Lukáš Tenora
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Vijaya Saradhi Mettu
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Arindom Pal
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Jesse Alt
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Barbara S. Slusher
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Departments of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Rana Rais
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Johns Hopkins Drug Discovery, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
19
|
Celeski M, Segreti A, Crisci F, Cricco R, Piscione M, Di Gioia G, Nusca A, Fossati C, Pigozzi F, Ussia GP, Solaro RJ, Grigioni F. The Role of Cardiac Troponin and Other Emerging Biomarkers Among Athletes and Beyond: Underlying Mechanisms, Differential Diagnosis, and Guide for Interpretation. Biomolecules 2024; 14:1630. [PMID: 39766337 PMCID: PMC11727179 DOI: 10.3390/biom14121630] [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: 11/22/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/15/2025] Open
Abstract
Cardiovascular (CV) disease remains the leading cause of morbidity and mortality worldwide, highlighting the necessity of understanding its underlying molecular and pathophysiological pathways. Conversely, physical activity (PA) and exercise are key strategies in reducing CV event risks. Detecting latent CV conditions in apparently healthy individuals, such as athletes, presents a unique challenge. The early identification and treatment of CV disorders are vital for long-term health and patient survival. Cardiac troponin is currently the most commonly used biomarker for assessing CV changes in both athletes and the general population. However, there remains considerable debate surrounding the mechanisms underlying exercise-induced troponin elevations and its release in non-ischemic contexts. Thus, there is a pressing need to identify and implement more sensitive and specific biomarkers for CV disorders in clinical practice. Indeed, research continues to explore reliable biomarkers for evaluating the health of athletes and the effectiveness of physical exercise. It is essential to analyze current evidence on troponin release in non-ischemic conditions, post-strenuous exercise, and the complex biological pathways that influence its detection. Furthermore, this study summarizes current research on cytokines and exosomes, including their physiological roles and their relevance in various CV conditions, especially in athletes. In addition, this paper gives special attention to underlying mechanisms, potential biomarkers, and future perspectives.
Collapse
Affiliation(s)
- Mihail Celeski
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Andrea Segreti
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Filippo Crisci
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Riccardo Cricco
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Mariagrazia Piscione
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Giuseppe Di Gioia
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
- Institute of Sports Medicine and Science, Italian National Olympic Committee, Largo Piero Gabrielli 1, 00197 Roma, Italy
| | - Annunziata Nusca
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Chiara Fossati
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Fabio Pigozzi
- Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, Piazza Lauro de Bosis 6, 00135 Roma, Italy
| | - Gian Paolo Ussia
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| | - Ross John Solaro
- Department of Physiology and Biophysics and Center for Cardiovascular Research, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA;
| | - Francesco Grigioni
- Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy (R.C.)
- Unit of Cardiovascular Sciences, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy
| |
Collapse
|
20
|
Kale V. Extracellular vesicles as standard-of-care therapy: will fast-tracking the regulatory processes help achieve the goal? Regen Med 2024; 19:617-635. [PMID: 39688586 PMCID: PMC11730413 DOI: 10.1080/17460751.2024.2442847] [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: 08/10/2024] [Accepted: 12/12/2024] [Indexed: 12/18/2024] Open
Abstract
Extracellular Vesicles (EVs) became a focus of clinical research when experimental and pre-clinical studies showed that they mimic their parent cells' regenerative and therapeutic effects and their cargo carries disease-specific diagnostic and prognostic biomarkers. Since the publication of data forms an endpoint of the study, this review specifically focused on the published clinical trials done with EVs. For brevity, this review was restricted to the last 10 years. Unexpectedly, the literature search showed that very few clinical trials assessing the therapeutic applications of EVs were published in this period indicating that they have not reached their desired endpoint. Conversely, most studies showed the potential of EVs present in various biofluids as a promising source of diagnostic and prognostic biomarkers for various diseases, and predictive markers to assess the effectiveness of therapy. This stark difference in the numbers could perhaps be due to the time-consuming regulatory processes involved in the clinical-grade preparation and characterization of EVs, and the determination of their safety and effective dose regimens. One wonders whether fast-tracking regulatory affairs could help accelerate the therapeutic use of EVs. This aspect needs urgent attention.
Collapse
Affiliation(s)
- Vaijayanti Kale
- Symbiosis Centre for Stem Cell Research, Symbiosis International University, Pune, India
| |
Collapse
|
21
|
Chen Q, Zheng Y, Jiang X, Wang Y, Chen Z, Wu D. Nature's carriers: leveraging extracellular vesicles for targeted drug delivery. Drug Deliv 2024; 31:2361165. [PMID: 38832506 DOI: 10.1080/10717544.2024.2361165] [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: 08/07/2023] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
With the rapid development of drug delivery systems, extracellular vesicles (EVs) have emerged as promising stars for improving targeting abilities and realizing effective delivery. Numerous studies have shown when compared to conventional strategies in targeted drug delivery (TDD), EVs-based strategies have several distinguished advantages besides targeting, such as participating in cell-to-cell communications and immune response, showing high biocompatibility and stability, penetrating through biological barriers, etc. In this review, we mainly focus on the mass production of EVs including the challenges and strategies for scaling up EVs production in a cost-effective and reproducible manner, the loading and active targeting methods, and examples of EVs as vehicles for TDD in consideration of potential safety and regulatory issues associated. We also conclude and discuss the rigor and reproducibility of EVs production, the current research status of the application of EVs-based strategies to targeted drug delivery, clinical conversion prospects, and existing chances and challenges.
Collapse
Affiliation(s)
- Qi Chen
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou, P. R. China
| | - Yuyi Zheng
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuhong Jiang
- Epilepsy Center, Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Yi Wang
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Zhejiang Rehabilitation Medical Center, The Third Affiliated Hospital of Zhejiang, Chinese Medical University, Hangzhou, PR China
| | - Zhong Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
- Epilepsy Center, Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, PR China
| | - Di Wu
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
22
|
Richard M, Moreau R, Croyal M, Mathiot L, Frénel J, Campone M, Dupont A, Gavard J, André‐Grégoire G, Guével L. Monitoring concentration and lipid signature of plasma extracellular vesicles from HR + metastatic breast cancer patients under CDK4/6 inhibitors treatment. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e70013. [PMID: 39691590 PMCID: PMC11650302 DOI: 10.1002/jex2.70013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/05/2024] [Accepted: 09/20/2024] [Indexed: 12/19/2024]
Abstract
Extracellular vesicles (EVs) are cell-derived small membrane structures that transport various molecules. They have emerged as potential circulating biomarkers for monitoring responses to cancer therapies. This study aimed to comprehensively characterize plasma-carried EVs in hormone receptor-positive (HR+) metastatic breast cancer (MBC) patients treated with first-line CDK4/6 inhibitors (iCDK4/6) combined with endocrine therapy. MBC patients were classified into three groups based on their response to therapy: resistant, intermediate or sensitive. In a prospective cohort, we monitored the concentration of circulating EVs, analyzed their lipid signature and correlated these factors with treatment response. To facilitate the translation of EV research to clinical practice, we established a three-step procedure: (1) EVs were isolated from plasma using semi-automatized size exclusion chromatography (SEC); (2) EV concentration, termed vesiclemia, was determined by drop counting via interferometric light microscopy (ILM); and (3) EV lipid composition was analyzed by mass spectrometry. ILM-based vesiclemia values were highly fluctuating upon iCDK4/6 treatment, while early increase associated with accelerated progression. Of note, vesiclemia remained a steady parameter over a 1-year period in age-matched healthy women. Additionally, analysis of the EV cargo unveiled a distinct sphingolipid profile, characterized by increased levels of ceramides and sphingomyelins in resistant patients within the first 2 months of treatment. Based on 16 sphingolipid species, sensitive and resistant patients were correctly classified with an overall accuracy of 82%. This specific sphingolipid pattern was exclusively discernible within EVs, and not in plasma, highlighting the significance of EVs in the early prediction of individual responses to iCDK4/6 and disease progression. Overall, this study provides insights of the longitudinal characterization of plasma-borne EVs in both a healthy group and HR+ MBC patients under iCDK4/6 therapies. Combined vesiclemia and EV sphingolipid profile emphasize the promising potential of EVs as non-invasive biomarkers for monitoring early treatment response.
Collapse
Affiliation(s)
- Mathilde Richard
- Team SOAP, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes‐Angers (CRCINA), InsermCNRS, Nantes UniversitéNantesFrance
- Équipe Labellisée Ligue Contre le CancerParisFrance
| | - Rosalie Moreau
- Team SOAP, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes‐Angers (CRCINA), InsermCNRS, Nantes UniversitéNantesFrance
- Équipe Labellisée Ligue Contre le CancerParisFrance
| | - Mikaël Croyal
- Nantes Université, CHU Nantes, CNRS, INSERMNantesFrance
- Nantes Université, CHU Nantes, Inserm, CNRS, SFR Santé, Inserm UMS 016, CNRS UMS 3556NantesFrance
- CRNH‐Ouest Mass Spectrometry Core FacilityNantesFrance
| | - Laurent Mathiot
- Institut de Cancérologie de l'Ouest (ICO), Site Rene GauducheauSaint HerblainFrance
| | | | - Mario Campone
- Institut de Cancérologie de l'Ouest (ICO), Site Rene GauducheauSaint HerblainFrance
| | - Aurélien Dupont
- SFR UMS CNRS 3480, INSERM 018, Biosit biologie, santé, innovation technologiqueRennesFrance
| | - Julie Gavard
- Team SOAP, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes‐Angers (CRCINA), InsermCNRS, Nantes UniversitéNantesFrance
- Équipe Labellisée Ligue Contre le CancerParisFrance
- Institut de Cancérologie de l'Ouest (ICO), Site Rene GauducheauSaint HerblainFrance
| | - Gwennan André‐Grégoire
- Team SOAP, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes‐Angers (CRCINA), InsermCNRS, Nantes UniversitéNantesFrance
- Équipe Labellisée Ligue Contre le CancerParisFrance
- Institut de Cancérologie de l'Ouest (ICO), Site Rene GauducheauSaint HerblainFrance
| | - Laëtitia Guével
- Team SOAP, Centre de Recherche en Cancérologie et Immunologie Intégrée Nantes‐Angers (CRCINA), InsermCNRS, Nantes UniversitéNantesFrance
- Équipe Labellisée Ligue Contre le CancerParisFrance
| |
Collapse
|
23
|
Yadav A, Xuan Y, Sen CK, Ghatak S. Standardized Reporting of Research on Exosomes to Ensure Rigor and Reproducibility. Adv Wound Care (New Rochelle) 2024; 13:584-599. [PMID: 38888007 DOI: 10.1089/wound.2024.0093] [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: 06/20/2024] Open
Abstract
Significance: The study of extracellular vesicles (EVs), especially exosomes, has unlocked new avenues in understanding cellular communication and potential therapeutic applications. Recent Advances: Advancements in EV research have shown significant contributions from the International Society for Extracellular Vesicles (ISEV), in establishing methodological standards. The evolution of the Minimal Information for Studies of Extracellular Vesicles (MISEV) guidelines from 2014 to 2023 reflects enhanced research rigor and reproducibility. The launch of EV-TRACK platform promotes uniformity and reproducibility by providing a centralized repository for data sharing and standardization practices. Furthermore, databases like EVpedia and ExoCarta have facilitated data sharing and collaboration within the scientific community. Concurrently, exosome-based therapies have emerged as a forefront area within regenerative medicine and targeted drug delivery, showcasing the potential of exosomes in promoting tissue regeneration. Critical Issues: Despite advancements, the field grapples with challenges such as vesicular heterogeneity, EV isolation complexity, and standardization. These issues impact research reproducibility and clinical applications. The inconsistency in exosomal preparations in clinical trials poses significant challenges to therapeutic efficacy and safety. Future Directions: The review outlines critical areas for future research, including the need for technological innovation in EV isolation and characterization, the establishment of standardized protocols, and a deeper understanding of exosome biology. The review also highlights the need to reassess guidelines, develop new EV isolation and characterization technologies, and establish standardized protocols to overcome current limitations. Emphasis is placed on interdisciplinary research and collaboration to address the complexities of EV biology, improve clinical trial design, and ultimately realize exosome's therapeutic and diagnostic potential. Continued evaluation and rigorous scientific validation are essential for successful exosome integration.
Collapse
Affiliation(s)
- Anita Yadav
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Yi Xuan
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Chandan K Sen
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Subhadip Ghatak
- McGowan Institute for Regenerative Medicine, Department of Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
24
|
Bobbili MR, Görgens A, Yan Y, Vogt S, Gupta D, Corso G, Barbaria S, Patrioli C, Weilner S, Pultar M, Jacak J, Hackl M, Schosserer M, Grillari R, Kjems J, Andaloussi SEL, Grillari J. Snorkel-tag based affinity chromatography for recombinant extracellular vesicle purification. J Extracell Vesicles 2024; 13:e12523. [PMID: 39400515 PMCID: PMC11472238 DOI: 10.1002/jev2.12523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid nanoparticles and play an important role in cell-cell communications, making them potential therapeutic agents and allowing to engineer for targeted drug delivery. The expanding applications of EVs in next generation medicine is still limited by existing tools for scaling standardized EV production, single EV tracing and analytics, and thus provide only a snapshot of tissue-specific EV cargo information. Here, we present the Snorkel-tag, for which we have genetically fused the EV surface marker protein CD81, to a series of tags with an additional transmembrane domain to be displayed on the EV surface, resembling a snorkel. This system enables the affinity purification of EVs from complex matrices in a non-destructive form while maintaining EV characteristics in terms of surface protein profiles, associated miRNA patterns and uptake into a model cell line. Therefore, we consider the Snorkel-tag to be a widely applicable tool in EV research, allowing for efficient preparation of EV standards and reference materials, or dissecting EVs with different surface markers when fusing to other tetraspanins in vitro or in vivo.
Collapse
Affiliation(s)
- Madhusudhan Reddy Bobbili
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue Regeneration
| | - André Görgens
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Institute for Transfusion Medicine, University Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Yan Yan
- Department of Molecular Biology and Genetics, Centre for Cellular Signal Patterns (CellPat), Interdisciplinary Nanoscience Centre (iNANO)Aarhus UniversityAarhus CDenmark
- Omiics ApSAarhus NDenmark
| | - Stefan Vogt
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Dhanu Gupta
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Institute of Developmental and Regenerative MedicineUniversity of Oxford, IMS‐Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, HeadingtonOxfordUnited Kingdom
- Department of PaediatricsUniversity of Oxford, South Parks RoadOxfordUnited Kingdom
| | - Giulia Corso
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Evercyte GmbHViennaAustria
| | - Samir Barbaria
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Carolina Patrioli
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Sylvia Weilner
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | | | - Jaroslaw Jacak
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- School of Medical Engineering and Applied Social ScienceUniversity of Applied Sciences Upper AustriaLinzAustria
| | - Matthias Hackl
- Austrian Cluster for Tissue Regeneration
- TAmiRNATAmiRNA GmbHViennaAustria
| | - Markus Schosserer
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Austrian Cluster for Tissue Regeneration
- Institute of Medical GeneticsCenter for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | - Regina Grillari
- Austrian Cluster for Tissue Regeneration
- Evercyte GmbHViennaAustria
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, Centre for Cellular Signal Patterns (CellPat), Interdisciplinary Nanoscience Centre (iNANO)Aarhus UniversityAarhus CDenmark
| | - Samir EL Andaloussi
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
| | - Johannes Grillari
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue Regeneration
| |
Collapse
|
25
|
Wang Y, Jia J, Wang F, Fang Y, Yang Y, Zhou Q, Yuan W, Gu X, Hu J, Yang S. Pre-metastatic niche: formation, characteristics and therapeutic implication. Signal Transduct Target Ther 2024; 9:236. [PMID: 39317708 PMCID: PMC11422510 DOI: 10.1038/s41392-024-01937-7] [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: 02/28/2024] [Revised: 06/29/2024] [Accepted: 07/23/2024] [Indexed: 09/26/2024] Open
Abstract
Distant metastasis is a primary cause of mortality and contributes to poor surgical outcomes in cancer patients. Before the development of organ-specific metastasis, the formation of a pre-metastatic niche is pivotal in promoting the spread of cancer cells. This review delves into the intricate landscape of the pre-metastatic niche, focusing on the roles of tumor-derived secreted factors, extracellular vesicles, and circulating tumor cells in shaping the metastatic niche. The discussion encompasses cellular elements such as macrophages, neutrophils, bone marrow-derived suppressive cells, and T/B cells, in addition to molecular factors like secreted substances from tumors and extracellular vesicles, within the framework of pre-metastatic niche formation. Insights into the temporal mechanisms of pre-metastatic niche formation such as epithelial-mesenchymal transition, immunosuppression, extracellular matrix remodeling, metabolic reprogramming, vascular permeability and angiogenesis are provided. Furthermore, the landscape of pre-metastatic niche in different metastatic organs like lymph nodes, lungs, liver, brain, and bones is elucidated. Therapeutic approaches targeting the cellular and molecular components of pre-metastatic niche, as well as interventions targeting signaling pathways such as the TGF-β, VEGF, and MET pathways, are highlighted. This review aims to enhance our understanding of pre-metastatic niche dynamics and provide insights for developing effective therapeutic strategies to combat tumor metastasis.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Jiachi Jia
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Fuqi Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Yingshuai Fang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Yabing Yang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Quanbo Zhou
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Xiaoming Gu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Junhong Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| |
Collapse
|
26
|
Mazzarella R, Cajas YN, Gonzalez Martínez ME, Rizos D. Extracellular vesicles: emerging paradigms in bovine embryo-maternal communication. Anim Reprod 2024; 21:e20240065. [PMID: 39286362 PMCID: PMC11404873 DOI: 10.1590/1984-3143-ar2024-0065] [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: 05/17/2024] [Accepted: 06/25/2024] [Indexed: 09/19/2024] Open
Abstract
The oviduct and uterus provide an optimal environment for early embryo development, where effective communication between the embryo and the maternal reproductive tract is crucial for establishing and maintaining pregnancy. Oviductal and uterine-derived EVs play pivotal roles in this maternal-embryonic communication and in facilitating early embryo development. However, despite the ability of in vitro culture methods to produce viable embryos, the lack of exchange between the embryo and the mother often results in lower-quality embryos than those derived in vivo. Therefore, there is a pressing need to increase our understanding of the physiological mechanisms underlying embryo interaction with the oviduct and endometrium through EVs and to develop models capable of mimicking the in vivo environment. This review aims to provide up-to-date insights into the communication between the mother and pre-implantation bovine embryo, exploring their applications and perspectives in the field.
Collapse
Affiliation(s)
- Rosane Mazzarella
- Department of Animal Reproduction, National Institute for Agricultural and Food Research and Technology, Spanish National Research Council - INIA-CSIC, Madrid, Spain
| | - Yulia Nathaly Cajas
- Department Agrarian Production, Technical University of Madrid -UPM, Madrid, Spain
- Departamento de Ciencias Biológicas, Universidad Técnica Particular de Loja - UTPL, Loja, Ecuador
| | - Maria Encina Gonzalez Martínez
- Department of Anatomy and Embryology, Veterinary Faculty of the Complutense University of Madrid - FV-UCM, Madrid, Spain
| | - Dimitrios Rizos
- Department of Animal Reproduction, National Institute for Agricultural and Food Research and Technology, Spanish National Research Council - INIA-CSIC, Madrid, Spain
| |
Collapse
|
27
|
Ważny Ł, Whiteside TL, Pietrowska M. Oncoviral Infections and Small Extracellular Vesicles. Viruses 2024; 16:1291. [PMID: 39205265 PMCID: PMC11359865 DOI: 10.3390/v16081291] [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: 07/10/2024] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
Small extracellular vesicles (sEV) are small membrane-bound nanovesicles with a size range below 200 nm that are released by all types of cells. sEV carry a diverse cargo of proteins, lipids, glycans, and nucleic acids that mimic the content of producer cells. sEV mediate intercellular communication and play a key role in a broad variety of physiological and pathological conditions. Recently, numerous reports have emerged examining the role of sEV in viral infections. A significant number of similarities in the sEV biogenesis pathways and the replication cycles of viruses suggest that sEV might influence the course of viral infections in diverse ways. Besides directly modulating virus propagation by transporting the viral cargo (complete virions, proteins, RNA, and DNA), sEV can also modify the host antiviral response and increase the susceptibility of cells to infection. The network of mutual interactions is particularly complex in the case of oncogenic viruses, deserving special consideration because of its significance in cancer progression. This review summarizes the current knowledge of interactions between sEV and oncogenic viruses, focusing on sEV abilities to modulate the carcinogenic properties of oncoviruses.
Collapse
Affiliation(s)
- Łukasz Ważny
- Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland;
| | - Theresa L. Whiteside
- UPMC Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232, USA;
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Monika Pietrowska
- Maria Sklodowska-Curie National Research Institute of Oncology, 44-102 Gliwice, Poland;
| |
Collapse
|
28
|
Vachon L, Jean G, Milasan A, Babran S, Lacroix E, Guadarrama Bello D, Villeneuve L, Rak J, Nanci A, Mihalache-Avram T, Tardif JC, Finnerty V, Ruiz M, Boilard E, Tessier N, Martel C. Platelet extracellular vesicles preserve lymphatic endothelial cell integrity and enhance lymphatic vessel function. Commun Biol 2024; 7:975. [PMID: 39128945 PMCID: PMC11317532 DOI: 10.1038/s42003-024-06675-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 08/02/2024] [Indexed: 08/13/2024] Open
Abstract
Lymphatic vessels are essential for preventing the accumulation of harmful components within peripheral tissues, including the artery wall. Various endogenous mechanisms maintain adequate lymphatic function throughout life, with platelets being essential for preserving lymphatic vessel integrity. However, since lymph lacks platelets, their impact on the lymphatic system has long been viewed as restricted to areas where lymphatics intersect with blood vessels. Nevertheless, platelets can also exert long range effects through the release of extracellular vesicles (EVs) upon activation. We observed that platelet EVs (PEVs) are present in lymph, a compartment to which they could transfer regulatory effects of platelets. Here, we report that PEVs in lymph exhibit a distinct signature enabling them to interact with lymphatic endothelial cells (LECs). In vitro experiments show that the internalization of PEVs by LECs maintains their functional integrity. Treatment with PEVs improves lymphatic contraction capacity in atherosclerosis-prone mice. We suggest that boosting lymphatic pumping with exogenous PEVs offers a novel therapeutic approach for chronic inflammatory diseases characterized by defective lymphatics.
Collapse
Affiliation(s)
- Laurent Vachon
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Gabriel Jean
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Andreea Milasan
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Sara Babran
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Elizabeth Lacroix
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | | | | | - Janusz Rak
- McGill University and Research, Institute of the McGill University Health Centre, Montreal, Canada
- Department of Experimental Medicine, McGill University, Montreal, Canada
| | - Antonio Nanci
- Department of Stomatology, Faculty of Dental Medicine, Université de Montréal, Montreal, Canada
- Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | | | - Jean-Claude Tardif
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | | | - Matthieu Ruiz
- Department of Nutrition, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Metabolomics platform, Montreal, Canada
| | - Eric Boilard
- Centre de Recherche ARThrite - Arthrite, Recherche, Traitements, Université Laval, Québec, Québec, Canada
- Infectious and Immune Diseases Axis, Centre de Recherche du Centre Hospitalier Universitaire de Québec-Université Laval, Québec, Québec, Canada
| | - Nolwenn Tessier
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Montreal Heart Institute, Montreal, Canada
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Canada.
- Montreal Heart Institute, Montreal, Canada.
| |
Collapse
|
29
|
Cui L, Perini G, Palmieri V, De Spirito M, Papi M. Plant-Derived Extracellular Vesicles as a Novel Frontier in Cancer Therapeutics. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1331. [PMID: 39195369 DOI: 10.3390/nano14161331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/29/2024]
Abstract
Recent advancements in nanomedicine and biotechnology have unveiled the remarkable potential of plant-derived extracellular vesicles (PDEVs) as a novel and promising approach for cancer treatment. These naturally occurring nanoscale particles exhibit exceptional biocompatibility, targeted delivery capabilities, and the capacity to load therapeutic agents, positioning them at the forefront of innovative cancer therapy strategies. PDEVs are distinguished by their unique properties that facilitate tumor targeting and penetration, thereby enhancing the efficacy of drug delivery systems. Their intrinsic biological composition allows for the evasion of the immune response, enabling the efficient transport of loaded therapeutic molecules directly to tumor sites. Moreover, PDEVs possess inherent anti-cancer properties, including the ability to induce cell cycle arrest and promote apoptotic pathways within tumor cells. These vesicles have also demonstrated antimetastatic effects, inhibiting the spread and growth of cancer cells. The multifunctional nature of PDEVs allows for the simultaneous delivery of multiple therapeutic agents, further enhancing their therapeutic potential. Engineering and modification techniques, such as encapsulation, and the loading of therapeutic agents via electroporation, sonication, and incubation, have enabled the customization of PDEVs to improve their targeting efficiency and therapeutic load capacity. This includes surface modifications to increase affinity for specific tumor markers and the encapsulation of various types of therapeutic agents, such as small molecule drugs, nucleic acids, and proteins. Their plant-derived origin offers an abundant and renewable source to produce therapeutic vesicles, reducing costs and facilitating scalability for clinical applications. This review provides an in-depth analysis of the latest research on PDEVs as emerging anti-cancer agents in cancer therapy.
Collapse
Affiliation(s)
- Lishan Cui
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
| | - Giordano Perini
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Valentina Palmieri
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
- Istituto dei Sistemi Complessi, Consiglio Nazionale delle Ricerche CNR, Via dei Taurini 19, 00185 Rome, Italy
| | - Marco De Spirito
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| | - Massimiliano Papi
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCSS, 00168 Rome, Italy
| |
Collapse
|
30
|
Bollard SM, Howard J, Casalou C, Mooney L, Peters S, Sweeney C, Ajaykumar A, Triana K, McCann A, Kelly PA, Potter SM. Comparative characterisation of extracellular vesicles from canine and human plasma: a necessary step in biomarker discovery. Vet Res Commun 2024; 48:2775-2782. [PMID: 38717732 PMCID: PMC11315736 DOI: 10.1007/s11259-024-10405-0] [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: 01/24/2024] [Accepted: 05/01/2024] [Indexed: 08/10/2024]
Abstract
Extracellular Vesicles (EV) have become an interesting focus as novel biomarkers of disease and are increasingly reported upon in humans and other species. The Minimal Information for Studies of Extracellular Vesicles 2018 (MISEV2018) guidelines were published to improve rigor and standardisation within the EV field and provide a framework for the reliable isolation and characterisation of EV populations. However, this rigor and standardisation has been challenging in the area of comparative medicine. Herein we present the successful isolation of EVs from human and canine plasma using Size Exclusion Chromatography and characterise these EVs according to best international practice. This study provides evidence for the reliable comparison of human and canine EVs isolated by this approach, and a baseline description of the EVs from healthy dogs to inform future biomarker studies. This work also demonstrates that the MISEV2018 guidelines can be successfully applied to EVs isolated from canine plasma.
Collapse
Affiliation(s)
- Stephanie Marie Bollard
- UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland.
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
- Department of Plastic & Reconstructive Surgery, Mater Misercordiae University Hospital, Eccles Street, Dublin 7, Ireland.
| | - J Howard
- UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - C Casalou
- UCD Charles Institute of Dermatology, University College Dublin, Belfield, Dublin 4, Ireland
| | - L Mooney
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - S Peters
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - C Sweeney
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - A Ajaykumar
- UCD Clinical Research Centre, University College Dublin, Belfield, Dublin 4, Ireland
| | - K Triana
- UCD Clinical Research Centre, University College Dublin, Belfield, Dublin 4, Ireland
| | - A McCann
- UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | - P A Kelly
- UCD School of Veterinary Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - S M Potter
- UCD School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
- Department of Plastic & Reconstructive Surgery, Mater Misercordiae University Hospital, Eccles Street, Dublin 7, Ireland
| |
Collapse
|
31
|
Shen S, Wang C, Gu J, Song F, Wu X, Qian F, Chen X, Wang L, Peng Q, Xing Z, Gu L, Wang F, Cheng X. A Predictive Model for Initial Platinum-Based Chemotherapy Efficacy in Patients with Postoperative Epithelial Ovarian Cancer Using Tissue-Derived Small Extracellular Vesicles. J Extracell Vesicles 2024; 13:e12486. [PMID: 39104279 DOI: 10.1002/jev2.12486] [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: 01/14/2024] [Revised: 06/02/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Epithelial ovarian cancer (EOC) is an often-fatal malignancy marked by the development of resistance to platinum-based chemotherapy. Thus, accurate prediction of platinum drug efficacy is crucial for strategically selecting postoperative interventions to mitigate the risks associated with suboptimal therapeutic outcomes and adverse effects. Tissue-derived extracellular vesicles (tsEVs), in contrast to their plasma counterparts, have emerged as a powerful tool for examining distinctive attributes of EOC tissues. In this study, 4D data-independent acquisition (DIA) proteomic sequencing was performed on tsEVs obtained from 58 platinum-sensitive and 30 platinum-resistant patients with EOC. The analysis revealed a notable enrichment of differentially expressed proteins that were predominantly associated with immune-related pathways. Moreover, pivotal immune-related proteins (IRPs) were identified by LASSO regression. These factors, combined with clinical parameters selected through univariate logistic regression, were used for the construction of a model employing multivariate logistic regression. This model integrated three tsEV IRPs, CCR1, IGHV_35 and CD72, with one clinical parameter, the presence of postoperative residual lesions. Thus, this model could predict the efficacy of initial platinum-based chemotherapy in patients with EOC post-surgery, providing prognostic insights even before the initiation of chemotherapy.
Collapse
Affiliation(s)
- Shizhen Shen
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Gynecologic Oncology, School of Medicine, Women's Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Conghui Wang
- Department of Gynecologic Oncology, School of Medicine, Women's Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiaxin Gu
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Feifei Song
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaodong Wu
- Department of Gynecologic Oncology, School of Medicine, Women's Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Fangfang Qian
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xiaojing Chen
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lingfang Wang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiaohua Peng
- Department of Gynecologic Oncology, School of Medicine, Women's Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ziyu Xing
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Lingkai Gu
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Fenfen Wang
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Gynecologic Oncology, School of Medicine, Women's Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaodong Cheng
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Gynecologic Oncology, School of Medicine, Women's Hospital, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
32
|
Takei Y, Yamada M, Saito K, Kameyama Y, Aihara T, Iwasaki Y, Murakami T, Kaiho Y, Ohkoshi A, Konno D, Shiga T, Takahashi K, Ikumi S, Toyama H, Ejima Y, Yamauchi M. Endothelium-Derived Extracellular Vesicles Expressing Intercellular Adhesion Molecules Reflect Endothelial Permeability and Sepsis Severity. Anesth Analg 2024; 139:385-396. [PMID: 39008867 DOI: 10.1213/ane.0000000000006988] [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: 07/17/2024]
Abstract
BACKGROUND Currently, clinical indicators for evaluating endothelial permeability in sepsis are unavailable. Endothelium-derived extracellular vesicles (EDEVs) are emerging as biomarkers of endothelial injury. Platelet endothelial cell adhesion molecule (PECAM) and vascular endothelial (VE)-cadherin are constitutively expressed endothelial intercellular adhesion molecules that regulate intercellular adhesion and permeability. Herein, we investigated the possible association between EDEVs expressing intercellular adhesion molecules (PECAM+ or VE-cadherin+ EDEVs) and endothelial permeability and sepsis severity. METHODS Human umbilical vein endothelial cells (HUVECs) were stimulated with tumor necrosis factor alpha (TNF-α) directly or after pretreatment with permeability-modifying reagents such as angiopoietin-1, prostacyclin, or vascular endothelial growth factor (VEGF) to alter TNF-α-induced endothelial hyperpermeability. Endothelial permeability was measured using the dextran assay or transendothelial electrical resistance. Additionally, a prospective cross-sectional observational study was conducted to analyze circulating EDEV levels in patients with sepsis. EDEVs were examined in HUVEC culture supernatants or patient plasma (nonsepsis, n = 30; sepsis, n = 30; septic shock, n = 42) using flow cytometry. The Wilcoxon rank-sum test was used for comparisons between 2 groups. Comparisons among 3 or more groups were performed using the Steel-Dwass test. Spearman's test was used for correlation analysis. Statistical significance was set at P < .05. RESULTS TNF-α stimulation of HUVECs significantly increased EDEV release and endothelial permeability. Pretreatment with angiopoietin-1 or prostacyclin suppressed the TNF-α-induced increase in endothelial permeability and inhibited the release of PECAM+ and VE-cadherin+ EDEVs. In contrast, pretreatment with VEGF increased TNF-α-induced endothelial permeability and the release of PECAM+ and VE-cadherin+ EDEVs. However, pretreatment with permeability-modifying reagents did not affect the release of EDEVs expressing inflammatory stimulus-inducible endothelial adhesion molecules such as E-selectin, intracellular adhesion molecule-1, or vascular cell adhesion molecule-1. The number of PECAM+ EDEVs on admission in the septic-shock group (232 [124, 590]/μL) was significantly higher (P = .043) than that in the sepsis group (138 [77,267]/μL), with an average treatment effect of 98/μL (95% confidence interval [CI], 2-270/μL), and the number of VE-cadherin+ EDEVs in the septic-shock group (173 [76,339]/μL) was also significantly higher (P = .004) than that in the sepsis group (81 [42,159]/μL), with an average treatment effect (ATE) of 79/μL (95% CI, 19-171/μL); these EDEV levels remained elevated until day 5. CONCLUSIONS EDEVs expressing intercellular adhesion molecules (PECAM+ or VE-cadherin+ EDEVs) may reflect increased endothelial permeability and could be valuable diagnostic and prognostic markers for sepsis.
Collapse
Affiliation(s)
- Yusuke Takei
- From the Department of Anesthesiology and Perioperative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuhiro Yamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Koji Saito
- Department of Intensive Care of Medicine, Tohoku University Hospital, Sendai, Japan
| | | | - Takanori Aihara
- Department of Anesthesiology, Osaki Citizen Hospital, Sendai, Japan
| | - Yudai Iwasaki
- From the Department of Anesthesiology and Perioperative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Toru Murakami
- From the Department of Anesthesiology and Perioperative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yu Kaiho
- From the Department of Anesthesiology and Perioperative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akira Ohkoshi
- Department of Otolaryngology-Head and Neck Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Daisuke Konno
- Department of Intensive Care of Medicine, Tohoku University Hospital, Sendai, Japan
| | - Takuya Shiga
- Department of Intensive Care of Medicine, Tohoku University Hospital, Sendai, Japan
| | - Kazuhiro Takahashi
- From the Department of Anesthesiology and Perioperative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Saori Ikumi
- From the Department of Anesthesiology and Perioperative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroaki Toyama
- From the Department of Anesthesiology and Perioperative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yutaka Ejima
- From the Department of Anesthesiology and Perioperative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masanori Yamauchi
- From the Department of Anesthesiology and Perioperative Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
33
|
Lin J, Lu W, Huang B, Yang W, Wang X. The role of tissue-derived extracellular vesicles in tumor microenvironment. Tissue Cell 2024; 89:102470. [PMID: 39002287 DOI: 10.1016/j.tice.2024.102470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 07/04/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024]
Abstract
The tumor microenvironment (TME) is a highly heterogeneous ecosystem that plays critical roles in the initiation, progression, invasion, and metastasis of cancers. Extracellular vesicles (EVs), as emerging components of the host-tumor communication, are lipid-bilayer membrane structures that are secreted by most cell types into TEM and increasingly recognized as critical elements that regulate the interaction between tumor cells and their surroundings. They contain a variety of bioactive molecules, such as proteins, nucleic acids, and lipids, and participate in various pathophysiological processes while regulating intercellular communication. While many studies have focused on the EVs derived from different body fluids or cell culture supernatants, the direct isolation of tissue-derived EVs (Ti-EVs) has garnered more attention due to the advantages of tissue specificity and accurate reflection of tissue microenvironment. In this review, we summarize the protocol for isolating Ti-EVs from different tissue interstitium, discuss the role of tumor-derived and adipose tissue-derived Ti-EVs in regulating TME. In addition, we sum up the latest application of Ti-EVs as potential biomarkers for cancer diseases.
Collapse
Affiliation(s)
- Jin Lin
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Wan Lu
- Jiangxi Provincial Key Laboratory of Birth Defect for Prevention and Control, Medical Genetics Center, Jiangxi Maternal and Child Health Hospital, Nanchang, China
| | - Bo Huang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Weiming Yang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Xiaozhong Wang
- Jiangxi Province Key Laboratory of Immunology and Inflammation, Jiangxi Provincial Clinical Research Center for Laboratory Medicine, Department of Clinical Laboratory, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
34
|
Shi P, Gao H, Cheng Z, Zhao K, Chen Y, Chen X, Gan W, Zhang A, Yang C, Zhang Y. Static magnetic field-modulated mesenchymal stem cell-derived mitochondria-containing microvesicles for enhanced intervertebral disc degeneration therapy. J Nanobiotechnology 2024; 22:457. [PMID: 39085827 PMCID: PMC11290117 DOI: 10.1186/s12951-024-02728-6] [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: 03/05/2024] [Accepted: 07/17/2024] [Indexed: 08/02/2024] Open
Abstract
Intervertebral disc degeneration (IVDD) is characterized by the senescence and declining vitality of nucleus pulposus cells (NPCs), often driven by mitochondrial dysfunction. This study elucidates that mesenchymal stem cells (MSCs) play a crucial role in attenuating NPC senescence by secreting mitochondria-containing microvesicles (mitoMVs). Moreover, it demonstrates that static magnetic fields (SMF) enhance the secretion of mitoMVs by MSCs. By distinguishing mitoMV generation from exosomes, this study shifts focus to understanding the molecular mechanisms of SMF intervention, emphasizing cargo transport and plasma membrane budding processes, with RNA sequencing indicating the potential involvement of the microtubule-based transport protein Kif5b. The study further confirms the interaction between Rab22a and Kif5b, revealing Rab22a's role in sorting mitoMVs into microvesicles (MVs) and potentially mediating subsequent plasma membrane budding. Subsequent construction of a gelatin methacrylate (GelMA) hydrogel delivery system further addresses the challenges of in vivo application and verifies the substantial potential of mitoMVs in delaying IVDD. This research not only sheds light on the molecular intricacies of SMF-enhanced mitoMV secretion but also provides innovative perspectives for future IVDD therapeutic strategies.
Collapse
Affiliation(s)
- Pengzhi Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Haiyang Gao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhangrong Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Kangcheng Zhao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuhang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xianglong Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Weikang Gan
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Anran Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Cao Yang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yukun Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
35
|
Tayanloo-Beik A, Eslami A, Sarvari M, Jalaeikhoo H, Rajaeinejad M, Nikandish M, Faridfar A, Rezaei-Tavirani M, Mafi AR, Larijani B, Arjmand B. Extracellular vesicles and cancer stem cells: a deadly duo in tumor progression. Oncol Rev 2024; 18:1411736. [PMID: 39091989 PMCID: PMC11291337 DOI: 10.3389/or.2024.1411736] [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: 04/03/2024] [Accepted: 06/27/2024] [Indexed: 08/04/2024] Open
Abstract
The global incidence of cancer is increasing, with estimates suggesting that there will be 26 million new cases and 17 million deaths per year by 2030. Cancer stem cells (CSCs) and extracellular vesicles (EVs) are key to the resistance and advancement of cancer. They play a crucial role in tumor dynamics and resistance to therapy. CSCs, initially discovered in acute myeloid leukemia, are well-known for their involvement in tumor initiation, progression, and relapse, mostly because of their distinct characteristics, such as resistance to drugs and the ability to self-renew. EVs, which include exosomes, microvesicles, and apoptotic bodies, play a vital role in facilitating communication between cells within the tumor microenvironment (TME). They have a significant impact on cellular behaviors and contribute to genetic and epigenetic changes. This paper analyzes the mutually beneficial association between CSCs and EVs, emphasizing their role in promoting tumor spread and developing resistance mechanisms. This review aims to investigate the interaction between these entities in order to discover new approaches for attacking the complex machinery of cancer cells. It highlights the significance of CSCs and EVs as crucial targets in the advancement of novel cancer treatments, which helps stimulate additional research, promote progress in ideas for cancer treatment, and provide renewed optimism in the effort to reduce the burden of cancer.
Collapse
Affiliation(s)
- Akram Tayanloo-Beik
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Azin Eslami
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hasan Jalaeikhoo
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Mohsen Rajaeinejad
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
- Student Research Committee, Aja University of medical sciences, Tehran, Iran
| | - Mohsen Nikandish
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | - Ali Faridfar
- AJA Cancer Epidemiology Research and Treatment Center (AJA-CERTC), AJA University of Medical Sciences, Tehran, Iran
| | | | - Ahmad Rezazadeh Mafi
- Department of Radiation Oncology, Imam Hossein Hospital, Shaheed Beheshti Medical University, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical sciences, Tehran, Iran
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
36
|
Bano A, Yadav P, Sharma M, Verma D, Vats R, Chaudhry D, Kumar P, Bhardwaj R. Extraction and characterization of exosomes from the exhaled breath condensate and sputum of lung cancer patients and vulnerable tobacco consumers-potential noninvasive diagnostic biomarker source. J Breath Res 2024; 18:046003. [PMID: 38988301 DOI: 10.1088/1752-7163/ad5eae] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 07/03/2024] [Indexed: 07/12/2024]
Abstract
Noninvasive sample sources of exosomes, such as exhaled breath and sputum, which are in close proximity to the tumor microenvironment and may contain biomarkers indicative of lung cancer, are far more permissive than invasive sample sources for biomarker screening. Standardized exosome extraction and characterization approaches for low-volume noninvasive samples are critically needed. We isolated and characterized exhaled breath condensate (EBC) and sputum exosomes from healthy nonsmokers (n= 30), tobacco smokers (n= 30), and lung cancer patients (n= 40) and correlated the findings with invasive sample sources. EBC samples were collected by using commercially available R-Tubes. To collect sputum samples the participants were directed to take deep breaths, hold their breath, and cough in a collection container. Dynamic light scattering, nanoparticle tracking analysis, and transmission electron microscopy were used to evaluate the exosome morphology. Protein isolation, western blotting, exosome quantification via EXOCET, and Fourier transform infrared spectroscopy were performed for molecular characterization. Exosomes were successfully isolated from EBC and sputum samples, and their yields were adequate and sufficiently pure for subsequent downstream processing and characterization. The exosomes were confirmed based on their size, shape, and surface marker expression. Remarkably, cancer exosomes were the largest in size not only in the plasma subgroups, but also in the EBC (p < 0.05) and sputum (p= 0.0036) subgroups, according to our findings. A significant difference in exosome concentrations were observed between the control sub-groups (p < 0.05). Our research confirmed that exosomes can be extracted from noninvasive sources, such as EBC and sputum, to investigate lung cancer diagnostic biomarkers for research, clinical, and early detection in smokers.
Collapse
Affiliation(s)
- Afsareen Bano
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| | - Pooja Yadav
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| | - Megha Sharma
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| | - Deepika Verma
- Department of Biochemistry, All India Institute of Medical Sciences, Delhi 110029, India
| | - Ravina Vats
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| | - Dhruva Chaudhry
- Department of Pulmonary & Critical Care Medicine, Pt. B. D. Sharma PGIMS, Rohtak, Haryana 124001, India
| | - Pawan Kumar
- Department of Pulmonary & Critical Care Medicine, Pt. B. D. Sharma PGIMS, Rohtak, Haryana 124001, India
| | - Rashmi Bhardwaj
- Centre for Medical Biotechnology, Maharshi Dayanand University, Rohtak, Haryana 124001, India
| |
Collapse
|
37
|
Chen X, Zhang A, Zhao K, Gao H, Shi P, Chen Y, Cheng Z, Zhou W, Zhang Y. The role of oxidative stress in intervertebral disc degeneration: Mechanisms and therapeutic implications. Ageing Res Rev 2024; 98:102323. [PMID: 38734147 DOI: 10.1016/j.arr.2024.102323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/19/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
Oxidative stress is one of the main driving mechanisms of intervertebral disc degeneration(IDD). Oxidative stress has been associated with inflammation in the intervertebral disc, cellular senescence, autophagy, and epigenetics of intervertebral disc cells. It and the above pathological mechanisms are closely linked through the common hub reactive oxygen species(ROS), and promote each other in the process of disc degeneration and promote the development of the disease. This reveals the important role of oxidative stress in the process of IDD, and the importance and great potential of IDD therapy targeting oxidative stress. The efficacy of traditional therapy is unstable or cannot be maintained. In recent years, due to the rise of materials science, many bioactive functional materials have been applied in the treatment of IDD, and through the combination with traditional drugs, satisfactory efficacy has been achieved. At present, the research review of antioxidant bioactive materials in the treatment of IDD is not complete. Based on the existing studies, the mechanism of oxidative stress in IDD and the common antioxidant therapy were summarized in this paper, and the strategies based on emerging bioactive materials were reviewed.
Collapse
Affiliation(s)
- Xianglong Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Anran Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Kangcheng Zhao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Haiyang Gao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Pengzhi Shi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuhang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhangrong Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Wenjuan Zhou
- Department of Operating Room, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yukun Zhang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
38
|
Kaur M, Fusco S, Van den Broek B, Aseervatham J, Rostami A, Iacovitti L, Grassi C, Lukomska B, Srivastava AK. Most recent advances and applications of extracellular vesicles in tackling neurological challenges. Med Res Rev 2024; 44:1923-1966. [PMID: 38500405 DOI: 10.1002/med.22035] [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: 01/02/2024] [Revised: 02/22/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
Over the past few decades, there has been a notable increase in the global burden of central nervous system (CNS) diseases. Despite advances in technology and therapeutic options, neurological and neurodegenerative disorders persist as significant challenges in treatment and cure. Recently, there has been a remarkable surge of interest in extracellular vesicles (EVs) as pivotal mediators of intercellular communication. As carriers of molecular cargo, EVs demonstrate the ability to traverse the blood-brain barrier, enabling bidirectional communication. As a result, they have garnered attention as potential biomarkers and therapeutic agents, whether in their natural form or after being engineered for use in the CNS. This review article aims to provide a comprehensive introduction to EVs, encompassing various aspects such as their diverse isolation methods, characterization, handling, storage, and different routes for EV administration. Additionally, it underscores the recent advances in their potential applications in neurodegenerative disorder therapeutics. By exploring their unique capabilities, this study sheds light on the promising future of EVs in clinical research. It considers the inherent challenges and limitations of these emerging applications while incorporating the most recent updates in the field.
Collapse
Affiliation(s)
- Mandeep Kaur
- Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Salvatore Fusco
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Bram Van den Broek
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jaya Aseervatham
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Abdolmohamad Rostami
- Department of Neurology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Lorraine Iacovitti
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Jefferson Stem Cell and Regenerative Neuroscience Center, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Claudio Grassi
- Department of Neuroscience, Università Cattolica del Sacro Cuore, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Barbara Lukomska
- NeuroRepair Department, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Amit K Srivastava
- Department of Medicine, Cardeza Foundation for Hematologic Research, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
39
|
Rahmani A, Soleymani A, Almukhtar M, Behzad Moghadam K, Vaziri Z, Hosein Tabar Kashi A, Adabi Firoozjah R, Jafari Tadi M, Zolfaghari Dehkharghani M, Valadi H, Moghadamnia AA, Gasser RB, Rostami A. Exosomes, and the potential for exosome-based interventions against COVID-19. Rev Med Virol 2024; 34:e2562. [PMID: 38924213 DOI: 10.1002/rmv.2562] [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: 09/08/2023] [Revised: 05/17/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024]
Abstract
Since late 2019, the world has been devastated by the coronavirus disease 2019 (COVID-19) induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with more than 760 million people affected and ∼seven million deaths reported. Although effective treatments for COVID-19 are currently limited, there has been a strong focus on developing new therapeutic approaches to address the morbidity and mortality linked to this disease. An approach that is currently being investigated is the use of exosome-based therapies. Exosomes are small, extracellular vesicles that play a role in many clinical diseases, including viral infections, infected cells release exosomes that can transmit viral components, such as miRNAs and proteins, and can also include receptors for viruses that facilitate viral entry into recipient cells. SARS-CoV-2 has the ability to impact the formation, secretion, and release of exosomes, thereby potentially facilitating or intensifying the transmission of the virus among cells, tissues and individuals. Therefore, designing synthetic exosomes that carry immunomodulatory cargo and antiviral compounds are proposed to be a promising strategy for the treatment of COVID-19 and other viral diseases. Moreover, exosomes generated from mesenchymal stem cells (MSC) might be employed as cell-free therapeutic agents, as MSC-derived exosomes can diminish the cytokine storm and reverse the suppression of host anti-viral defences associated with COVID-19, and boost the repair of lung damage linked to mitochondrial activity. The present article discusses the significance and roles of exosomes in COVID-19, and explores potential future applications of exosomes in combating this disease. Despite the challenges posed by COVID-19, exosome-based therapies could represent a promising avenue for improving patient outcomes and reducing the impact of this disease.
Collapse
Affiliation(s)
- Abolfazl Rahmani
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ali Soleymani
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | | | - Kimia Behzad Moghadam
- Independent Researcher, Former University of California, San Francisco (UCSF), San Francisco, California, USA
| | - Zahra Vaziri
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Ali Hosein Tabar Kashi
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Reza Adabi Firoozjah
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Mehrdad Jafari Tadi
- Department of Cell and Molecular Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Maryam Zolfaghari Dehkharghani
- Department of Healthcare Administration and Policy, School of Public Health, University of Nevada Las Vegas (UNLV), Las Vegas, Nevada, USA
| | - Hadi Valadi
- Department of Rheumatology and Inflammation Research Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Ali Akbar Moghadamnia
- Department of Pharmacology and Toxicology, Babol University of Medical Sciences, Babol, Iran
- Pharmaceutical Sciences Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| | - Robin B Gasser
- Department of Veterinary Biosciences, Faculty of Science, Melbourne Veterinary School, The University of Melbourne, Parkville, Victoria, Australia
| | - Ali Rostami
- Infectious Diseases and Tropical Medicine Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
40
|
Jennings H, McMorrow S, Chlebeck P, Heise G, Levitsky M, Verhoven B, Kink JA, Weinstein K, Hong S, Al‐Adra DP. Normothermic liver perfusion derived extracellular vesicles have concentration-dependent immunoregulatory properties. J Extracell Vesicles 2024; 13:e12485. [PMID: 39051751 PMCID: PMC11270586 DOI: 10.1002/jev2.12485] [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: 12/24/2023] [Accepted: 06/27/2024] [Indexed: 07/27/2024] Open
Abstract
Extracellular vesicles (EVs) are major contributors to immunological responses following solid organ transplantation. Donor derived EVs are best known for their role in transplant rejection through transferring donor major histocompatibility complex proteins to recipient antigen presenting cells, a phenomenon known as ‛cross-decoration'. In contrast, donor liver-derived EVs are associated with organ tolerance in small animal models. Therefore, the cellular source of EVs and their cargo could influence their downstream immunological effects. To investigate the immunological effects of EVs released by the liver in a physiological and transplant-relevant model, we isolated EVs being produced during normothermic ex vivo liver perfusion (NEVLP), a novel method of liver storage prior to transplantation. We found EVs were produced by the liver during NEVLP, and these EVs contained multiple anti-inflammatory miRNA species. In terms of function, liver-derived EVs were able to cross-decorate allogeneic cells and suppress the immune response in allogeneic mixed lymphocyte reactions in a concentration-dependent fashion. In terms of cytokine response, the addition of 1 × 109 EVs to the mixed lymphocyte reactions significantly decreased the production of the inflammatory cytokines TNF-α, IL-10 and IFN-γ. In conclusion, we determined physiologically produced liver-derived EVs are immunologically regulatory, which has implications for their role and potential modification in solid organ transplantation.
Collapse
Affiliation(s)
- Heather Jennings
- Department of Surgery, Division of TransplantationUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Stacey McMorrow
- Department of Surgery, Division of TransplantationUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Peter Chlebeck
- Department of Surgery, Division of TransplantationUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Grace Heise
- Department of Surgery, Division of TransplantationUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Mia Levitsky
- Department of Surgery, Division of TransplantationUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Bret Verhoven
- Department of Surgery, Division of TransplantationUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - John A. Kink
- Department of MedicineUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Kristin Weinstein
- Department of Surgery, Division of TransplantationUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of PharmacyUniversity of Wisconsin‐MadisonMadisonWisconsinUSA
| | - David P. Al‐Adra
- Department of Surgery, Division of TransplantationUniversity of Wisconsin School of Medicine and Public HealthMadisonWisconsinUSA
| |
Collapse
|
41
|
Baruah H, Sarma A, Basak D, Das M. Exosome: From biology to drug delivery. Drug Deliv Transl Res 2024; 14:1480-1516. [PMID: 38252268 DOI: 10.1007/s13346-024-01515-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] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
In recent years, different advancements have been observed in nanosized drug delivery systems. Factors such as stability, safety and targeting efficiency cause hindrances in the clinical translation of these synthetic nanocarriers. Therefore, researchers employed endogenous nanocarriers like exosomes as drug delivery vehicles that have an inherent ability to target more efficiently after appropriate functionalization and show higher biocompatibility and less immunogenicity and facilitate penetration through the biological barriers more quickly than the other available carriers. Exosomes are biologically derived lipid bilayer-enclosed nanosized extracellular vesicles (size ranges from 30 to 150 nm) secreted from both prokaryotic and eukaryotic cells and appears significantly in the extracellular space. These EVs (extracellular vesicles) can exist in different sources, including mammals, plants and microorganisms. Different advanced techniques have been introduced for the isolation of exosomes to overcome the existing barriers present with conventional methods. Extensive research on the application of exosomes in therapeutic delivery for treating various diseases related to central nervous system, bone, cancer, skin, etc. has been employed. Several studies are on different stages of clinical trials, and many exosomes patents have been registered.
Collapse
Affiliation(s)
- Himakshi Baruah
- Advanced Drug Delivery Laboratory, Department of Pharmaceutics, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Guwahati, 781017, Assam, India
| | - Anupam Sarma
- Advanced Drug Delivery Laboratory, Department of Pharmaceutics, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Guwahati, 781017, Assam, India.
| | - Debojeet Basak
- Advanced Drug Delivery Laboratory, Department of Pharmaceutics, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Guwahati, 781017, Assam, India
| | - Mridusmita Das
- Advanced Drug Delivery Laboratory, Department of Pharmaceutics, School of Pharmaceutical Sciences, Girijananda Chowdhury University, Guwahati, 781017, Assam, India
| |
Collapse
|
42
|
Tiwari P, Yadav K, Shukla RP, Bakshi AK, Panwar D, Das S, Mishra PR. Extracellular vesicles-powered immunotherapy: Unleashing the potential for safer and more effective cancer treatment. Arch Biochem Biophys 2024; 756:110022. [PMID: 38697343 DOI: 10.1016/j.abb.2024.110022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024]
Abstract
Cancer treatment has seen significant advancements with the introduction of Onco-immunotherapies (OIMTs). Although some of these therapies have received approval for use, others are either undergoing testing or are still in the early stages of development. Challenges persist in making immunotherapy widely applicable to cancer treatment. To maximize the benefits of immunotherapy and minimize potential side effects, it's essential to improve response rates across different immunotherapy methods. A promising development in this area is the use of extracellular vesicles (EVs) as novel delivery systems. These small vesicles can effectively deliver immunotherapies, enhancing their effectiveness and reducing harmful side effects. This article discusses the importance of integrating nanomedicines into OIMTs, highlighting the challenges with current anti-OIMT methods. It also explores key considerations for designing nanomedicines tailored for OIMTs, aiming to improve upon existing immunotherapy techniques. Additionally, the article looks into innovative approaches like biomimicry and the use of natural biomaterial-based nanocarriers (NCs). These advancements have the potential to transform the delivery of immunotherapy. Lastly, the article addresses the challenges of moving OIMTs from theory to clinical practice, providing insights into the future of using advanced nanotechnology in cancer treatment.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Jawaharlal Nehru University, New Delhi, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Dilip Panwar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Sweety Das
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, 201002, U.P., India.
| |
Collapse
|
43
|
Lin Y, Wang Z, Liu S, Liu J, Zhang Z, Ouyang Y, Su Z, Chen D, Guo L, Luo T. Roles of extracellular vesicles on macrophages in inflammatory bone diseases. Mol Cell Biochem 2024; 479:1401-1414. [PMID: 37436653 DOI: 10.1007/s11010-023-04809-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 07/02/2023] [Indexed: 07/13/2023]
Abstract
Inflammatory bone disease is a general term for a series of diseases caused by chronic inflammation, which leads to the destruction of bone homeostasis, that is, the osteolytic activity of osteoclasts increases, and the osteogenic activity of osteoblasts decreases, leading to osteolysis. Macrophages are innate immune cell with plasticity, and their polarization is related to inflammatory bone diseases. The dynamic balance of macrophages between the M1 phenotype and the M2 phenotype affects the occurrence and development of diseases. In recent years, an increasing number of studies have shown that extracellular vesicles existing in the extracellular environment can act on macrophages, affecting the progress of inflammatory diseases. This process is realized by influencing the physiological activity or functional activity of macrophages, inducing macrophages to secrete cytokines, and playing an anti-inflammatory or pro-inflammatory role. In addition, by modifying and editing extracellular vesicles, the potential of targeting macrophages can be used to provide new ideas for developing new drug carriers for inflammatory bone diseases.
Collapse
Affiliation(s)
- Yifan Lin
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ziyan Wang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Shirong Liu
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiaohong Liu
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhiyi Zhang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuanting Ouyang
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Zhikang Su
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Ding Chen
- Guangzhou Medical University, Guangzhou, Guangdong, China
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lvhua Guo
- Guangzhou Medical University, Guangzhou, Guangdong, China.
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Tao Luo
- Guangzhou Medical University, Guangzhou, Guangdong, China.
- Department of Prosthodontics, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Affiliated Stomatology Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China.
| |
Collapse
|
44
|
Javdani-Mallak A, Salahshoori I. Environmental pollutants and exosomes: A new paradigm in environmental health and disease. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 925:171774. [PMID: 38508246 DOI: 10.1016/j.scitotenv.2024.171774] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/16/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024]
Abstract
This study investigates the intricate interplay between environmental pollutants and exosomes, shedding light on a novel paradigm in environmental health and disease. Cellular stress, induced by environmental toxicants or disease, significantly impacts the production and composition of exosomes, crucial mediators of intercellular communication. The heat shock response (HSR) and unfolded protein response (UPR) pathways, activated during cellular stress, profoundly influence exosome generation, cargo sorting, and function, shaping intercellular communication and stress responses. Environmental pollutants, particularly lipophilic ones, directly interact with exosome lipid bilayers, potentially affecting membrane stability, release, and cellular uptake. The study reveals that exposure to environmental contaminants induces significant changes in exosomal proteins, miRNAs, and lipids, impacting cellular function and health. Understanding the impact of environmental pollutants on exosomal cargo holds promise for biomarkers of exposure, enabling non-invasive sample collection and real-time insights into ongoing cellular responses. This research explores the potential of exosomal biomarkers for early detection of health effects, assessing treatment efficacy, and population-wide screening. Overcoming challenges requires advanced isolation techniques, standardized protocols, and machine learning for data analysis. Integration with omics technologies enhances comprehensive molecular analysis, offering a holistic understanding of the complex regulatory network influenced by environmental pollutants. The study underscores the capability of exosomes in circulation as promising biomarkers for assessing environmental exposure and systemic health effects, contributing to advancements in environmental health research and disease prevention.
Collapse
Affiliation(s)
- Afsaneh Javdani-Mallak
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Iman Salahshoori
- Department of Polymer Processing, Iran Polymer and Petrochemical Institute, Tehran, Iran; Department of Chemical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
45
|
Shahsavari M, Nieuwland R, van Leeuwen TG, van der Pol E. Poloxamer-188 as a wetting agent for microfluidic resistive pulse sensing measurements of extracellular vesicles. PLoS One 2024; 19:e0295849. [PMID: 38696491 PMCID: PMC11065227 DOI: 10.1371/journal.pone.0295849] [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: 07/25/2023] [Accepted: 11/30/2023] [Indexed: 05/04/2024] Open
Abstract
INTRODUCTION Microfluidic resistive pulse sensing (MRPS) can determine the concentration and size distribution of extracellular vesicles (EVs) by measuring the electrical resistance of single EVs passing through a pore. To ensure that the sample flows through the pore, the sample needs to contain a wetting agent, such as bovine serum albumin (BSA). BSA leaves EVs intact but occasionally results in unstable MRPS measurements. Here, we aim to find a new wetting agent by evaluating Poloxamer-188 and Tween-20. METHODS An EV test sample was prepared using an outdated erythrocyte blood bank concentrate. The EV test sample was diluted in Dulbecco's phosphate-buffered saline (DPBS) or DPBS containing 0.10% BSA (w/v), 0.050% Poloxamer-188 (v/v) or 1.00% Tween-20 (v/v). The effect of the wetting agents on the concentration and size distribution of EVs was determined by flow cytometry. To evaluate the precision of sample volume determination with MRPS, the interquartile range (IQR) of the particles transit time through the pore was examined. To validate that DPBS containing Poloxamer-188 yields reliable MRPS measurements, the repeatability of MRPS in measuring blood plasma samples was examined. RESULTS Flow cytometry results show that the size distribution of EVs in Tween 20, in contrast to Poloxamer-188, differs from the control measurements (DPBS and DPBS containing BSA). MRPS results show that Poloxamer-188 improves the precision of sample volume determination compared to BSA and Tween-20, because the IQR of the transit time of EVs in the test sample is 11 μs, which is lower than 56 μs for BSA and 16 μs for Tween-20. Furthermore, the IQR of the transit time of particles in blood samples with Poloxamer-188 are 14, 16, and 14 μs, which confirms the reliability of MRPS measurements. CONCLUSION The solution of 0.050% Poloxamer-188 in DPBS does not lyse EVs and results in repeatable and unimpeded MRPS measurements.
Collapse
Affiliation(s)
- Mona Shahsavari
- Amsterdam Vesicle Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Rienk Nieuwland
- Amsterdam Vesicle Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Microcirculation, Amsterdam, The Netherlands
| | - Ton G. van Leeuwen
- Amsterdam Vesicle Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Edwin van der Pol
- Amsterdam Vesicle Center, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Laboratory of Experimental Clinical Chemistry, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Biomedical Engineering and Physics, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
- Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| |
Collapse
|
46
|
Arif S, Richer M, Larochelle S, Moulin VJ. Microvesicles derived from dermal myofibroblasts modify the integrity of the blood and lymphatic barriers using distinct endocytosis pathways. JOURNAL OF EXTRACELLULAR BIOLOGY 2024; 3:e151. [PMID: 38939570 PMCID: PMC11080715 DOI: 10.1002/jex2.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 02/06/2024] [Accepted: 04/15/2024] [Indexed: 06/29/2024]
Abstract
Microvesicles (MVs) are a subtype of extracellular vesicles that can transfer biological information from their producer cells to target cells. This communication can in turn affect both normal and pathological processes. Mounting evidence has revealed that dermal wound myofibroblasts (Wmyo) produce MVs, which can transfer biomolecules impacting receptor cells such as human dermal microvascular endothelial cells (HDMECs). While the effects of MVs on HDMECs are generally well described in the literature, little is known about the transport of MVs across the HDMEC barrier, and their potential effect on the barrier integrity remains unknown. Here, we investigated these roles of Wmyo-derived MVs on two sub-populations of HDMECs, blood endothelial cells (BECs) and lymphatic endothelial cells (LECs). Using an in vitro model to mimic the endothelial barrier, we showed that MVs crossed the LEC barrier but not the BEC barrier. In addition, we demonstrated that MVs were able to influence the cell-cell junctions of HDMECs. Specifically, we observed that after internalization via the predominantly caveolin-dependent pathway, MVs induced the opening of junctions in BECs. Conversely, in LECs, MVs mainly use the macropinocytosis pathway and induce closure of these junctions. Moreover, proteins in the MV membrane were responsible for this effect, but not specifically those belonging to the VEGF family. Finally, we found that once the LEC barrier permeability was reduced by MV stimuli, MVs ceased to cross the barrier. Conversely, when the BEC barrier was rendered permeable following stimulation with MVs, they were subsequently able to cross the barrier via the paracellular pathway. Taken together, these results suggest that the study of Wmyo-derived MVs offers valuable insights into their interaction with the HDMEC barrier in the context of wound healing. They highlight the potential significance of these MVs in the overall process.
Collapse
Affiliation(s)
- Syrine Arif
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEXCentre de recherche du CHU de Québec‐Université LavalQuebecCanada
| | - Megan Richer
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEXCentre de recherche du CHU de Québec‐Université LavalQuebecCanada
| | - Sébastien Larochelle
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEXCentre de recherche du CHU de Québec‐Université LavalQuebecCanada
| | - Véronique J. Moulin
- Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEXCentre de recherche du CHU de Québec‐Université LavalQuebecCanada
- Department of Surgery, Faculty of MedicineUniversité LavalQuebecCanada
| |
Collapse
|
47
|
Shahi S, Kang T, Fonseka P. Extracellular Vesicles in Pathophysiology: A Prudent Target That Requires Careful Consideration. Cells 2024; 13:754. [PMID: 38727289 PMCID: PMC11083420 DOI: 10.3390/cells13090754] [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: 02/07/2024] [Revised: 04/22/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Extracellular vesicles (EVs) are membrane-bound particles released by cells to perform multitudes of biological functions. Owing to their significant implications in diseases, the pathophysiological role of EVs continues to be extensively studied, leading research to neglect the need to explore their role in normal physiology. Despite this, many identified physiological functions of EVs, including, but not limited to, tissue repair, early development and aging, are attributed to their modulatory role in various signaling pathways via intercellular communication. EVs are widely perceived as a potential therapeutic strategy for better prognosis, primarily through utilization as a mode of delivery vehicle. Moreover, disease-associated EVs serve as candidates for the targeted inhibition by pharmacological or genetic means. However, these attempts are often accompanied by major challenges, such as off-target effects, which may result in adverse phenotypes. This renders the clinical efficacy of EVs elusive, indicating that further understanding of the specific role of EVs in physiology may enhance their utility. This review highlights the essential role of EVs in maintaining cellular homeostasis under different physiological settings, and also discusses the various aspects that may potentially hinder the robust utility of EV-based therapeutics.
Collapse
Affiliation(s)
| | | | - Pamali Fonseka
- Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia; (S.S.); (T.K.)
| |
Collapse
|
48
|
Liu J, Lin C, Li B, Huang Q, Chen X, Tang S, Luo X, Lu R, Liu Y, Liao S, Ding X. Biochanin A inhibits endothelial dysfunction induced by IL‑6‑stimulated endothelial microparticles in Perthes disease via the NFκB pathway. Exp Ther Med 2024; 27:137. [PMID: 38476892 PMCID: PMC10928846 DOI: 10.3892/etm.2024.12425] [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: 02/09/2023] [Accepted: 12/15/2023] [Indexed: 03/14/2024] Open
Abstract
Endothelial dysfunction caused by the stimulation of endothelial microparticles (EMPs) by the inflammatory factor IL-6 is one of the pathogenic pathways associated with Perthes disease. The natural active product biochanin A (BCA) has an anti-inflammatory effect; however, whether it can alleviate endothelial dysfunction in Perthes disease is not known. The present in vitro experiments on human umbilical vein endothelial cells showed that 0-100 pg/ml IL-6-EMPs could induce endothelial dysfunction in a concentration-dependent manner, and the results of the Cell Counting Kit 8 assay revealed that, at concentrations of <20 µM, BCA had no cytotoxic effect. Reverse transcription-quantitative PCR demonstrated that BCA reduced the expression levels of the endothelial dysfunction indexes E-selectin and intercellular cell adhesion molecule-1 (ICAM-1) in a concentration-dependent manner. Immunofluorescence and western blotting illustrated that BCA increased the expression levels of zonula occludens-1 and decreased those of ICAM-1. Mechanistic studies showed that BCA inhibited activation of the NFκB pathway. In vivo experiments demonstrated that IL-6 was significantly increased in the rat model of ischemic necrosis of the femoral head, whereas BCA inhibited IL-6 production. Therefore, in Perthes disease, BCA may inhibit the NFκB pathway to suppress IL-6-EMP-induced endothelial dysfunction, and could thus be regarded as a potential treatment for Perthes disease.
Collapse
Affiliation(s)
- Jianhong Liu
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Chengsen Lin
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Boxiang Li
- Department of Orthopedics, Minzu Hospital of Guangxi Zhuang Autonomous Region, Nanning, Guangxi Zhuang Autonomous Region 530001, P.R. China
| | - Qian Huang
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xianxiang Chen
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Shengping Tang
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiaolin Luo
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Rongbin Lu
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Yun Liu
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Shijie Liao
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| | - Xiaofei Ding
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
- Guangxi Key Laboratory of Regenerative Medicine, Research Centre for Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region 530021, P.R. China
| |
Collapse
|
49
|
Cheng X, Henick BS, Cheng K. Anticancer Therapy Targeting Cancer-Derived Extracellular Vesicles. ACS NANO 2024; 18:6748-6765. [PMID: 38393984 DOI: 10.1021/acsnano.3c06462] [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] [Indexed: 02/25/2024]
Abstract
Extracellular vesicles (EVs) are natural lipid nanoparticles secreted by most types of cells. In malignant cancer, EVs derived from cancer cells contribute to its progression and metastasis by facilitating tumor growth and invasion, interfering with anticancer immunity, and establishing premetastasis niches in distant organs. In recent years, multiple strategies targeting cancer-derived EVs have been proposed to improve cancer patient outcomes, including inhibiting EV generation, disrupting EVs during trafficking, and blocking EV uptake by recipient cells. Developments in EV engineering also show promising results in harnessing cancer-derived EVs as anticancer agents. Here, we summarize the current understanding of the origin and functions of cancer-derived EVs and review the recent progress in anticancer therapy targeting these EVs.
Collapse
Affiliation(s)
- Xiao Cheng
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
- Joint Department of Biomedical EngineeringNorth Carolina State University, Raleigh, North Carolina 27606, United States
| | - Brian S Henick
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, United States
| | - Ke Cheng
- Department of Biomedical Engineering, Columbia University, New York, New York 10027, United States
| |
Collapse
|
50
|
Goryunov K, Ivanov M, Kulikov A, Shevtsova Y, Burov A, Podurovskaya Y, Zubkov V, Degtyarev D, Sukhikh G, Silachev D. A Review of the Use of Extracellular Vesicles in the Treatment of Neonatal Diseases: Current State and Problems with Translation to the Clinic. Int J Mol Sci 2024; 25:2879. [PMID: 38474125 PMCID: PMC10932115 DOI: 10.3390/ijms25052879] [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: 01/15/2024] [Revised: 02/22/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
Neonatal disorders, particularly those resulting from prematurity, pose a major challenge in health care and have a significant impact on infant mortality and long-term child health. The limitations of current therapeutic strategies emphasize the need for innovative treatments. New cell-free technologies utilizing extracellular vesicles (EVs) offer a compelling opportunity for neonatal therapy by harnessing the inherent regenerative capabilities of EVs. These nanoscale particles, secreted by a variety of organisms including animals, bacteria, fungi and plants, contain a repertoire of bioactive molecules with therapeutic potential. This review aims to provide a comprehensive assessment of the therapeutic effects of EVs and mechanistic insights into EVs from stem cells, biological fluids and non-animal sources, with a focus on common neonatal conditions such as hypoxic-ischemic encephalopathy, respiratory distress syndrome, bronchopulmonary dysplasia and necrotizing enterocolitis. This review summarizes evidence for the therapeutic potential of EVs, analyzes evidence of their mechanisms of action and discusses the challenges associated with the implementation of EV-based therapies in neonatal clinical practice.
Collapse
Affiliation(s)
- Kirill Goryunov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Mikhail Ivanov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Andrey Kulikov
- Medical Institute, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Moscow 117198, Russia;
| | - Yulia Shevtsova
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| | - Artem Burov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Yulia Podurovskaya
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Victor Zubkov
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Dmitry Degtyarev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Gennady Sukhikh
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
| | - Denis Silachev
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology, Moscow 117198, Russia; (K.G.); (M.I.); (Y.S.); (A.B.); (Y.P.); (V.Z.); (D.D.); (G.S.)
- A.N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia
| |
Collapse
|