1
|
Owliaee I, Khaledian M, Shojaeian A, Boroujeni AK. The role of small extracellular vesicles in spreading and inhibiting arthropod-borne diseases. GMS HYGIENE AND INFECTION CONTROL 2024; 19:Doc48. [PMID: 39553300 PMCID: PMC11565596 DOI: 10.3205/dgkh000503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Arthropod-borne diseases (ABDs) refer to a group of viral pathogens that affect a wide range of vertebrate hosts, including humans and non-human primates. In addition to being transmitted by mosquitoes and ticks, arthropods can also spread pathogens that cause severe human diseases. On the other hand, extracellular vesicles (EVs) can serve as cross-placental drug delivery vehicles (DDVs) to the fetus and even as antigen-presenting cells (APCs). To this end, the current review aimed to examine the role of small EVs (sEVs) in the transmission and inhibition of arthropod-borne viruses, also known as arboviruses. First, a deeper understanding of the mechanistic aspects of how these vesicles function during insect-pathogen interactions is required. Next, scalability and yield optimization must be addressed while introducing EV-based therapeutics on an industrial scale in order to implement them effectively. Finally,it is recommended to consider that sEV-mediated transfer plays a crucial role in the spread of ABDs. This is because it transfers pathogenic agents between cells within vectors, resulting in subsequent transmission to hosts. Consequently, sEVs provide potential targets for the development of novel therapies that inhibit pathogen replication or reduce arthropod vector populations. Future research in this area should emphasize how these vesicles function within host-vector systems, using advanced imaging techniques - such as high-resolution microscopy (HRM) - and cost-effective methods, in order to produce sufficient quantities for large-scale implementation.
Collapse
Affiliation(s)
- Iman Owliaee
- Department of Medical Virology, Faculty of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Mehran Khaledian
- Department of Medical Entomology, Faculty of Medicine, Hamadan, Iran
| | - Ali Shojaeian
- Research Center for Molecular Medicine, Institute of Cancer, Avicenna Health Research Institute (AHRI), Hamadan University of Medical Sciences, Hamadan, Iran
| | - Armin Khaghani Boroujeni
- Skin Disease and Leishmaniasis Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
2
|
Athira AP, Sreekanth S, Chandran A, Lahon A. Dual Role of Extracellular Vesicles as Orchestrators of Emerging and Reemerging Virus Infections. Cell Biochem Biophys 2024:10.1007/s12013-024-01495-3. [PMID: 39225901 DOI: 10.1007/s12013-024-01495-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/21/2024] [Indexed: 09/04/2024]
Abstract
Current decade witnessed the emergence and re-emergence of many viruses, which affected public health significantly. Viruses mainly utilize host cell machinery to promote its growth, and spread of these diseases. Numerous factors influence virus-host cell interactions, of which extracellular vesicles play an important role, where they transfer information both locally and distally by enclosing viral and host-derived proteins and RNAs as their cargo. Thus, they play a dual role in mediating virus infections by promoting virus dissemination and evoking immune responses in host organisms. Moreover, it acts as a double-edged sword during these infections. Advances in extracellular vesicles regulating emerging and reemerging virus infections, particularly in the context of SARS-CoV-2, Dengue, Ebola, Zika, Chikungunya, West Nile, and Japanese Encephalitis viruses are discussed in this review.
Collapse
Affiliation(s)
- A P Athira
- Department of Viral Vaccines, Institute of Advanced Virology, Bio 360 Life Science Park, Thiruvananthapuram, Kerala, India
| | - Smrithi Sreekanth
- Department of Viral Vaccines, Institute of Advanced Virology, Bio 360 Life Science Park, Thiruvananthapuram, Kerala, India
| | - Ananthu Chandran
- Department of Viral Vaccines, Institute of Advanced Virology, Bio 360 Life Science Park, Thiruvananthapuram, Kerala, India
| | - Anismrita Lahon
- Department of Viral Vaccines, Institute of Advanced Virology, Bio 360 Life Science Park, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
3
|
Mousavi MJ, Arefinia N, Azarsa M, Hoseinnezhad T, Behboudi E. MicroRNA profiles in Zika virus infection: Insights from diverse sources. Indian J Med Microbiol 2024; 51:100697. [PMID: 39103054 DOI: 10.1016/j.ijmmb.2024.100697] [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: 11/08/2023] [Revised: 07/17/2024] [Accepted: 07/30/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND Zika virus (ZIKV) stands as one of the most significant reemerging viral pathogens, linked to neurological diseases such as meningoencephalitis and congenital microcephaly. Today there are no effective therapies for treating ZIKV-infected patients. MiRNAs play a critical role in regulating cellular signaling and physiological conditions, and alterations in their profiles can bear great significance in disease progression. OBJECTIVES Despite significant progress in understanding the interaction between the ZIKV and its host since the outbreak, a more comprehensive understanding on these interactions is imperative. This review aims to summarize the studies in the field and shed light on the intricate relationship between ZIKV and its host at the molecular level. CONTENT We found that in ZIKV-infected humans, over-expression of miR-431-5p and miR-30e-5p plays a crucial role in innate immune responses and contributes to neurological damage. Additionally, in ZIKA-infected mice, we observed upregulated expression of all the targets of miR-124-3p including CCL2, IL7, IRF1, and SBNO2. Notably, other targets of this miRNA include TLR6, TNF, STAT3, and NF-kB also exhibited upregulation in the central nervous system (CNS) of infected mice. Conversely, miR-654-3p levels were reduced, correlating with the upregulation of its predicted targets including FLT3LG, LITAF, CD69, and TLR2. In the case of insects, aae-miR-286a/b-3p was predicted to target all ZIKV genotypes. This specific miRNA is typically found in ovaries and can be transferred to embryos. In conclusion, our findings suggest that host microRNAs and ZIKV-encoded microRNAs hold promise as potential targets for the diagnosis of ZIKV infections and may even serve as a therapeutic approach for managing this infectious disease.
Collapse
Affiliation(s)
- Mohammad Javad Mousavi
- Department of Hematology, School of Para-Medicine, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Nasir Arefinia
- Bio Environmental Health Hazards Research Center, Jiroft University of Medical Sciences, Jiroft, Iran.
| | - Mohammad Azarsa
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran
| | - Taraneh Hoseinnezhad
- Student Research Committee, Bushehr University of Medical Sciences, Bushehr, Iran.
| | - Emad Behboudi
- Department of Basic Medical Sciences, Khoy University of Medical Sciences, Khoy, Iran.
| |
Collapse
|
4
|
Nguyen VVT, Welsh JA, Tertel T, Choo A, van de Wakker SI, Defourny KAY, Giebel B, Vader P, Padmanabhan J, Lim SK, Nolte‐'t Hoen ENM, Verhaar MC, Bostancioglu RB, Zickler AM, Hong JM, Jones JC, EL Andaloussi S, van Balkom BWM, Görgens A. Inter-laboratory multiplex bead-based surface protein profiling of MSC-derived EV preparations identifies MSC-EV surface marker signatures. J Extracell Vesicles 2024; 13:e12463. [PMID: 38868945 PMCID: PMC11170075 DOI: 10.1002/jev2.12463] [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: 10/20/2023] [Revised: 04/15/2024] [Accepted: 05/21/2024] [Indexed: 06/14/2024] Open
Abstract
Mesenchymal stromal cells (MSCs) are promising regenerative therapeutics that primarily exert their effects through secreted extracellular vesicles (EVs). These EVs - being small and non-living - are easier to handle and possess advantages over cellular products. Consequently, the therapeutic potential of MSC-EVs is increasingly investigated. However, due to variations in MSC-EV manufacturing strategies, MSC-EV products should be considered as highly diverse. Moreover, the diverse array of EV characterisation technologies used for MSC-EV characterisation further complicates reliable interlaboratory comparisons of published data. Consequently, this study aimed to establish a common method that can easily be used by various MSC-EV researchers to characterise MSC-EV preparations to facilitate interlaboratory comparisons. To this end, we conducted a comprehensive inter-laboratory assessment using a novel multiplex bead-based EV flow cytometry assay panel. This assessment involved 11 different MSC-EV products from five laboratories with varying MSC sources, culture conditions, and EV preparation methods. Through this assay panel covering a range of mostly MSC-related markers, we identified a set of cell surface markers consistently positive (CD44, CD73 and CD105) or negative (CD11b, CD45 and CD197) on EVs of all explored MSC-EV preparations. Hierarchical clustering analysis revealed distinct surface marker profiles associated with specific preparation processes and laboratory conditions. We propose CD73, CD105 and CD44 as robust positive markers for minimally identifying MSC-derived EVs and CD11b, CD14, CD19, CD45 and CD79 as reliable negative markers. Additionally, we highlight the influence of culture medium components, particularly human platelet lysate, on EV surface marker profiles, underscoring the influence of culture conditions on resulting EV products. This standardisable approach for MSC-EV surface marker profiling offers a tool for routine characterisation of manufactured EV products in pre-clinical and clinical research, enhances the quality control of MSC-EV preparations, and hopefully paves the way for higher consistency and reproducibility in the emerging therapeutic MSC-EV field.
Collapse
Affiliation(s)
| | - Joshua A. Welsh
- Translational Nanobiology Section, Laboratory of Pathology, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
- The Measuring Stick, LtdPeterboroughUK
- Advanced Technology GroupBecton DickinsonSan JoseCaliforniaUSA
| | - Tobias Tertel
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Andre Choo
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Simonides I. van de Wakker
- Department of Cardiology, Experimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Kyra A. Y. Defourny
- Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Bernd Giebel
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Pieter Vader
- Department of Cardiology, Experimental Cardiology LaboratoryUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
- CDL ResearchUniversity Medical Center Utrecht, Utrecht UniversityUtrechtThe Netherlands
| | - Jayanthi Padmanabhan
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Sai Kiang Lim
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Esther N. M. Nolte‐'t Hoen
- Division of Infectious Diseases & Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | | | - R. Beklem Bostancioglu
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
| | - Antje M. Zickler
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| | - Jia Mei Hong
- Bioprocessing Technology Institute (BTI)Agency for Science, Technology and Research (A*STAR)SingaporeSingapore
| | - Jennifer C. Jones
- Translational Nanobiology Section, Laboratory of Pathology, National Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Samir EL Andaloussi
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| | | | - André Görgens
- Institute for Transfusion MedicineUniversity Hospital EssenUniversity of Duisburg‐EssenEssenGermany
- Division of Biomolecular and Cellular Medicine, Department of Laboratory MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Karolinska ATMP CenterANA FuturaHuddingeSweden
| |
Collapse
|
5
|
Moreno E, Ciordia S, Fátima SM, Jiménez D, Martínez-Sanz J, Vizcarra P, Ron R, Sánchez-Conde M, Bargiela R, Sanchez-Carrillo S, Moreno S, Corrales F, Ferrer M, Serrano-Villar S. Proteomic snapshot of saliva samples predicts new pathways implicated in SARS-CoV-2 pathogenesis. Clin Proteomics 2024; 21:37. [PMID: 38778280 PMCID: PMC11112864 DOI: 10.1186/s12014-024-09482-9] [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: 10/13/2023] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND Information on the microbiome's human pathways and active members that can affect SARS-CoV-2 susceptibility and pathogenesis in the salivary proteome is very scarce. Here, we studied a unique collection of samples harvested from April to June 2020 from unvaccinated patients. METHODS We compared 10 infected and hospitalized patients with severe (n = 5) and moderate (n = 5) coronavirus disease (COVID-19) with 10 uninfected individuals, including non-COVID-19 but susceptible individuals (n = 5) and non-COVID-19 and nonsusceptible healthcare workers with repeated high-risk exposures (n = 5). RESULTS By performing high-throughput proteomic profiling in saliva samples, we detected 226 unique differentially expressed (DE) human proteins between groups (q-value ≤ 0.05) out of 3376 unambiguously identified proteins (false discovery rate ≤ 1%). Major differences were observed between the non-COVID-19 and nonsusceptible groups. Bioinformatics analysis of DE proteins revealed human proteomic signatures related to inflammatory responses, central cellular processes, and antiviral activity associated with the saliva of SARS-CoV-2-infected patients (p-value ≤ 0.0004). Discriminatory biomarker signatures from human saliva include cystatins, protective molecules present in the oral cavity, calprotectins, involved in cell cycle progression, and histones, related to nucleosome functions. The expression levels of two human proteins related to protein transport in the cytoplasm, DYNC1 (p-value, 0.0021) and MAPRE1 (p-value, 0.047), correlated with angiotensin-converting enzyme 2 (ACE2) plasma activity. Finally, the proteomes of microorganisms present in the saliva samples showed 4 main microbial functional features related to ribosome functioning that were overrepresented in the infected group. CONCLUSION Our study explores potential candidates involved in pathways implicated in SARS-CoV-2 susceptibility, although further studies in larger cohorts will be necessary.
Collapse
Affiliation(s)
- Elena Moreno
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain.
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain.
| | - Sergio Ciordia
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología (CNB), CSIC, 28049, Madrid, Spain
| | - Santos Milhano Fátima
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología (CNB), CSIC, 28049, Madrid, Spain
| | - Daniel Jiménez
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
| | - Javier Martínez-Sanz
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Pilar Vizcarra
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Raquel Ron
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Matilde Sánchez-Conde
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| | - Rafael Bargiela
- Centre for Environmental Biotechnology, School of Natural Sciences, Bangor University, Bangor, LL57 2UW, UK
| | - Sergio Sanchez-Carrillo
- Instituto de Catalisis y Petroleoquimica (ICP), CSIC, 28049, Madrid, Spain
- Centro de Biologia Molecular Severo Ochoa (CBM), CSIC-UAM, 28049, Madrid, Spain
| | - Santiago Moreno
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
- Facultad de Medicina, Universidad de Alcalá de Henares, 28801, Alcalá de Henares, Madrid, Spain
| | - Fernando Corrales
- Functional Proteomics Laboratory, Centro Nacional de Biotecnología (CNB), CSIC, 28049, Madrid, Spain
| | - Manuel Ferrer
- Instituto de Catalisis y Petroleoquimica (ICP), CSIC, 28049, Madrid, Spain
| | - Sergio Serrano-Villar
- Department of Infectious Diseases, Facultad de Medicina, Hospital Universitario Ramón y Cajal, IRYCIS, Carretera de Colmenar Viejo, Km 9.100, 28034, Madrid, Spain
- CIBERINFEC, Instituto de Salud Carlos III, 28029, Madrid, Spain
| |
Collapse
|
6
|
Cui L, Zheng J, Lu Y, Lin P, Lin Y, Zheng Y, Xu R, Mai Z, Guo B, Zhao X. New frontiers in salivary extracellular vesicles: transforming diagnostics, monitoring, and therapeutics in oral and systemic diseases. J Nanobiotechnology 2024; 22:171. [PMID: 38610017 PMCID: PMC11015696 DOI: 10.1186/s12951-024-02443-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Salivary extracellular vesicles (EVs) have emerged as key tools for non-invasive diagnostics, playing a crucial role in the early detection and monitoring of diseases. These EVs surpass whole saliva in biomarker detection due to their enhanced stability, which minimizes contamination and enzymatic degradation. The review comprehensively discusses methods for isolating, enriching, quantifying, and characterizing salivary EVs. It highlights their importance as biomarkers in oral diseases like periodontitis and oral cancer, and underscores their potential in monitoring systemic conditions. Furthermore, the review explores the therapeutic possibilities of salivary EVs, particularly in personalized medicine through engineered EVs for targeted drug delivery. The discussion also covers the current challenges and future prospects in the field, emphasizing the potential of salivary EVs in advancing clinical practice and disease management.
Collapse
Affiliation(s)
- Li Cui
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Jiarong Zheng
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Ye Lu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Pei Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Yunfan Lin
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Yucheng Zheng
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Rongwei Xu
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Zizhao Mai
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China
| | - Bing Guo
- Department of Dentistry, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510080, China
| | - Xinyuan Zhao
- Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, 510280, China.
| |
Collapse
|
7
|
Margolis L. Extracellular vesicles block viral entryways. Nat Microbiol 2024; 9:882-883. [PMID: 38528149 DOI: 10.1038/s41564-024-01651-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Affiliation(s)
- Leonid Margolis
- Faculty of Natural Sciences and Medicine, Ilia State University, Tbilisi, Georgia.
| |
Collapse
|
8
|
Groß R, Reßin H, von Maltitz P, Albers D, Schneider L, Bley H, Hoffmann M, Cortese M, Gupta D, Deniz M, Choi JY, Jansen J, Preußer C, Seehafer K, Pöhlmann S, Voelker DR, Goffinet C, Pogge-von Strandmann E, Bunz U, Bartenschlager R, El Andaloussi S, Sparrer KMJ, Herker E, Becker S, Kirchhoff F, Münch J, Müller JA. Phosphatidylserine-exposing extracellular vesicles in body fluids are an innate defence against apoptotic mimicry viral pathogens. Nat Microbiol 2024; 9:905-921. [PMID: 38528146 PMCID: PMC10994849 DOI: 10.1038/s41564-024-01637-6] [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: 03/16/2023] [Accepted: 02/14/2024] [Indexed: 03/27/2024]
Abstract
Some viruses are rarely transmitted orally or sexually despite their presence in saliva, breast milk, or semen. We previously identified that extracellular vesicles (EVs) in semen and saliva inhibit Zika virus infection. However, the antiviral spectrum and underlying mechanism remained unclear. Here we applied lipidomics and flow cytometry to show that these EVs expose phosphatidylserine (PS). By blocking PS receptors, targeted by Zika virus in the process of apoptotic mimicry, they interfere with viral attachment and entry. Consequently, physiological concentrations of EVs applied in vitro efficiently inhibited infection by apoptotic mimicry dengue, West Nile, Chikungunya, Ebola and vesicular stomatitis viruses, but not severe acute respiratory syndrome coronavirus 2, human immunodeficiency virus 1, hepatitis C virus and herpesviruses that use other entry receptors. Our results identify the role of PS-rich EVs in body fluids in innate defence against infection via viral apoptotic mimicries, explaining why these viruses are primarily transmitted via PS-EV-deficient blood or blood-ingesting arthropods rather than direct human-to-human contact.
Collapse
Affiliation(s)
- Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Hanna Reßin
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Pascal von Maltitz
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Dan Albers
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Laura Schneider
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Hanna Bley
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Markus Hoffmann
- Infection Biology Unit, German Primate Center, Göttingen, Germany
- Georg-August University Göttingen, Göttingen, Germany
| | - Mirko Cortese
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Dhanu Gupta
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
- Department of Paediatrics, University of Oxford, Oxford, UK
| | - Miriam Deniz
- Clinic for Gynecology and Obstetrics, Ulm University Medical Center, Ulm, Germany
| | - Jae-Yeon Choi
- Department of Medicine, National Jewish Health, Denver, CO, USA
| | - Jenny Jansen
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Preußer
- Core Facility Extracellular Vesicles, Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
| | - Kai Seehafer
- Organisch-Chemisches Institut, Ruprecht-Karls-Universität, Heidelberg, Germany
| | - Stefan Pöhlmann
- Infection Biology Unit, German Primate Center, Göttingen, Germany
- Georg-August University Göttingen, Göttingen, Germany
| | | | - Christine Goffinet
- Institute of Virology, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
| | - Elke Pogge-von Strandmann
- Core Facility Extracellular Vesicles, Institute for Tumor Immunology, Center for Tumor Biology and Immunology, Philipps University Marburg, Marburg, Germany
| | - Uwe Bunz
- Organisch-Chemisches Institut, Ruprecht-Karls-Universität, Heidelberg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, University of Heidelberg, Heidelberg, Germany
| | - Samir El Andaloussi
- Biomolecular Medicine, Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Eva Herker
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University Marburg, Marburg, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
| | - Janis A Müller
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
- Institute of Virology, Philipps University Marburg, Marburg, Germany.
| |
Collapse
|
9
|
Latanova A, Karpov V, Starodubova E. Extracellular Vesicles in Flaviviridae Pathogenesis: Their Roles in Viral Transmission, Immune Evasion, and Inflammation. Int J Mol Sci 2024; 25:2144. [PMID: 38396820 PMCID: PMC10889558 DOI: 10.3390/ijms25042144] [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: 12/29/2023] [Revised: 02/04/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
The members of the Flaviviridae family are becoming an emerging threat for public health, causing an increasing number of infections each year and requiring effective treatment. The consequences of these infections can be severe and include liver inflammation with subsequent carcinogenesis, endothelial damage with hemorrhage, neuroinflammation, and, in some cases, death. The mechanisms of Flaviviridae pathogenesis are being actively investigated, but there are still many gaps in their understanding. Extracellular vesicles may play important roles in these mechanisms, and, therefore, this topic deserves detailed research. Recent data have revealed the involvement of extracellular vesicles in steps of Flaviviridae pathogenesis such as transmission, immune evasion, and inflammation, which is critical for disease establishment. This review covers recent papers on the roles of extracellular vesicles in the pathogenesis of Flaviviridae and includes examples of clinical applications of the accumulated data.
Collapse
Affiliation(s)
- Anastasia Latanova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia; (V.K.); (E.S.)
| | | | | |
Collapse
|
10
|
Wu J, Liu G, Jia R, Guo J. Salivary Extracellular Vesicles: Biomarkers and Beyond in Human Diseases. Int J Mol Sci 2023; 24:17328. [PMID: 38139157 PMCID: PMC10743646 DOI: 10.3390/ijms242417328] [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: 10/18/2023] [Revised: 11/29/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
Extracellular vesicles, as bioactive molecules, have been extensively studied. There are abundant studies in the literature on their biogenesis, secretion, structure, and content, and their roles in pathophysiological processes. Extracellular vesicles have been reviewed as biomarkers for use in diagnostic tools. Saliva contains many extracellular vesicles, and compared with other body fluids, it is easier to obtain in a non-invasive way, making its acquisition more easily accepted by patients. In recent years, there have been numerous new studies investigating the role of salivary extracellular vesicles as biomarkers. These studies have significant implications for future clinical diagnosis. Therefore, in this paper, we summarize and review the potential applications of salivary extracellular vesicles as biomarkers, and we also describe their other functions (e.g., hemostasis, innate immune defense) in both oral and non-oral diseases.
Collapse
Affiliation(s)
- Jialing Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; (J.W.); (G.L.); (R.J.)
| | - Gege Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; (J.W.); (G.L.); (R.J.)
| | - Rong Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; (J.W.); (G.L.); (R.J.)
| | - Jihua Guo
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan 430072, China; (J.W.); (G.L.); (R.J.)
- Department of Endodontics, School & Hospital of Stomatology, Wuhan University, Wuhan 430079, China
| |
Collapse
|
11
|
Jayasinghe M, Gao C, Yap G, Yeo BZJ, Vu LT, Tay DJW, Loh WX, Aw ZQ, Chen H, Phung DC, Hoang DV, Prajogo RC, Hooi L, Lim FQ, Pirisinu M, Mok CK, Lim KW, Tang SJ, Tan KS, Chow EKH, Chen L, Phan AT, Chu JJH, Le MTN. Red Blood Cell-Derived Extracellular Vesicles Display Endogenous Antiviral Effects and Enhance the Efficacy of Antiviral Oligonucleotide Therapy. ACS NANO 2023; 17:21639-21661. [PMID: 37852618 PMCID: PMC10655171 DOI: 10.1021/acsnano.3c06803] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023]
Abstract
The COVID-19 pandemic has resulted in a large number of fatalities and, at present, lacks a readily available curative treatment for patients. Here, we demonstrate that unmodified red blood cell-derived extracellular vesicles (RBCEVs) can inhibit SARS-CoV-2 infection in a phosphatidylserine (PS) dependent manner. Using T cell immunoglobulin mucin domain-1 (TIM-1) as an example, we demonstrate that PS receptors on cells can significantly increase the adsorption and infection of authentic and pseudotyped SARS-CoV-2 viruses. RBCEVs competitively inhibit this interaction and block TIM-1-mediated viral entry into cells. We further extend the therapeutic efficacy of this antiviral treatment by loading antisense oligonucleotides (ASOs) designed to target conserved regions of key SARS-CoV-2 genes into RBCEVs. We establish that ASO-loaded RBCEVs are efficiently taken up by cells in vitro and in vivo to suppress SARS-CoV-2 replication. Our findings indicate that this RBCEV-based SARS-CoV-2 therapeutic displays promise as a potential treatment capable of inhibiting SARS-CoV-2 entry and replication.
Collapse
Affiliation(s)
- Migara
K. Jayasinghe
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
- Department
of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Chang Gao
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
- Department
of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Gracemary Yap
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
- Department
of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Brendon Zhi Jie Yeo
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
- Department
of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Luyen Tien Vu
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
- Department
of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Douglas Jie Wen Tay
- Infectious
Diseases Translational Research Programme and Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117545, Singapore
- Biosafety
Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Wen Xiu Loh
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
- Department
of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Zhen Qin Aw
- Infectious
Diseases Translational Research Programme and Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117545, Singapore
- Biosafety
Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Huixin Chen
- Infectious
Diseases Translational Research Programme and Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117545, Singapore
- Biosafety
Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Dai Cao Phung
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
- Department
of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Dong Van Hoang
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Rebecca Carissa Prajogo
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
- Department
of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore
| | - Lissa Hooi
- Cancer
Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117599, Singapore
| | - Fang Qing Lim
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Marco Pirisinu
- Department
of Biomedical Sciences, Jockey Club College of Veterinary Medicine
and Life Sciences, City University of Hong
Kong, Tat Chee Avenue, Kowloon, Hong Kong SAR 999077, China
| | - Chee Keng Mok
- Infectious
Diseases Translational Research Programme and Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117545, Singapore
- Biosafety
Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Kah Wai Lim
- Division
of Physics & Applied Physics, School of Physical & Mathematical
Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Sze Jing Tang
- Cancer
Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117599, Singapore
| | - Kai Sen Tan
- Infectious
Diseases Translational Research Programme and Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117545, Singapore
- Biosafety
Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
| | - Edward Kai-Hua Chow
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
- Cancer
Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117599, Singapore
| | - Leilei Chen
- Cancer
Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117599, Singapore
- Department
of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117594, Singapore
| | - Anh Tuan Phan
- Division
of Physics & Applied Physics, School of Physical & Mathematical
Sciences, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Justin Jang Hann Chu
- Infectious
Diseases Translational Research Programme and Department of Microbiology
and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117545, Singapore
- Biosafety
Level 3 Core Facility, Yong Loo Lin School of Medicine, National University of Singapore, 14 Medical Drive, Singapore 117599, Singapore
- Institute
of Molecular and Cell Biology, Agency for
Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| | - Minh TN Le
- Institute
for Digital Medicine and Department of Pharmacology, Yong Loo Lin
School of Medicine, National University
of Singapore, 16 Medical Drive, Singapore 117600, Singapore
- Department
of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, Singapore 119228, Singapore
- Institute
of Molecular and Cell Biology, Agency for
Science, Technology and Research, 61 Biopolis Drive, Singapore 138673, Singapore
| |
Collapse
|
12
|
Chernyshev VS, Yashchenok A, Ivanov M, Silachev DN. Filtration-based technologies for isolation, purification and analysis of extracellular vesicles. Phys Chem Chem Phys 2023; 25:23344-23357. [PMID: 37646109 DOI: 10.1039/d3cp03129b] [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: 09/01/2023]
Abstract
The involvement of extracellular vesicles (EVs) in cellular communication with multifactorial and multifaceted biological activity has generated significant interest, highlighting their potential diagnostic and therapeutic applications. EVs are found in nearly all biological fluids creating a broad spectrum of where potential disease markers can be found for liquid biopsy development and what subtypes can be used for treatment of diseases. Complexity of biological fluids has generated a variety of different approaches for EV isolation and identification that may in one way or another be most optimal for research studies or clinical use. Each approach has its own advantages and disadvantages, significance of which can be evaluated depending on the end goal of the study. One of the methods is based on filtration which has received attention in the past years due its versatility, low cost and other advantages. Introduction of different approaches for EV capture and analysis that are based on filtration gave rise to new subcategories of filtration techniques which are presented in this overview. Miniaturization and combination of filtration-based approaches with microfluidics is also highlighted due its future prospects in healthcare, especially point-of-need technologies.
Collapse
Affiliation(s)
- Vasiliy S Chernyshev
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov, 117997, Moscow, Russia.
| | - Alexey Yashchenok
- Skoltech Center for Photonic Science and Engineering, Skolkovo Institute of Science and Technology Skolkovo Innovation Center, 121205, Moscow, Russia
| | - Mikhail Ivanov
- National Medical Research Center for Obstetrics, Gynecology and Perinatology named after Academician V.I. Kulakov, 117997, Moscow, Russia.
| | - Denis N Silachev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
| |
Collapse
|
13
|
Zhao F, Xu Y, Liu N, Lv D, Chen Y, Liu Z, Jin X, Xiao M, Lavillette D, Zhong J, Bartenschlager R, Long G. Extracellular vesicles from Zika virus-infected cells display viral E protein that binds ZIKV-neutralizing antibodies to prevent infection enhancement. EMBO J 2023; 42:e112096. [PMID: 36734074 PMCID: PMC10015360 DOI: 10.15252/embj.2022112096] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 12/26/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023] Open
Abstract
Mosquito-borne flaviviruses including Zika virus (ZIKV) represent a public health problem in some parts of the world. Although ZIKV infection is predominantly asymptomatic or associated with mild symptoms, it can lead to neurological complications. ZIKV infection can also cause antibody-dependent enhancement (ADE) of infection with similar viruses, warranting further studies of virion assembly and the function of envelope (E) protein-specific antibodies. Although extracellular vesicles (EVs) from flavivirus-infected cells have been reported to transmit infection, this interpretation is challenged by difficulties in separating EVs from flavivirions due to their similar biochemical composition and biophysical properties. In the present study, a rigorous EV-virion separation method combining sequential ultracentrifugation and affinity capture was developed to study EVs from ZIKV-infected cells. We find that these EVs do not transmit infection, but EVs display abundant E proteins which have an antigenic landscape similar to that of virions carrying E. ZIKV E-coated EVs attenuate antibody-dependent enhancement mediated by ZIKV E-specific and DENV-cross-reactive antibodies in both cell culture and mouse models. We thus report an alternative route for Flavivirus E protein secretion. These results suggest that modulation of E protein release via virions and EVs may present a new approach to regulating flavivirus-host interactions.
Collapse
Affiliation(s)
- Fanfan Zhao
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Biosafety Level 3 LaboratoryShanghai Institute of Infectious Disease and Biosecurity, Fudan UniversityShanghaiChina
- CAS Key Laboratory of Molecular Virology and ImmunologyInstitut Pasteur of Shanghai, Chinese Academy of SciencesShanghaiChina
| | - Yongfen Xu
- CAS Key Laboratory of Molecular Virology and ImmunologyInstitut Pasteur of Shanghai, Chinese Academy of SciencesShanghaiChina
| | - Na Liu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Biosafety Level 3 LaboratoryShanghai Institute of Infectious Disease and Biosecurity, Fudan UniversityShanghaiChina
| | - Dawei Lv
- CAS Key Laboratory of Molecular Virology and ImmunologyInstitut Pasteur of Shanghai, Chinese Academy of SciencesShanghaiChina
| | - Yujie Chen
- CAS Key Laboratory of Molecular Virology and ImmunologyInstitut Pasteur of Shanghai, Chinese Academy of SciencesShanghaiChina
| | - Zhi Liu
- CAS Key Laboratory of Molecular Virology and ImmunologyInstitut Pasteur of Shanghai, Chinese Academy of SciencesShanghaiChina
| | - Xia Jin
- CAS Key Laboratory of Molecular Virology and ImmunologyInstitut Pasteur of Shanghai, Chinese Academy of SciencesShanghaiChina
| | - Mingbing Xiao
- Department of Gastroenterology and Research Center of Clinical MedicineAffiliated Hospital of Nantong UniversityNantongChina
| | - Dimitri Lavillette
- CAS Key Laboratory of Molecular Virology and ImmunologyInstitut Pasteur of Shanghai, Chinese Academy of SciencesShanghaiChina
| | - Jin Zhong
- CAS Key Laboratory of Molecular Virology and ImmunologyInstitut Pasteur of Shanghai, Chinese Academy of SciencesShanghaiChina
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular VirologyHeidelberg UniversityHeidelbergGermany
- German Center for Infectious Diseases, Heidelberg Partner SiteHeidelbergGermany
| | - Gang Long
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Medical College, Biosafety Level 3 LaboratoryShanghai Institute of Infectious Disease and Biosecurity, Fudan UniversityShanghaiChina
- CAS Key Laboratory of Molecular Virology and ImmunologyInstitut Pasteur of Shanghai, Chinese Academy of SciencesShanghaiChina
| |
Collapse
|
14
|
Liang JQ, Xie MY, Hou LJ, Wang HL, Luo JY, Sun JJ, Xi QY, Jiang QY, Chen T, Zhang YL. miRNAs derived from milk small extracellular vesicles inhibit porcine epidemic diarrhea virus infection. Antiviral Res 2023; 212:105579. [PMID: 36907442 DOI: 10.1016/j.antiviral.2023.105579] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/12/2023]
Abstract
Porcine epidemic diarrhea virus (PEDV), a member of the genus Alphacoronavirus in the family Coronaviridae, causes acute diarrhea and/or vomiting, dehydration, and high mortality in neonatal piglets. It has caused huge economic losses to animal husbandry worldwide. Current commercial PEDV vaccines do not provide enough protection against variant and evolved virus strains. No specific drugs are available to treat PEDV infection. The development of more effective therapeutic anti-PEDV agents is urgently needed. Our previous study suggested that porcine milk small extracellular vesicles (sEV) facilitate intestinal tract development and prevent lipopolysaccharide-induced intestinal injury. However, the effects of milk sEV during viral infection remain unclear. Our study found that porcine milk sEV, which was isolated and purified by differential ultracentrifugation, could inhibit PEDV replication in IPEC-J2 and Vero cells. Simultaneously, we constructed a PEDV infection model for piglet intestinal organoids and found that milk sEV also inhibited PEDV infection. Subsequently, in vivo experiments showed that milk sEV pre-feeding exerted robust protection of piglets from PEDV-induced diarrhea and mortality. Strikingly, we found that the miRNAs extracted from milk sEV inhibited PEDV infection. miRNA-seq, bioinformatics analysis, and experimental verification demonstrated that miR-let-7e and miR-27b, which were identified in milk sEV targeted PEDV N and host HMGB1, suppressed viral replication. Taken together, we revealed the biological function of milk sEV in resisting PEDV infection and proved its cargo miRNAs, miR-let-7e and miR-27b, possess antiviral functions. This study is the first description of the novel function of porcine milk sEV in regulating PEDV infection. It provides a better understanding of milk sEV resistance to coronavirus infection, warranting further studies to develop sEV as an attractive antiviral.
Collapse
Affiliation(s)
- Jia Qi Liang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Mei-Ying Xie
- Guangdong Eco-Engineering Polytechnic, Guangzhou, Guangdong, 510520, China
| | - Lian-Jie Hou
- The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan, Guangdong, 511518, China
| | - Hai-Long Wang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jun-Yi Luo
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Jia-Jie Sun
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qian-Yun Xi
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Qing-Yan Jiang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, 510642, China
| | - Ting Chen
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| | - Yong-Liang Zhang
- College of Animal Science, Guangdong Province Key Laboratory of Animal Nutritional Regulation, and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| |
Collapse
|
15
|
Venkatesan G, Wan Ab Rahman WS, Shahidan WNS, Iberahim S, Muhd Besari@Hashim AB. Plasma-derived exosomal miRNA as potential biomarker for diagnosis and prognosis of vector-borne diseases: A review. Front Microbiol 2023; 14:1097173. [PMID: 37125151 PMCID: PMC10133507 DOI: 10.3389/fmicb.2023.1097173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 03/21/2023] [Indexed: 05/02/2023] Open
Abstract
Early disease diagnosis is critical for better management and treatment outcome of patients. Therefore, diagnostic methods should ideally be accurate, consistent, easy to perform at low cost and preferably non-invasive. In recent years, various biomarkers have been studied for the detection of cardiovascular diseases, cerebrovascular diseases, infectious diseases, diabetes mellitus and malignancies. Exosomal microRNA (miRNA) are small non-coding RNA molecules that influence gene expression after transcription. Previous studies have shown that these types of miRNAs can potentially be used as biomarkers for cancers of the breast and colon, as well as diffuse large B-cell lymphoma. It may also be used to indicate viral and bacterial infections, such as the human immunodeficiency virus (HIV), tuberculosis and hepatitis. However, its use in the diagnosis of vector-borne diseases is rather limited. Therefore, this review aims to introduce several miRNAs derived from exosomal plasma that may potentially serve as a disease biomarker due to the body's immune response, with special focus on the early detection of vector-borne diseases.
Collapse
Affiliation(s)
| | - Wan Suriana Wan Ab Rahman
- School of Dental Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
- *Correspondence: Wan Suriana Wan Ab Rahman,
| | | | - Salfarina Iberahim
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Alwi bin Muhd Besari@Hashim
- Department of Internal Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
16
|
Tsuchiya H. The Oral Cavity Potentially Serving as a Reservoir for SARS-CoV-2 but Not Necessarily Facilitating the Spread of COVID-19 in Dental Practice. Eur J Dent 2022. [DOI: 10.1055/s-0042-1757909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
AbstractIntraoral tissues, secretions, and microenvironments may provide severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) with the conditions necessary for viral cellular entry and inhabitation. The aim of the present study is to overview the oral cavity that potentially serves as a reservoir for SARS-CoV-2, and then discuss the possibility that such oral cavity facilitates the spread of coronavirus disease 2019 (COVID-19) in dental practice. Articles were retrieved from PubMed/Medline, LitCovid, ProQuest, Google Scholar, and preprint medRxiv databases. Results of the literature search indicated that SARS-CoV-2 host cell entry-relevant receptor and virus/cell membrane fusion mediators are expressed in major and minor salivary glands, tongue, taste bud, periodontal tissue, and dental pulp, which would be a target and reservoir for SARS-CoV-2. SARS-CoV-2 is present in saliva and gingival crevicular fluid of COVID-19 patients. These secretions would contaminate dental aerosol and droplet with SARS-CoV-2. SARS-CoV-2 inhabits periodontal pocket, gingival sulcus, and dental caries lesion, which could provide SARS-CoV-2 with a habitat. SARS-CoV-2 ribonucleic acid is preserved in dental calculus, which may inform of the previous infection with SARS-CoV-2. Despite involvement of the oral cavity in SARS-CoV-2 transmission and infection, to date, there have been no clusters of COVID-19 in dental practice. Dental settings are much less likely to facilitate the spread of COVID-19 compared with general medical settings, which may be explained by the situation of dentistry that the number of patients to visit dental offices/clinics was decreased during the COVID-19 pandemic, the characteristics of dentistry that dental professionals have maintained high awareness of viral infection prevention, adhered to a strict protocol for infection control, and been using personal protective equipment for a long time, the experimental results that dental devices generate only small amounts of aerosol responsible for the airborne viral transmission, irrigant from the dental unit contributes to the aerosol microbiota much rather than saliva, and the commonly used evacuation or suction system effectively reduces aerosol and droplet generation, and the possibility that human saliva exhibits the antiviral activity and the property to inhibit SARS-CoV-2 infection. It is considered that dental treatment and oral health care can be delivered safely in the COVID-19 era.
Collapse
Affiliation(s)
- Hironori Tsuchiya
- Department of Dental Basic Education, Asahi University School of Dentistry, Mizuho, Gifu, Japan
| |
Collapse
|
17
|
Tey SK, Yam JWP. The importance of activated TMPRSS2 in the proviral role of small extracellular vesicles in SARS-CoV-2 infection. J Extracell Vesicles 2022; 11:e12296. [PMID: 36541555 PMCID: PMC9769082 DOI: 10.1002/jev2.12296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Sze Keong Tey
- Department of SurgerySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| | - Judy Wai Ping Yam
- Department of PathologySchool of Clinical MedicineLi Ka Shing Faculty of MedicineThe University of Hong KongHong KongChina
| |
Collapse
|
18
|
Berry F, Morin‐Dewaele M, Majidipur A, Jamet T, Bartier S, Ignjatovic E, Toniutti D, Gaspar Lopes J, Soyeux‐Porte P, Maillé P, Saldana C, Brillet R, Ahnou N, Softic L, Couturaud B, Huet É, Ahmed‐Belkacem A, Fourati S, Louis B, Coste A, Béquignon É, de la Taille A, Destouches D, Vacherot F, Pawlotsky J, Firlej V, Bruscella P. Proviral role of human respiratory epithelial cell-derived small extracellular vesicles in SARS-CoV-2 infection. J Extracell Vesicles 2022; 11:e12269. [PMID: 36271885 PMCID: PMC9587708 DOI: 10.1002/jev2.12269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/20/2022] [Accepted: 09/14/2022] [Indexed: 11/06/2022] Open
Abstract
Small Extracellular Vesicles (sEVs) are 50-200 nm in diameter vesicles delimited by a lipid bilayer, formed within the endosomal network or derived from the plasma membrane. They are secreted in various biological fluids, including airway nasal mucus. The goal of this work was to understand the role of sEVs present in the mucus (mu-sEVs) produced by human nasal epithelial cells (HNECs) in SARS-CoV-2 infection. We show that uninfected HNECs produce mu-sEVs containing SARS-CoV-2 receptor ACE2 and activated protease TMPRSS2. mu-sEVs cleave prefusion viral Spike proteins at the S1/S2 boundary, resulting in higher proportions of prefusion S proteins exposing their receptor binding domain in an 'open' conformation, thereby facilitating receptor binding at the cell surface. We show that the role of nasal mu-sEVs is to complete prefusion Spike priming performed by intracellular furin during viral egress from infected cells. This effect is mediated by vesicular TMPRSS2 activity, rendering SARS-CoV-2 virions prone to entry into target cells using the 'early', TMPRSS2-dependent pathway instead of the 'late', cathepsin-dependent route. These results indicate that prefusion Spike priming by mu-sEVs in the nasal cavity plays a role in viral tropism. They also show that nasal mucus does not protect from SARS-CoV-2 infection, but instead facilitates it.
Collapse
Affiliation(s)
- François Berry
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance
| | - Margot Morin‐Dewaele
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance
| | - Amene Majidipur
- Team “Therapeutic Resistance in Prostate Cancer” (TRePCa)Univ Paris Est CreteilCréteilFrance
| | - Thibaud Jamet
- Team “Therapeutic Resistance in Prostate Cancer” (TRePCa)Univ Paris Est CreteilCréteilFrance
| | - Sophie Bartier
- Department of ENT and Cervico‐Facial SurgeryAP‐HP, Centre Hospitalier Intercommunal de CréteilCréteilFrance,Department of ENT and Cervico‐Facial SurgeryAP‐HP, Centre Hospitalier Universitaire Henri MondorCréteilFrance,Department of PulmonologyAP‐HP, Centre Hospitalier Universitaire Henri MondorCréteilFrance,Institut Mondor de Recherche Biomédicale, INSERM U955, CNRS EMR 7000, Team “Biomechanics and Respiratory System”Univ Paris Est CreteilCréteilFrance
| | - Eva Ignjatovic
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance
| | - Donatella Toniutti
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance
| | - Jeanne Gaspar Lopes
- Team “Therapeutic Resistance in Prostate Cancer” (TRePCa)Univ Paris Est CreteilCréteilFrance
| | - Pascale Soyeux‐Porte
- Team “Therapeutic Resistance in Prostate Cancer” (TRePCa)Univ Paris Est CreteilCréteilFrance
| | - Pascale Maillé
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance,Department of PathologyAP‐HP, Centre Hospitalier Universitaire Henri MondorCréteilFrance
| | - Carolina Saldana
- Team “Therapeutic Resistance in Prostate Cancer” (TRePCa)Univ Paris Est CreteilCréteilFrance,Department of OncologyAP‐HP, Centre Hospitalier Universitaire Henri MondorCréteilFrance
| | - Rozenn Brillet
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance
| | - Nazim Ahnou
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance
| | - Laurent Softic
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance
| | - Benoit Couturaud
- Institute of Chemistry and Materials (ICMPE)Univ Paris Est Creteil, CNRS UMR7182CréteilFrance
| | - Éric Huet
- Team “Therapeutic Resistance in Prostate Cancer” (TRePCa)Univ Paris Est CreteilCréteilFrance
| | - Abdelhakim Ahmed‐Belkacem
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance
| | - Slim Fourati
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance,Department of VirologyAP‐HP, Centre Hospitalier Universitaire Henri MondorCréteilFrance
| | - Bruno Louis
- Institut Mondor de Recherche Biomédicale, INSERM U955, CNRS EMR 7000, Team “Biomechanics and Respiratory System”Univ Paris Est CreteilCréteilFrance
| | - André Coste
- Department of ENT and Cervico‐Facial SurgeryAP‐HP, Centre Hospitalier Intercommunal de CréteilCréteilFrance,Department of ENT and Cervico‐Facial SurgeryAP‐HP, Centre Hospitalier Universitaire Henri MondorCréteilFrance,Department of PulmonologyAP‐HP, Centre Hospitalier Universitaire Henri MondorCréteilFrance,Institut Mondor de Recherche Biomédicale, INSERM U955, CNRS EMR 7000, Team “Biomechanics and Respiratory System”Univ Paris Est CreteilCréteilFrance
| | - Émilie Béquignon
- Department of ENT and Cervico‐Facial SurgeryAP‐HP, Centre Hospitalier Intercommunal de CréteilCréteilFrance,Department of ENT and Cervico‐Facial SurgeryAP‐HP, Centre Hospitalier Universitaire Henri MondorCréteilFrance,Department of PulmonologyAP‐HP, Centre Hospitalier Universitaire Henri MondorCréteilFrance,Institut Mondor de Recherche Biomédicale, INSERM U955, CNRS EMR 7000, Team “Biomechanics and Respiratory System”Univ Paris Est CreteilCréteilFrance
| | - Alexandre de la Taille
- Team “Therapeutic Resistance in Prostate Cancer” (TRePCa)Univ Paris Est CreteilCréteilFrance,Department of UrologyAP‐HP, Centre Hospitalier Universitaire Henri MondorCréteilFrance
| | - Damien Destouches
- Team “Therapeutic Resistance in Prostate Cancer” (TRePCa)Univ Paris Est CreteilCréteilFrance
| | - Francis Vacherot
- Team “Therapeutic Resistance in Prostate Cancer” (TRePCa)Univ Paris Est CreteilCréteilFrance
| | - Jean‐Michel Pawlotsky
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance,Department of VirologyAP‐HP, Centre Hospitalier Universitaire Henri MondorCréteilFrance
| | - Virginie Firlej
- Team “Therapeutic Resistance in Prostate Cancer” (TRePCa)Univ Paris Est CreteilCréteilFrance
| | - Patrice Bruscella
- Institut Mondor de Recherche Biomédicale, INSERM U955, Team “Viruses, Hepatology, Cancer”Univ Paris Est CreteilCréteilFrance
| |
Collapse
|
19
|
Tertel T, Tomić S, Đokić J, Radojević D, Stevanović D, Ilić N, Giebel B, Kosanović M. Serum-derived extracellular vesicles: Novel biomarkers reflecting the disease severity of COVID-19 patients. J Extracell Vesicles 2022; 11:e12257. [PMID: 35979935 PMCID: PMC9451525 DOI: 10.1002/jev2.12257] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 06/10/2022] [Accepted: 07/26/2022] [Indexed: 11/09/2022] Open
Abstract
COVID-19 is characterized by a wide spectrum of disease severity, whose indicators and underlying mechanisms need to be identified. The role of extracellular vesicles (EVs) in COVID-19 and their biomarker potential, however, remains largely unknown. Aiming to identify specific EV signatures of patients with mild compared to severe COVID-19, we characterized the EV composition of 20 mild and 26 severe COVID-19 patients along with 16 sex and age-matched healthy donors with a panel of eight different antibodies by imaging flow cytometry (IFCM). We correlated the obtained data with 37 clinical, prerecorded biochemical and immunological parameters. Severe patients' sera contained increased amounts of CD13+ and CD82+ EVs, which positively correlated with IL-6-producing and circulating myeloid-derived suppressor cells (MDSCs) and with the serum concentration of proinflammatory cytokines, respectively. Sera of mild COVID-19 patients contained more HLA-ABC+ EVs than sera of the healthy donors and more CD24+ EVs than severe COVID-19 patients. Their increased abundance negatively correlated with disease severity and accumulation of MDSCs, being considered as key drivers of immunopathogenesis in COVID-19. Altogether, our results support the potential of serum EVs as powerful biomarkers for COVID-19 severity and pave the way for future investigations aiming to unravel the role of EVs in COVID-19 progression.
Collapse
Affiliation(s)
- Tobias Tertel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Sergej Tomić
- Institute for the Application of Nuclear Energy, INEP, University of Belgrade, Zemun-Belgrade, Serbia
| | - Jelena Đokić
- Institute of Molecular Genetics and Genetic Engineering (IMGGI), University of Belgrade, Belgrade, Serbia
| | - Dušan Radojević
- Institute of Molecular Genetics and Genetic Engineering (IMGGI), University of Belgrade, Belgrade, Serbia
| | - Dejan Stevanović
- Clinical Hospital Center Zemun, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nataša Ilić
- Institute for the Application of Nuclear Energy, INEP, University of Belgrade, Zemun-Belgrade, Serbia
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Maja Kosanović
- Institute for the Application of Nuclear Energy, INEP, University of Belgrade, Zemun-Belgrade, Serbia
| |
Collapse
|
20
|
Yenuganti VR, Afroz S, Khan RA, Bharadwaj C, Nabariya DK, Nayak N, Subbiah M, Chintala K, Banerjee S, Reddanna P, Khan N. Milk exosomes elicit a potent anti-viral activity against dengue virus. J Nanobiotechnology 2022; 20:317. [PMID: 35794557 PMCID: PMC9258094 DOI: 10.1186/s12951-022-01496-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 06/08/2022] [Indexed: 11/10/2022] Open
Abstract
Background Exosomes are nano-sized vesicles secreted by various cells into the intra and extracellular space and hence is an integral part of biological fluids including milk. In the last few decades, many research groups have proved the potential of milk exosomes as a sustainable, economical and non-immunogenic drug delivery and therapeutic agent against different pathological conditions. However, its anti-viral properties still remain to be unearthed. Methods Here, we have been able to isolate, purify and characterize the milk derived exosomes from Cow (CME) and Goat (GME) and further studied its antiviral properties against Dengue virus (DENV), Newcastle Disease Virus strain Komarov (NDV-K) and Human Immunodeficiency Virus (HIV-1) using an in-vitro infection system. Results TEM, NTA and DLS analysis validated the appropriate size of the isolated cow and goat milk exosomes (30–150 nm). Real-time PCR and immunoblotting results confirmed the presence of several milk exosomal miRNAs and protein markers. Our findings suggest that GME significantly decreased the infectivity of DENV. In addition, we confirmed that GME significantly reduces DENV replication and reduced the secretion of mature virions. Furthermore, heat inactivation of GME did not show any inhibition on DENV infection, replication, and secretion of mature virions. RNase treatment of GME abrogates the anti-viral properties indicating direct role of exosomes in DENV inhibition. In addition GME inhibited the infectivity of NDV-K, but not HIV-1, suggesting that the GME mediated antiviral activity might be virus specific. Conclusion This study demonstrates the anti-viral properties of milk exosomes and opens new avenues for the development of exosome-based therapies to treat viral diseases. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01496-5.
Collapse
|
21
|
Ali NB, Abdull Razis AF, Ooi DJ, Chan KW, Ismail N, Foo JB. Theragnostic Applications of Mammal and Plant-Derived Extracellular Vesicles: Latest Findings, Current Technologies, and Prospects. Molecules 2022; 27:3941. [PMID: 35745063 PMCID: PMC9228370 DOI: 10.3390/molecules27123941] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/26/2022] [Accepted: 02/11/2022] [Indexed: 11/16/2022] Open
Abstract
The way cells communicate is not fully understood. However, it is well-known that extracellular vesicles (EVs) are involved. Researchers initially thought that EVs were used by cells to remove cellular waste. It is now clear that EVs function as signaling molecules released by cells to communicate with one another, carrying a cargo representing the mother cell. Furthermore, these EVs can be found in all biological fluids, making them the perfect non-invasive diagnostic tool, as their cargo causes functional changes in the cells upon receiving, unlike synthetic drug carriers. EVs last longer in circulation and instigate minor immune responses, making them the perfect drug carrier. This review sheds light on the latest development in EVs isolation, characterization and, application as therapeutic cargo, novel drug loading techniques, and diagnostic tools. We also address the advancement in plant-derived EVs, their characteristics, and applications; since plant-derived EVs only recently gained focus, we listed the latest findings. Although there is much more to learn about, EV is a wide field of research; what scientists have discovered so far is fascinating. This paper is suitable for those new to the field seeking to understand EVs and those already familiar with it but wanting to review the latest findings.
Collapse
Affiliation(s)
- Nada Basheir Ali
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
| | - Ahmad Faizal Abdull Razis
- Department of Food Science, Faculty of Food Science and Technology, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia;
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (K.W.C.); (N.I.)
| | - Der Jiun Ooi
- Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University, Bandar Saujana Putra, Jenjarom 42610, Selangor, Malaysia
| | - Kim Wei Chan
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (K.W.C.); (N.I.)
| | - Norsharina Ismail
- Natural Medicines and Products Research Laboratory, Institute of Bioscience, Universiti Putra Malaysia (UPM), Serdang 43400, Selangor, Malaysia; (K.W.C.); (N.I.)
| | - Jhi Biau Foo
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor’s University, Subang Jaya 47500, Selangor, Malaysia;
| |
Collapse
|
22
|
Lim K, Nishide G, Yoshida T, Watanabe‐Nakayama T, Kobayashi A, Hazawa M, Hanayama R, Ando T, Wong RW. Millisecond dynamic of SARS-CoV-2 spike and its interaction with ACE2 receptor and small extracellular vesicles. J Extracell Vesicles 2021; 10:e12170. [PMID: 34874124 PMCID: PMC8650025 DOI: 10.1002/jev2.12170] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 09/21/2021] [Accepted: 11/08/2021] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 spike protein (S) binds to human angiotensin-converting enzyme 2 (hACE2), allowing virus to dock on cell membrane follow by viral entry. Here, we use high-speed atomic force microscopy (HS-AFM) for real-time visualization of S, and its interaction with hACE2 and small extracellular vesicles (sEVs). Results show conformational heterogeneity of S, flexibility of S stalk and receptor-binding domain (RBD), and pH/temperature-induced conformational change of S. S in an S-ACE2 complex appears as an all-RBD up conformation. The complex acquires a distinct topology upon acidification. S and S2 subunit demonstrate different membrane docking mechanisms on sEVs. S-hACE2 interaction facilitates S to dock on sEVs, implying the feasibility of ACE2-expressing sEVs for viral neutralization. In contrary, S2 subunit docks on lipid layer and enters sEV using its fusion peptide, mimicking the viral entry scenario. Altogether, our study provides a platform that is suitable for real-time visualization of various entry inhibitors, neutralizing antibodies, and sEV-based decoy in blocking viral entry. Teaser: Comprehensive observation of SARS-CoV-2 spike and its interaction with receptor ACE2 and sEV-based decoy in real time using HS-AFM.
Collapse
Affiliation(s)
- Keesiang Lim
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Goro Nishide
- Division of Nano Life Science in the Graduate School of Frontier Science InitiativeWISE Program for Nano‐Precision MedicineScience and TechnologyKanazawa UniversityKanazawaIshikawaJapan
| | - Takeshi Yoshida
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Department of ImmunologyKanazawa University Graduate School of Medical SciencesKanazawaIshikawaJapan
| | | | - Akiko Kobayashi
- Cell‐Bionomics Research UnitInstitute for Frontier Science Initiative (INFINITI)Kanazawa UniversityKanazawaIshikawaJapan
| | - Masaharu Hazawa
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Cell‐Bionomics Research UnitInstitute for Frontier Science Initiative (INFINITI)Kanazawa UniversityKanazawaIshikawaJapan
| | - Rikinari Hanayama
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Department of ImmunologyKanazawa University Graduate School of Medical SciencesKanazawaIshikawaJapan
| | - Toshio Ando
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
| | - Richard W. Wong
- WPI‐Nano Life Science InstituteKanazawa UniversityKanazawaIshikawaJapan
- Cell‐Bionomics Research UnitInstitute for Frontier Science Initiative (INFINITI)Kanazawa UniversityKanazawaIshikawaJapan
| |
Collapse
|
23
|
Wong FC, Ye L, Demir IE, Kahlert C. Schwann cell-derived exosomes: Janus-faced mediators of regeneration and disease. Glia 2021; 70:20-34. [PMID: 34519370 DOI: 10.1002/glia.24087] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 08/23/2021] [Accepted: 08/25/2021] [Indexed: 12/20/2022]
Abstract
The phenotypic plasticity of Schwann cells (SCs) has contributed to the regenerative potential of the peripheral nervous system (PNS), but also pathological processes. This double-sided effect has led to an increasing attention to the role of extracellular vesicles (EVs) or exosomes in SCs to examine the intercellular communication between SCs and their surroundings. Here, we first describe the current knowledge of SC and EV biology, which forms the basis for the updates on advances in SC-derived exosomes research. We seek to explore in-depth the exosome-mediated molecular mechanisms involved in the regulation of SCs and their microenvironment. This review concludes with potential applications of SC-derived exosomes as delivery vehicles for therapeutics and biomarkers. The goal of this review is to emphasize the crucial role of SC-derived exosomes in the functional integration of the PNS, highlighting an emerging area in which there is much to explore and re-explore.
Collapse
Affiliation(s)
- Fang Cheng Wong
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Linhan Ye
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.,Germany German Cancer Consortium (DKTK), Partner Site, Munich, Germany.,CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany
| | - Ihsan Ekin Demir
- Department of Surgery, Klinikum rechts der Isar, Technical University of Munich, School of Medicine, Munich, Germany.,Germany German Cancer Consortium (DKTK), Partner Site, Munich, Germany.,Department of General Surgery, HPB-Unit, School of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey.,CRC 1321 Modelling and Targeting Pancreatic Cancer, Munich, Germany.,Else Kröner Clinician Scientist Professor for "Translational Pancreatic Surgery
| | - Christoph Kahlert
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany.,National Center for Tumor Diseases (NCT/UCC), Dresden, Germany
| |
Collapse
|
24
|
Jackson KK, Powell RR, Bruce TF, Marcus RK. Rapid isolation of extracellular vesicles from diverse biofluid matrices via capillary-channeled polymer fiber solid-phase extraction micropipette tips. Analyst 2021; 146:4314-4325. [PMID: 34105528 DOI: 10.1039/d1an00373a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Extracellular vesicles (EVs) play essential roles in biological systems based on their ability to carry genetic and protein cargos, intercede in cellular communication and serve as vectors in intercellular transport. As such, EVs are species of increasing focus from the points of view of fundamental biochemistry, clinical diagnostics, and therapeutics delivery. Of particular interest are 30-200 nm EVs called exosomes, which have demonstrated high potential for use in diagnostic and targeted delivery applications. The ability to collect exosomes from patient biofluid samples would allow for comprehensive yet remote diagnoses to be performed. While several exosome isolation methods are in common use, they generally produce low recoveries, whose purities are compromised by concomitant inclusion of lipoproteins, host cell proteins, and protein aggregates. Those methods often work on lengthy timescales (multiple hours) and result in very low throughput. In this study, capillary-channeled polymer (C-CP) fiber micropipette tips were employed in a hydrophobic interaction chromatography (HIC) solid-phase extraction (SPE) workflow. Demonstrated is the isolation of exosomes from human urine, saliva, cervical mucus, serum, and goat milk matrices. This method allows for quick (<15 min) and low-cost (<$1 per tip) isolations at sample volume and time scales relevant for clinical applications. The tip isolation was evaluated using absorbance (scattering) detection, nanoparticle tracking analysis (NTA), and transmission electron microscopy (TEM). Exosome purity was assessed by Bradford assay, based on the removal of free proteins. An enzyme-linked immunosorbent assay (ELISA) to the CD81 tetraspanin protein was used to confirm the presence of the known exosomal-biomarker on the vesicles.
Collapse
Affiliation(s)
- Kaylan K Jackson
- Clemson University, Department of Chemistry, Clemson, SC 29634, USA.
| | - Rhonda R Powell
- Clemson University, Clemson Light Imaging Facility, Clemson, SC 29634, USA
| | - Terri F Bruce
- Clemson University, Department of Bioengineering, Clemson, SC 29634, USA
| | - R Kenneth Marcus
- Clemson University, Department of Chemistry, Clemson, SC 29634, USA.
| |
Collapse
|
25
|
Malhotra S, Amin ZM, Dobhal G, Cottam S, Nann T, Goreham RV. Novel devices for isolation and detection of bacterial and mammalian extracellular vesicles. Mikrochim Acta 2021; 188:139. [PMID: 33772384 DOI: 10.1007/s00604-021-04790-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/15/2021] [Indexed: 12/11/2022]
Abstract
Extracellular vesicles are spherical nanoparticles inherently released by almost all cell types. They acquire the cell's membrane and cytoplasmic characteristics offering abundant identical units that can be captured to recognize the cell of origin. The abundance of vital cell information and multifunctional roles in cellular processes has rendered them attention, particularly as promising biomarkers for disease diagnosis and use in potential drug delivery systems. This review provides insights into standard approaches towards cultivation and isolation of mammalian and bacterial extracellular vesicles. We assess gaps in conventional separation and detection technologies while also tracking developments in ongoing research. The review focuses on highlighting alternative state-of-the-art microfluidic devices that offer avenues for fast, cost-effective, precision-oriented capture and sensing of extracellular vesicles. Combining different detection technologies on an integrated "lab-on-a-chip" system has the prospective to provide customizable opportunities for clinical use of extracellular vesicles in disease diagnostics and therapeutic applications.
Collapse
Affiliation(s)
- Shiana Malhotra
- School of Mathematical and Physical Sciences, University of Newcastle, Callaghan, 2308, Australia
| | - Zarinah M Amin
- School of Mathematical and Physical Sciences, University of Newcastle, Callaghan, 2308, Australia
| | - Garima Dobhal
- School of Mathematical and Physical Sciences, University of Newcastle, Callaghan, 2308, Australia
| | - Sophie Cottam
- School of Mathematical and Physical Sciences, University of Newcastle, Callaghan, 2308, Australia
| | - Thomas Nann
- School of Mathematical and Physical Sciences, University of Newcastle, Callaghan, 2308, Australia
| | - Renee V Goreham
- School of Mathematical and Physical Sciences, University of Newcastle, Callaghan, 2308, Australia.
| |
Collapse
|
26
|
Chen R, Zhang W, Gong M, Wang F, Wu H, Liu W, Gao Y, Liu B, Chen S, Lu W, Yu X, Liu A, Han R, Chen Y, Han D. Characterization of an Antiviral Component in Human Seminal Plasma. Front Immunol 2021; 12:580454. [PMID: 33679733 PMCID: PMC7933687 DOI: 10.3389/fimmu.2021.580454] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 01/04/2021] [Indexed: 02/04/2023] Open
Abstract
Numerous types of viruses have been found in human semen, which raises concerns about the sexual transmission of these viruses. The overall effect of semen on viral infection and transmission have yet to be fully investigated. In the present study, we aimed at the effect of seminal plasma (SP) on viral infection by focusing on the mumps viral (MuV) infection of HeLa cells. MuV efficiently infected HeLa cells in vitro. MuV infection was strongly inhibited by the pre-treatment of viruses with SP. SP inhibited MuV infection through the impairment of the virus's attachment to cells. The antiviral activity of SP was resistant to the treatment of SP with boiling water, Proteinase K, RNase A, and DNase I, suggesting that the antiviral factor would not be proteins and nucleic acids. PNGase or PLA2 treatments did not abrogate the antiviral effect of SP against MuV. Further, we showed that the prostatic fluid (PF) showed similar inhibition as SP, whereas the epididymal fluid and seminal vesicle extract did not inhibit MuV infection. Both SP and PF also inhibited MuV infection of other cell types, including another human cervical carcinoma cell line C33a, mouse primary epididymal epithelial cells, and Sertoli cell line 15P1. Moreover, this inhibitory effect was not specific to MuV, as the herpes simplex virus 1, dengue virus 2, and adenovirus 5 infections were also inhibited by SP and PF. Our findings suggest that SP contains a prostate-derived pan-antiviral factor that may limit the sexual transmission of various viruses.
Collapse
Affiliation(s)
- Ran Chen
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Wenjing Zhang
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Maolei Gong
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Fei Wang
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Han Wu
- Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Weihua Liu
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yunxiao Gao
- Department of Andrology, China-Japan Friendship Hospital, Beijing, China
| | - Baoxing Liu
- Department of Andrology, China-Japan Friendship Hospital, Beijing, China
| | - Song Chen
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Lu
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaoqin Yu
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Aijie Liu
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Ruiqin Han
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yongmei Chen
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Daishu Han
- Institute of Basic Medical Sciences, School of Basic Medicine, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
27
|
Noschka R, Gerbl F, Löffler F, Kubis J, Rodríguez AA, Mayer D, Grieshober M, Holch A, Raasholm M, Forssmann WG, Spellerberg B, Wiese S, Weidinger G, Ständker L, Stenger S. Unbiased Identification of Angiogenin as an Endogenous Antimicrobial Protein With Activity Against Virulent Mycobacterium tuberculosis. Front Microbiol 2021; 11:618278. [PMID: 33537017 PMCID: PMC7848861 DOI: 10.3389/fmicb.2020.618278] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022] Open
Abstract
Tuberculosis is a highly prevalent infectious disease with more than 1.5 million fatalities each year. Antibiotic treatment is available, but intolerable side effects and an increasing rate of drug-resistant strains of Mycobacterium tuberculosis (Mtb) may hamper successful outcomes. Antimicrobial peptides (AMPs) offer an alternative strategy for treatment of infectious diseases in which conventional antibiotic treatment fails. Human serum is a rich resource for endogenous AMPs. Therefore, we screened a library generated from hemofiltrate for activity against Mtb. Taking this unbiased approach, we identified Angiogenin as the single compound in an active fraction. The antimicrobial activity of endogenous Angiogenin against extracellular Mtb could be reproduced by synthetic Angiogenin. Using computational analysis, we identified the hypothetical active site and optimized the lytic activity by amino acid exchanges. The resulting peptide-Angie1-limited the growth of extra- and intracellular Mtb and the fast-growing pathogens Escherichia coli, Pseudomonas aeruginosa, and Klebsiella pneumoniae. Toward our long-term goal of evaluating Angie1 for therapeutic efficacy in vivo, we demonstrate that the peptide can be efficiently delivered into human macrophages via liposomes and is not toxic for zebrafish embryos. Taken together, we define Angiogenin as a novel endogenous AMP and derive the small, bioactive fragment Angie1, which is ready to be tested for therapeutic activity in animal models of tuberculosis and infections with fast-growing bacterial pathogens.
Collapse
Affiliation(s)
- Reiner Noschka
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Fabian Gerbl
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Florian Löffler
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Jan Kubis
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Armando A Rodríguez
- Core Unit Mass Spectrometry and Proteomics, Ulm University, Ulm, Germany.,Core Facility of Functional Peptidomics, Ulm University, Ulm, Germany
| | - Daniel Mayer
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Mark Grieshober
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Armin Holch
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Martina Raasholm
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | | | - Barbara Spellerberg
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Ulm University, Ulm, Germany
| | - Gilbert Weidinger
- Institute of Biochemistry and Molecular Biology, Ulm University, Ulm, Germany
| | - Ludger Ständker
- Core Facility of Functional Peptidomics, Ulm University, Ulm, Germany
| | - Steffen Stenger
- Institute of Medical Microbiology and Hygiene, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
28
|
Liangsupree T, Multia E, Riekkola ML. Modern isolation and separation techniques for extracellular vesicles. J Chromatogr A 2020; 1636:461773. [PMID: 33316564 DOI: 10.1016/j.chroma.2020.461773] [Citation(s) in RCA: 263] [Impact Index Per Article: 52.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/26/2020] [Accepted: 11/28/2020] [Indexed: 02/07/2023]
Abstract
Extracellular vesicles (EVs) are heterogenous membrane-bound vesicles released from various origins. EVs play a crucial role in cellular communication and mediate several physiological and pathological processes, highlighting their potential therapeutic and diagnostic applications. Due to the rapid increase in interests and needs to elucidate EV properties and functions, numerous isolation and separation approaches for EVs have been developed to overcome limitations of conventional techniques, such as ultracentrifugation. This review focuses on recently emerging and modern EV isolation and separation techniques, including size-, charge-, and affinity-based techniques while excluding ultracentrifugation and precipitation-based techniques due to their multiple limitations. The advantages and drawbacks of each technique are discussed together with insights into their applications. Emerging approaches all share similar features in terms of being time-effective, easy-to-operate, and capable of providing EVs with suitable and desirable purity and integrity for applications of interest. Combination and hyphenation of techniques have been used for EV isolation and separation to yield EVs with the best quality. The most recent development using an automated on-line system including selective affinity-based trapping unit and asymmetrical flow field-flow fractionation allows reliable isolation and fractionation of EV subpopulations from human plasma.
Collapse
Affiliation(s)
| | - Evgen Multia
- Department of Chemistry, P.O. Box 55, FI-00014 University of Helsinki, Finland
| | | |
Collapse
|