1
|
Wijerathne SVT, Pandit R, Ezeuko CC, Matthews QL. Comparative Examination of Feline Coronavirus and Canine Coronavirus Effects on Extracellular Vesicles Acquired from A-72 Canine Fibrosarcoma Cell Line. Vet Sci 2025; 12:477. [PMID: 40431570 PMCID: PMC12115506 DOI: 10.3390/vetsci12050477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 05/02/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Introduction: Coronavirus (CoV) is an extremely contagious, enveloped positive-single-stranded RNA virus, which has become a global pandemic that causes several illnesses in humans and animals. Hence, it is necessary to investigate viral-induced reactions across diverse hosts. Herein, we propose utilizing naturally secreted extracellular vesicles (EVs), mainly focusing on exosomes to examine virus-host responses following CoV infection. Exosomes are small membrane-bound vesicles originating from the endosomal pathway, which play a pivotal role in intracellular communication and physiological and pathological processes. We suggested that CoV could impact EV formation, content, and diverse immune responses in vitro. Methods: In this study, we infected A-72, which is a canine fibroblast cell line, with a feline coronavirus (FCoV) and canine coronavirus (CCoV) independently in an exosome-free media at 0.001 multiplicity of infection (MOI), with incubation periods of 48 and 72 h. The cell viability was significantly downregulated with increased incubation time following FCoV and CCoV infection, which was identified by performing the 3-(4,5-dimethylthiazo-1-2yl)-2,5-diphenyltetrazolium bromide (MTT) assay. After the infection, EVs were isolated through ultracentrifugation, and the subsequent analysis involved quantifying and characterizing the purified EVs using various techniques. Results: NanoSight particle tracking analysis (NTA) verified that EV dimensions fell between 100 and 200 nm at both incubation periods. At both periods, total protein and RNA levels were significantly upregulated in A-72-derived EVs following FCoV and CCoV infections. However, total DNA levels were gradually upregulated with increased incubation time. Dot blot analysis indicated that the expression levels of ACE2, IL-1β, Flotillin-1, CD63, caspase-8, and Hsp90 were modified in A-72-derived EVs following both CoV infections. Conclusions: Our results indicated that FCoV and CCoV infections could modulate the EV production and content, which could play a role in the development of viral diseases. Investigating diverse animal CoV will provide in-depth insight into host exosome biology during CoV infection. Hence, our findings contribute to the comprehension and characterization of EVs in virus-host interactions during CoV infection.
Collapse
Affiliation(s)
- Sandani V. T. Wijerathne
- Microbiology Program, Alabama State University, Montgomery, AL 36104, USA; (S.V.T.W.); (R.P.); (C.C.E.)
| | - Rachana Pandit
- Microbiology Program, Alabama State University, Montgomery, AL 36104, USA; (S.V.T.W.); (R.P.); (C.C.E.)
| | - Chioma C. Ezeuko
- Microbiology Program, Alabama State University, Montgomery, AL 36104, USA; (S.V.T.W.); (R.P.); (C.C.E.)
| | - Qiana L. Matthews
- Microbiology Program, Alabama State University, Montgomery, AL 36104, USA; (S.V.T.W.); (R.P.); (C.C.E.)
- Department of Biological Sciences, College of Science, Technology, Engineering, and Mathematics, Alabama State University, Montgomery, AL 36104, USA
| |
Collapse
|
2
|
Sato Y, Hatanaka Y, Sato Y, Matsumoto K, Osana S, Nagatomi R, Nishizawa S. Viral Membrane-Targeting Amphipathic Helical Peptide-Based Fluorogenic Probes for the Analysis of Infectious Titers of Enveloped Viruses. Anal Chem 2025; 97:5480-5487. [PMID: 39840494 PMCID: PMC11923947 DOI: 10.1021/acs.analchem.4c04852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/23/2025]
Abstract
Enveloped viruses have caused the majority of epidemics and pandemics over the past decade. Direct sensing of virus particles (virions) holds great potential for the functional analysis of enveloped viruses. Here, we explore a series of viral membrane-targeting amphipathic helical (AH) peptide-based molecular probes for the assessment of infectious titers of the human coronavirus 229E virus (HCoV-229E). The M2-protein-derived AH peptide is identified as a strong binder for HCoV-229E, and its conjugate with Nile Red, M2-NR, exhibits fluorogenic response upon selective binding to the viral membrane of HCoV-229E. We demonstrate that the response of M2-NR toward the HCoV-229E virus enables the rapid, simple, and reliable assessment of the infectivity of HCoV-229E. In addition, the present fluorescence assay for infectivity analysis is applicable to various kinds of enveloped virus including influenza A virus, herpes simplex virus-1, and lentivirus.
Collapse
Affiliation(s)
- Yusuke Sato
- Department
of Chemistry, Graduate School of Science, Tohoku University, 6-3
Aramaki-Aza Aoba, Aoba-ku, Sendai 980-8578, Japan
- JST,
PRESTO, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
- JST,
FOREST, 4-1-8 Honcho, Kawaguchi, Saitama 332-0012, Japan
| | - Yusaku Hatanaka
- Department
of Chemistry, Graduate School of Science, Tohoku University, 6-3
Aramaki-Aza Aoba, Aoba-ku, Sendai 980-8578, Japan
| | - Yoshitaka Sato
- Department
of Virology, Nagoya University Graduate
School of Medicine, 65
Tsurumai-cho, Showa-ku, Nagoya 466-8550, Japan
| | - Kota Matsumoto
- Department
of Chemistry, Graduate School of Science, Tohoku University, 6-3
Aramaki-Aza Aoba, Aoba-ku, Sendai 980-8578, Japan
| | - Shion Osana
- Department
of Medicine and Science in Sports and Exercise, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8578, Japan
| | - Ryoichi Nagatomi
- Department
of Medicine and Science in Sports and Exercise, Tohoku University, 2-1 Seiryo-machi, Aoba-ku, Sendai 980-8578, Japan
| | - Seiichi Nishizawa
- Department
of Chemistry, Graduate School of Science, Tohoku University, 6-3
Aramaki-Aza Aoba, Aoba-ku, Sendai 980-8578, Japan
| |
Collapse
|
3
|
Qian H, Zang R, Zhang R, Zheng G, Qiu G, Meng J, Wang J, Xia J, Huang R, Le Z, Shu Q, Xu J. Circulating extracellular vesicles from severe COVID-19 patients induce lung inflammation. mSphere 2024; 9:e0076424. [PMID: 39475319 PMCID: PMC11580465 DOI: 10.1128/msphere.00764-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024] Open
Abstract
Circulating extracellular vesicles (EVs) have been associated with the development of COVID-19 due to their roles in viral infection, inflammatory response, and thrombosis. However, the direct induction of lung inflammation by circulating EVs from severe COVID-19 patients remains unknown. EVs were extracted from the plasma of severe COVID-19 patients admitted to intensive care and healthy controls. To study the effect of COVID-19 EVs on lung inflammation, mice were intratracheally instilled with EVs. To examine the proinflammatory effects of EVs in vitro, bone marrow-derived macrophages were treated with EVs. COVID-19 but not control EVs triggered lung inflammation, as assessed by total protein level, total cell count, neutrophil count, and levels of proinflammatory cytokines in the bronchoalveolar lavage. COVID-19 EVs also promoted M1 polarization of alveolar macrophages in vivo. Treatment of bone marrow-derived macrophages with COVID-19 EVs enhanced the M1 phenotype and augmented the production of IL-1β, IL-6, and TNF-α. In summary, circulating EVs from severe COVID-19 patients induce lung inflammation in mice. EVs could become a potential therapeutic target for alleviating lung injury in COVID-19. IMPORTANCE Extracellular vesicles (EVs) have been reported to facilitate cytokine storm, coagulation, vascular dysfunction, and the spread of the virus in COVID-19. The direct role of circulating EVs from severe COVID-19 patients in lung injury remains unrecognized. Our study demonstrated that plasma EVs obtained from severe COVID-19 patients induced lung inflammation and polarization of alveolar macrophages in vivo. In vitro experiments also revealed the proinflammatory effects of COVID-19 EVs. The present study sheds fresh insight into the mechanisms of COVID-19-induced lung injury, highlighting EVs as a potential therapeutic target in combating the disease.
Collapse
Affiliation(s)
- Huifeng Qian
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Ruoxi Zang
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ruoyang Zhang
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | | | - Guanguan Qiu
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
| | - Jianbiao Meng
- Tongde Hospital of Zhejiang, Hangzhou, Zhejiang, China
| | - Jiangmei Wang
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jie Xia
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ruoqiong Huang
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Zhenkai Le
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiang Shu
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jianguo Xu
- Shaoxing Second Hospital, Shaoxing, Zhejiang, China
- National Clinical Research Center for Child Health, The Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
4
|
Wijerathne SVT, Pandit R, Ipinmoroti AO, Crenshaw BJ, Matthews QL. Feline coronavirus influences the biogenesis and composition of extracellular vesicles derived from CRFK cells. Front Vet Sci 2024; 11:1388438. [PMID: 39091390 PMCID: PMC11292801 DOI: 10.3389/fvets.2024.1388438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Introduction Coronavirus (CoV) has become a public health crisis that causes numerous illnesses in humans and certain animals. Studies have identified the small, lipid-bound structures called extracellular vesicles (EVs) as the mechanism through which viruses can enter host cells, spread, and evade the host's immune defenses. EVs are able to package and carry numerous viral compounds, including proteins, genetic substances, lipids, and receptor proteins. We proposed that the coronavirus could alter EV production and content, as well as influence EV biogenesis and composition in host cells. Methods In the current research, Crandell-Rees feline kidney (CRFK) cells were infected with feline coronavirus (FCoV) in an exosome-free media at a multiplicity of infection (MOI) of 2,500 infectious units (IFU) at 48 h and 72 h time points. Cell viability was analyzed and found to be significantly decreased by 9% (48 h) and 15% (72 h) due to FCoV infection. EVs were isolated by ultracentrifugation, and the surface morphology of isolated EVs was analyzed via Scanning Electron Microscope (SEM). Results NanoSight particle tracking analysis (NTA) confirmed that the mean particle sizes of control EVs were 131.9 nm and 126.6 nm, while FCoV infected-derived EVs were 143.4 nm and 120.9 nm at 48 and 72 h, respectively. Total DNA, RNA, and protein levels were determined in isolated EVs at both incubation time points; however, total protein was significantly increased at 48 h. Expression of specific protein markers such as TMPRSS2, ACE2, Alix, TSG101, CDs (29, 47, 63), TLRs (3, 6, 7), TNF-α, and others were altered in infection-derived EVs when compared to control-derived EVs after FCoV infection. Discussion Our findings suggested that FCoV infection could alter the EV production and composition in host cells, which affects the infection progression and disease evolution. One purpose of studying EVs in various animal coronaviruses that are in close contact with humans is to provide significant information about disease development, transmission, and adaptation. Hence, this study suggests that EVs could provide diagnostic and therapeutic applications in animal CoVs, and such understanding could provide information to prevent future coronavirus outbreaks.
Collapse
Affiliation(s)
| | - Rachana Pandit
- Microbiology Program, Alabama State University, Montgomery, AL, United States
| | | | | | - Qiana L. Matthews
- Microbiology Program, Alabama State University, Montgomery, AL, United States
- Department of Biological Sciences, College of Science, Technology, Engineering, and Mathematics, Alabama State University, Montgomery, AL, United States
| |
Collapse
|
5
|
Ameratunga R, Jordan A, Lehnert K, Leung E, Mears ER, Snell R, Steele R, Woon ST. SARS-CoV-2 evolution has increased resistance to monoclonal antibodies and first-generation COVID-19 vaccines: Is there a future therapeutic role for soluble ACE2 receptors for COVID-19? Antiviral Res 2024; 227:105894. [PMID: 38677595 DOI: 10.1016/j.antiviral.2024.105894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Revised: 04/17/2024] [Accepted: 04/22/2024] [Indexed: 04/29/2024]
Abstract
COVID-19 has caused calamitous health, economic and societal consequences. Although several COVID-19 vaccines have received full authorization for use, global deployment has faced political, financial and logistical challenges. The efficacy of first-generation COVID-19 vaccines is waning and breakthrough infections are allowing ongoing transmission and evolution of SARS-CoV-2. Furthermore, COVID-19 vaccine efficacy relies on a functional immune system. Despite receiving three primary doses and three or more heterologous boosters, some immunocompromised patients may not be adequately protected by COVID-19 vaccines and remain vulnerable to severe disease. The evolution of new SARS-CoV-2 variants has also resulted in the rapid obsolescence of monoclonal antibodies. Convalescent plasma from COVID-19 survivors has produced inconsistent results. New drugs such as Paxlovid (nirmatrelvir/ritonavir) are beyond the reach of low- and middle-income countries. With widespread use of Paxlovid, it is likely nirmatrelvir-resistant clades of SARS-CoV-2 will emerge in the future. There is thus an urgent need for new effective anti-SARS-CoV-2 treatments. The in vitro efficacy of soluble ACE2 against multiple SARS-CoV-2 variants including omicron (B.1.1.529), was recently described using a competitive ELISA assay as a surrogate marker for virus neutralization. This indicates soluble wild-type ACE2 receptors are likely to be resistant to viral evolution. Nasal and inhaled treatment with soluble ACE2 receptors has abrogated severe disease in animal models of COVID-19. There is an urgent need for clinical trials of this new class of antiviral therapeutics, which could complement vaccines and Paxlovid.
Collapse
Affiliation(s)
- Rohan Ameratunga
- Department of Clinical Immunology, Auckland Hospital, Park Rd, Grafton, 1010, Auckland, New Zealand; Department of Virology and Immunology, Auckland Hospital, Park Rd, Grafton, 1010, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand.
| | - Anthony Jordan
- Department of Clinical Immunology, Auckland Hospital, Park Rd, Grafton, 1010, Auckland, New Zealand
| | - Klaus Lehnert
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Euphemia Leung
- Auckland Cancer Society Research Centre, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Emily R Mears
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Russell Snell
- Applied Translational Genetics Group, School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Richard Steele
- Department of Virology and Immunology, Auckland Hospital, Park Rd, Grafton, 1010, Auckland, New Zealand
| | - See-Tarn Woon
- Department of Virology and Immunology, Auckland Hospital, Park Rd, Grafton, 1010, Auckland, New Zealand; Department of Molecular Medicine and Pathology, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
6
|
Hwang J, Kim BK, Moon S, Park W, Kim KW, Yoon JH, Oh H, Jung S, Park Y, Kim S, Kim M, Kim S, Jung Y, Park M, Kim JH, Jung ST, Kim SJ, Kim YS, Chung WJ, Song MS, Kweon DH. Conversion of Host Cell Receptor into Virus Destructor by Immunodisc to Neutralize Diverse SARS-CoV-2 Variants. Adv Healthc Mater 2024; 13:e2302803. [PMID: 38329411 DOI: 10.1002/adhm.202302803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 12/29/2023] [Indexed: 02/09/2024]
Abstract
The decreasing efficacy of antiviral drugs due to viral mutations highlights the challenge of developing a single agent targeting multiple strains. Using host cell viral receptors as competitive inhibitors is promising, but their low potency and membrane-bound nature have limited this strategy. In this study, the authors show that angiotensin-converting enzyme 2 (ACE2) in a planar membrane patch can effectively neutralize all tested severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants that emerged during the COVID-19 pandemic. The ACE2-incorporated membrane patch implemented using nanodiscs replicated the spike-mediated membrane fusion process outside the host cell, resulting in virus lysis, extracellular RNA release, and potent antiviral activity. While neutralizing antibodies became ineffective as the SARS-CoV-2 evolved to better penetrate host cells the ACE2-incorporated nanodiscs became more potent, highlighting the advantages of using receptor-incorporated nanodiscs for antiviral purposes. ACE2-incorporated immunodisc, an Fc fusion nanodisc developed in this study, completely protected humanized mice infected with SARS-CoV-2 after prolonged retention in the airways. This study demonstrates that the incorporation of viral receptors into immunodisc transforms the entry gate into a potent virucide for all current and future variants, a concept that can be extended to different viruses.
Collapse
Affiliation(s)
- Jaehyeon Hwang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Beom Kyu Kim
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Seokoh Moon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Wonbeom Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Kyeong Won Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jeong Hyeon Yoon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyunseok Oh
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Research Center, Mvrix Inc., Anyang, 14058, Republic of Korea
| | - Sangwon Jung
- Research Center, Mvrix Inc., Anyang, 14058, Republic of Korea
| | - Youngseo Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Suhyun Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Misoo Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Soomin Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Younghun Jung
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Myungseo Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jun-Ho Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Sang Taek Jung
- Department of Biomedical Sciences, Graduate School, Korea University, Seoul, 02841, Republic of Korea
| | - Sang Jick Kim
- Synthetic Biology and Bioengineering Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, 34141, Republic of Korea
| | - Yong-Sung Kim
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Woo-Jae Chung
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Min-Suk Song
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, 28644, Republic of Korea
| | - Dae-Hyuk Kweon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Research Center, Mvrix Inc., Anyang, 14058, Republic of Korea
| |
Collapse
|
7
|
Schaubmayr W, Hochreiter B, Hunyadi-Gulyas E, Riegler L, Schmidt K, Tiboldi A, Moser B, Klein KU, Krenn K, Scharbert G, Mohr T, Schmid JA, Spittler A, Tretter V. The Proteome of Extracellular Vesicles Released from Pulmonary Microvascular Endothelium Reveals Impact of Oxygen Conditions on Biotrauma. Int J Mol Sci 2024; 25:2415. [PMID: 38397093 PMCID: PMC10889365 DOI: 10.3390/ijms25042415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 02/09/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
The lung can experience different oxygen concentrations, low as in hypoxia, high as under supplemental oxygen therapy, or oscillating during intermittent hypoxia as in obstructive sleep apnea or intermittent hypoxia/hyperoxia due to cyclic atelectasis in the ventilated patient. This study aimed to characterize the oxygen-condition-specific protein composition of extracellular vesicles (EVs) released from human pulmonary microvascular endothelial cells in vitro to decipher their potential role in biotrauma using quantitative proteomics with bioinformatic evaluation, transmission electron microscopy, flow cytometry, and non-activated thromboelastometry (NATEM). The release of vesicles enriched in markers CD9/CD63/CD81 was enhanced under intermittent hypoxia, strong hyperoxia and intermittent hypoxia/hyperoxia. Particles with exposed phosphatidylserine were increased under intermittent hypoxia. A small portion of vesicles were tissue factor-positive, which was enhanced under intermittent hypoxia and intermittent hypoxia/hyperoxia. EVs from treatment with intermittent hypoxia induced a significant reduction of Clotting Time in NATEM analysis compared to EVs isolated after normoxic exposure, while after intermittent hypoxia/hyperoxia, tissue factor in EVs seems to be inactive. Gene set enrichment analysis of differentially expressed genes revealed that EVs from individual oxygen conditions potentially induce different biological processes such as an inflammatory response under strong hyperoxia and intermittent hypoxia/hyperoxia and enhancement of tumor invasiveness under intermittent hypoxia.
Collapse
Affiliation(s)
- Wolfgang Schaubmayr
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Beatrix Hochreiter
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Eva Hunyadi-Gulyas
- Laboratory of Proteomics Research, HUN-REN Biological Research Centre, 6726 Szeged, Hungary;
| | - Louise Riegler
- Department of Orthopedics and Trauma Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Katy Schmidt
- Core Facility of Cell Imaging and Ultrastructure Research, University of Vienna, 1090 Vienna, Austria
| | - Akos Tiboldi
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Bernhard Moser
- Department of Thoracic Surgery, Medical University of Vienna, 1090 Vienna, Austria
| | - Klaus U. Klein
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Katharina Krenn
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Gisela Scharbert
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| | - Thomas Mohr
- Institute of Cancer Research, Department of Medicine I, Comprehensive Cancer Center, Medical University of Vienna, 1090 Vienna, Austria
| | - Johannes A. Schmid
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, 1090 Vienna, Austria;
| | - Andreas Spittler
- Department of Surgery and Core Facility Flow Cytometry, Medical University of Vienna, 1090 Vienna, Austria;
| | - Verena Tretter
- Department of Anesthesia, General Intensive Care and Pain Medicine, Medical University of Vienna, 1090 Vienna, Austria (B.H.); (K.K.)
| |
Collapse
|
8
|
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
|
9
|
Hambo S, Harb H. Extracellular Vesicles and Their Role in Lung Infections. Int J Mol Sci 2023; 24:16139. [PMID: 38003329 PMCID: PMC10671184 DOI: 10.3390/ijms242216139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 11/07/2023] [Accepted: 11/08/2023] [Indexed: 11/26/2023] Open
Abstract
Lung infections are one of the most common causes of death and morbidity worldwide. Both bacterial and viral lung infections cause a vast number of infections with varying severities. Extracellular vesicles (EVs) produced by different cells due to infection in the lung have the ability to modify the immune system, leading to either better immune response or worsening of the disease. It has been shown that both bacteria and viruses have the ability to produce their EVs and stimulate the immune system for that. In this review, we investigate topics from EV biogenesis and types of EVs to lung bacterial and viral infections caused by various bacterial species. Mycobacterium tuberculosis, Staphylococcus aureus, and Streptococcus pneumoniae infections are covered intensively in this review. Moreover, various viral lung infections, including SARS-CoV-2 infections, have been depicted extensively. In this review, we focus on eukaryotic-cell-derived EVs as an important component of disease pathogenesis. Finally, this review holds high novelty in its findings and literature review. It represents the first time to cover all different information on immune-cell-derived EVs in both bacterial and viral lung infections.
Collapse
Affiliation(s)
| | - Hani Harb
- Institute for Medical Microbiology and Virology, University Hospital Dresden, Technical University Dresden, Fetscherstr. 74, 01307 Dresden, Germany;
| |
Collapse
|
10
|
Bou JV, Taguwa S, Matsuura Y. Trick-or-Trap: Extracellular Vesicles and Viral Transmission. Vaccines (Basel) 2023; 11:1532. [PMID: 37896936 PMCID: PMC10611016 DOI: 10.3390/vaccines11101532] [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: 07/24/2023] [Revised: 09/15/2023] [Accepted: 09/23/2023] [Indexed: 10/29/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid membrane-enclosed particles produced by most cells, playing important roles in various biological processes. They have been shown to be involved in antiviral mechanisms such as transporting antiviral molecules, transmitting viral resistance, and participating in antigen presentation. While viral transmission was traditionally thought to occur through independent viral particles, the process of viral infection is complex, with multiple barriers and challenges that viruses must overcome for successful infection. As a result, viruses exploit the intercellular communication pathways of EVs to facilitate cluster transmission, increasing their chances of infecting target cells. Viral vesicle transmission offers two significant advantages. Firstly, it enables the collective transmission of viral genomes, increasing the chances of infection and promoting interactions between viruses in subsequent generations. Secondly, the use of vesicles as vehicles for viral transmission provides protection to viral particles against environmental factors, while also expanding the cell tropism allowing viruses to reach cells in a receptor-independent manner. Understanding the role of EVs in viral transmission is crucial for comprehending virus evolution and developing innovative antiviral strategies, therapeutic interventions, and vaccine approaches.
Collapse
Affiliation(s)
- Juan-Vicente Bou
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, 2-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Shuhei Taguwa
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, 2-8 Yamadaoka, Suita, Osaka 565-0871, Japan
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center for Advanced Modalities and DDS, Osaka University, 2-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yoshiharu Matsuura
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, 2-8 Yamadaoka, Suita, Osaka 565-0871, Japan
- Research Institute for Microbial Diseases, Osaka University, 3-1 Yamadaoka, Suita, Osaka 565-0871, Japan
- Center for Advanced Modalities and DDS, Osaka University, 2-8 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
11
|
Fu Y, Zi R, Xiong S. Infection by exosome-carried Coxsackievirus B3 induces immune escape resulting in an aggravated pathogenesis. Microbes Infect 2023; 25:105148. [PMID: 37156458 DOI: 10.1016/j.micinf.2023.105148] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 05/10/2023]
Abstract
Increasing evidence has shown that extracellular vesicles or exosomes released from virus-infected cells contain viral particles, genomes, or other pathogenic factors that move to neighbor cells, contributing to virus dissemination and productive infection. Our recent study demonstrated that exosomes carrying CVB3 virions exhibited greater infection efficiency than free virions because they accessed various entry routes, overcoming restrictions to viral tropism. However, the pathogenicity of exosomes carried CVB3 and their effect on immunological properties have not yet been completely explained. In the current study, we sought to explore whether exosomes exert their effect on the CVB3-induced pathogenesis or evade the immune attack. Our results showed that exosomes-carried CVB3 could effectively infect viral receptor-negative immune cells in vivo, resulting in inducing immune system loss. Importantly, the exosomes-carried CVB3 had the ability to escape the neutralizing antibodies activity resulting in inducing the severe onset of myocarditis. Using the genetically engineered mouse with deficiency of exosomes, we observed that the exosomes-carried CVB3 reinforced an aggravated pathogenesis. By understanding how exosomes promote the course of viral disease, clinical applications of exosomes can be developed.
Collapse
Affiliation(s)
- Yuxuan Fu
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Ruidong Zi
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China
| | - Sidong Xiong
- Jiangsu Key Laboratory of Infection and Immunity, Institutes of Biology and Medical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
12
|
Beetler DJ, Di Florio DN, Bruno KA, Ikezu T, March KL, Cooper LT, Wolfram J, Fairweather D. Extracellular vesicles as personalized medicine. Mol Aspects Med 2023; 91:101155. [PMID: 36456416 PMCID: PMC10073244 DOI: 10.1016/j.mam.2022.101155] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/14/2022] [Accepted: 10/26/2022] [Indexed: 11/29/2022]
Abstract
Extracellular vesicles (EVs) are released from all cells in the body, forming an important intercellular communication network that contributes to health and disease. The contents of EVs are cell source-specific, inducing distinct signaling responses in recipient cells. The specificity of EVs and their accumulation in fluid spaces that are accessible for liquid biopsies make them highly attractive as potential biomarkers and therapies for disease. The duality of EVs as favorable (therapeutic) or unfavorable (pathological) messengers is context dependent and remains to be fully determined in homeostasis and various disease states. This review describes the use of EVs as biomarkers, drug delivery vehicles, and regenerative therapeutics, highlighting examples involving viral infections, cancer, and neurological diseases. There is growing interest to provide personalized therapy based on individual patient and disease characteristics. Increasing evidence suggests that EV biomarkers and therapeutic approaches are ideal for personalized medicine due to the diversity and multifunctionality of EVs.
Collapse
Affiliation(s)
- Danielle J Beetler
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, 55902, USA; Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Damian N Di Florio
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, 55902, USA; Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Katelyn A Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA; Center for Regenerative Medicine, University of Florida, Gainesville, FL, 32611, USA; Division of Cardiology, University of Florida, Gainesville, FL, 32611, USA
| | - Tsuneya Ikezu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Keith L March
- Center for Regenerative Medicine, University of Florida, Gainesville, FL, 32611, USA; Division of Cardiology, University of Florida, Gainesville, FL, 32611, USA
| | - Leslie T Cooper
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Joy Wolfram
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD, 4072, Australia; Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - DeLisa Fairweather
- Center for Clinical and Translational Science, Mayo Clinic, Rochester, MN, 55902, USA; Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL, 32224, USA; Department of Environmental Health Sciences and Engineering, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
| |
Collapse
|
13
|
Hernández-Díazcouder A, Díaz-Godínez C, Carrero JC. Extracellular vesicles in COVID-19 prognosis, treatment, and vaccination: an update. Appl Microbiol Biotechnol 2023; 107:2131-2141. [PMID: 36917275 PMCID: PMC10012322 DOI: 10.1007/s00253-023-12468-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 02/27/2023] [Accepted: 03/02/2023] [Indexed: 03/16/2023]
Abstract
The lethality of the COVID 19 pandemic became the trigger for one of the most meteoric races on record in the search for strategies of disease control. Those include development of rapid and sensitive diagnostic methods, therapies to treat severe cases, and development of anti-SARS-CoV-2 vaccines, the latter responsible for the current relative control of the disease. However, the commercially available vaccines are still far from conferring protection against acquiring the infection, so the development of more efficient vaccines that can cut the transmission of the variants of concerns that currently predominate and those that will emerge is a prevailing need. On the other hand, considering that COVID 19 is here to stay, the development of new diagnosis and treatment strategies is also desirable. In this sense, there has recently been a great interest in taking advantage of the benefits offered by extracellular vesicles (EVs), membrane structures of nanoscale size that carry information between cells participating in this manner in many physiological homeostatic and pathological processes. The interest has been focused on the fact that EVs are relatively easy to obtain and manipulate, allowing the design of natural nanocarriers that deliver molecules of interest, as well as the information about the pathogens, which can be exploited for the aforementioned purposes. Studies have shown that infection with SARS-CoV-2 induces the release of EVs from different sources, including platelets, and that their increase in blood, as well as some of their markers, could be used as a prognosis of disease severity. Likewise, EVs from different sources are being used as the ideal carriers for delivering active molecules and drugs to treat the disease, as well as vaccine antigens. In this review, we describe the progress that has been made in these three years of pandemic regarding the use of EVs for diagnosis, treatment, and vaccination against SARS-CoV-2 infection. KEY POINTS: • Covid-19 still requires more effective and specific treatments and vaccines. • The use of extracellular vesicles is emerging as an option with multiple advantages. • Association of EVs with COVID 19 and engineered EVs for its control are presented.
Collapse
Affiliation(s)
- Adrián Hernández-Díazcouder
- Departamento de Inmunología, Instituto Nacional de Cardiología Ignacio Chávez, Ciudad de México, México
- Departamento de Ciencias de La Salud, Universidad Tecnológica de México (UNITEC), Estado de México, Los Reyes, México
| | - César Díaz-Godínez
- Departamento de Ciencias de La Salud, Universidad Tecnológica de México (UNITEC), Estado de México, Los Reyes, México
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México
| | - Julio César Carrero
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), 04510, Ciudad de México, México.
| |
Collapse
|
14
|
Kong L, Yang C, Zhang Z. Organism-Generated Biological Vesicles In Situ: An Emerging Drug Delivery Strategy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204178. [PMID: 36424135 PMCID: PMC9839880 DOI: 10.1002/advs.202204178] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 10/13/2022] [Indexed: 06/16/2023]
Abstract
Biological vesicles, containing genetic materials and proteins of the original cells, are usually used for local or systemic communications among cells. Currently, studies on biological vesicles as therapeutic strategies or drug delivery carriers mainly focus on exogenously generated biological vesicles. However, the limitations of yield and purity caused by the complex purification process still hinder their clinical transformation. Recently, it has been reported that living organisms, including cells and bacteria, can produce functional/therapeutic biological vesicles within body automatically. Therefore, using organisms to produce endogenous biological vesicles in body as drug/bio-information delivery carriers has become a potential therapeutic strategy. In this review, the current development status and application prospects of in situ organism-produced biological vesicles are introduced. The advantages and effects of this endogenous biological vesicles-based strategy in drug delivery and disease treatments are analyzed. According to the type of endogenous biological vesicles, they are divided into four categories: exosomes, platelet-derived microparticles, apoptotic bodies, and bacteria-released outer membrane vesicles. And finally, the shortcomings of current research and future development are analyzed. This review is believed to open up the application of endogenous biological vesicles in the field of biomedicine and shed light on current research.
Collapse
Affiliation(s)
- Li Kong
- Tongji School of PharmacyHuazhong University of Science and TechnologyWuhan430030P. R. China
| | - Conglian Yang
- Tongji School of PharmacyHuazhong University of Science and TechnologyWuhan430030P. R. China
| | - Zhiping Zhang
- Tongji School of PharmacyHuazhong University of Science and TechnologyWuhan430030P. R. China
- Hubei Engineering Research Center for Novel Drug Delivery SystemHuazhong University of Science and TechnologyWuhan430030P. R. China
- National Engineering Research Center for NanomedicineHuazhong University of Science and TechnologyWuhan430030P. R. China
| |
Collapse
|
15
|
Chen X, Li H, Song H, Wang J, Zhang X, Han P, Wang X. Meet changes with constancy: Defence, antagonism, recovery, and immunity roles of extracellular vesicles in confronting SARS-CoV-2. J Extracell Vesicles 2022; 11:e12288. [PMID: 36450704 PMCID: PMC9712136 DOI: 10.1002/jev2.12288] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 11/12/2022] [Accepted: 11/16/2022] [Indexed: 12/03/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has wrought havoc on the world economy and people's daily lives. The inability to comprehensively control COVID-19 is due to the difficulty of early and timely diagnosis, the lack of effective therapeutic drugs, and the limited effectiveness of vaccines. The body contains billions of extracellular vesicles (EVs), which have shown remarkable potential in disease diagnosis, drug development, and vaccine carriers. Recently, increasing evidence has indicated that EVs may participate or assist the body in defence, antagonism, recovery and acquired immunity against SARS-CoV-2. On the one hand, intercepting and decrypting the general intelligence carried in circulating EVs from COVID-19 patients will provide an important hint for diagnosis and treatment; on the other hand, engineered EVs modified by gene editing in the laboratory will amplify the effectiveness of inhibiting infection, replication and destruction of ever-mutating SARS-CoV-2, facilitating tissue repair and making a better vaccine. To comprehensively understand the interaction between EVs and SARS-CoV-2, providing new insights to overcome some difficulties in the diagnosis, prevention and treatment of COVID-19, we conducted a rounded review in this area. We also explain numerous critical challenges that these tactics face before they enter the clinic, and this work will provide previous 'meet change with constancy' lessons for responding to future similar public health disasters. Extracellular vesicles (EVs) provide a 'meet changes with constancy' strategy to combat SARS-CoV-2 that spans defence, antagonism, recovery, and acquired immunity. Targets for COVID-19 diagnosis, therapy, and prevention of progression may be found by capture of the message decoding in circulating EVs. Engineered and biomimetic EVs can boost effects of the natural EVs, especially anti-SARS-CoV-2, targeted repair of damaged tissue, and improvement of vaccine efficacy.
Collapse
Affiliation(s)
- Xiaohang Chen
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
- Fujian Key Laboratory of Oral Diseases, School and Hospital of StomatologyFujian Medical UniversityFuzhouChina
| | - Huifei Li
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
| | - Haoyue Song
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
| | - Jie Wang
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
| | - Xiaoxuan Zhang
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
| | - Pengcheng Han
- CAS Key Laboratory of Pathogen Microbiology and ImmunologyInstitute of Microbiology, Chinese Academy of SciencesBeijingChina
- School of MedicineZhongda Hospital, Southeast UniversityNanjingChina
| | - Xing Wang
- Shanxi Medical University School and Hospital of StomatologyTaiyuanChina
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New MaterialsTaiyuanChina
| |
Collapse
|
16
|
Kongsomros S, Pongsakul N, Panachan J, Khowawisetsut L, Somkird J, Sangma C, Kanjanapruthipong T, Wongtrakoongate P, Chairoungdua A, Pattanapanyasat K, Newburg DS, Morrow AL, Hongeng S, Thitithanyanont A, Chutipongtanate S. Comparison of viral inactivation methods on the characteristics of extracellular vesicles from SARS-CoV-2 infected human lung epithelial cells. J Extracell Vesicles 2022; 11:e12291. [PMID: 36468940 PMCID: PMC9721205 DOI: 10.1002/jev2.12291] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 11/15/2022] [Accepted: 11/23/2022] [Indexed: 12/12/2022] Open
Abstract
The interaction of SARS-CoV-2 infection with extracellular vesicles (EVs) is of particular interest at the moment. Studying SARS-CoV-2 contaminated-EV isolates in instruments located outside of the biosafety level-3 (BSL-3) environment requires knowing how viral inactivation methods affect the structure and function of extracellular vesicles (EVs). Therefore, three common viral inactivation methods, ultraviolet-C (UVC; 1350 mJ/cm2 ), β-propiolactone (BPL; 0.005%), heat (56°C, 45 min) were performed on defined EV particles and their proteins, RNAs, and function. Small EVs were isolated from the supernatant of SARS-CoV-2-infected human lung epithelial Calu-3 cells by stepwise centrifugation, ultrafiltration and qEV size-exclusion chromatography. The EV isolates contained SARS-CoV-2. UVC, BPL and heat completely abolished SARS-CoV-2 infectivity of the contaminated EVs. Particle detection by electron microscopy and nanoparticle tracking was less affected by UVC and BPL than heat treatment. Western blot analysis of EV markers was not affected by any of these three methods. UVC reduced SARS-CoV-2 spike detectability by quantitative RT-PCR and slightly altered EV-derived β-actin detection. Fibroblast migration-wound healing activity of the SARS-CoV-2 contaminated-EV isolate was only retained after UVC treatment. In conclusion, specific viral inactivation methods are compatible with specific measures in SARS-CoV-2 contaminated-EV isolates. UVC treatment seems preferable for studying functions of EVs released from SARS-CoV-2 infected cells.
Collapse
Affiliation(s)
- Supasek Kongsomros
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi HospitalMahidol UniversitySamut PrakanThailand
- Pediatric Translational Research Unit, Department of PediatricsFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Department of Microbiology, Faculty of ScienceMahidol UniversityBangkokThailand
| | - Nutkridta Pongsakul
- Pediatric Translational Research Unit, Department of PediatricsFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
| | - Jirawan Panachan
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi HospitalMahidol UniversityBangkokThailand
| | - Ladawan Khowawisetsut
- Department of Parasitology, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Jinjuta Somkird
- Department of Parasitology, Faculty of Medicine Siriraj HospitalMahidol UniversityBangkokThailand
| | - Chak Sangma
- Department of Chemistry, Faculty of ScienceKasetsart UniversityBangkokThailand
| | | | | | - Arthit Chairoungdua
- Department of Biochemistry, Faculty of ScienceMahidol UniversityBangkokThailand
| | - Kovit Pattanapanyasat
- Center of Excellence for Microparticle and Exosome in Diseases, Research DepartmentFaculty of Medicine Siriraj Hospital, Mahidol UniversityBangkokThailand
| | - David S. Newburg
- Division of Epidemiology, Department of Environmental and Public Health SciencesUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| | - Ardythe L. Morrow
- Division of Epidemiology, Department of Environmental and Public Health SciencesUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
- Division of Infectious Diseases, Department of PediatricsCincinnati Children's Hospital Medical CenterCincinnatiOhioUSA
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine Ramathibodi HospitalMahidol UniversityBangkokThailand
| | | | - Somchai Chutipongtanate
- Chakri Naruebodindra Medical Institute, Faculty of Medicine Ramathibodi HospitalMahidol UniversitySamut PrakanThailand
- Pediatric Translational Research Unit, Department of PediatricsFaculty of Medicine Ramathibodi Hospital, Mahidol UniversityBangkokThailand
- Division of Epidemiology, Department of Environmental and Public Health SciencesUniversity of Cincinnati College of MedicineCincinnatiOhioUSA
| |
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
|
Mustajab T, Kwamboka MS, Choi DA, Kang DW, Kim J, Han KR, Han Y, Lee S, Song D, Chwae YJ. Update on Extracellular Vesicle-Based Vaccines and Therapeutics to Combat COVID-19. Int J Mol Sci 2022; 23:ijms231911247. [PMID: 36232549 PMCID: PMC9569487 DOI: 10.3390/ijms231911247] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
The COVID-19 pandemic has had a deep impact on people worldwide since late 2019 when SARS-CoV-2 was first identified in Wuhan, China. In addition to its effect on public health, it has affected humans in various aspects of life, including social, economic, cultural, and political. It is also true that researchers have made vigorous efforts to overcome COVID-19 throughout the world, but they still have a long way to go. Accordingly, innumerable therapeutics and vaccine candidates have been studied for their efficacies and have been tried clinically in a very short span of time. For example, the versatility of extracellular vesicles, which are membrane-bound particles released from all types of cells, have recently been highlighted in terms of their effectiveness, biocompatibility, and safety in the fight against COVID-19. Thus, here, we tried to explain the use of extracellular vesicles as therapeutics and for the development of vaccines against COVID-19. Along with the mechanisms and a comprehensive background of their application in trapping the coronavirus or controlling the cytokine storm, we also discuss the obstacles to the clinical use of extracellular vesicles and how these could be resolved in the future.
Collapse
Affiliation(s)
- Tamanna Mustajab
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Moriasi Sheba Kwamboka
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Da Ae Choi
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Dae Wook Kang
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Junho Kim
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Kyu Ri Han
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Yujin Han
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Sorim Lee
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Dajung Song
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
| | - Yong-Joon Chwae
- Department of Microbiology, School of Medicine, Ajou University, Suwon 16499, Korea
- Department of Biomedical Science, Graduate School of Ajou University, Suwon 16499, Korea
- Correspondence: ; Tel.: +82-031-219-5073
| |
Collapse
|