1
|
Pawlik MT, Rinneberg G, Koch A, Meyringer H, Loew TH, Kjellberg A. Is there a rationale for hyperbaric oxygen therapy in the patients with Post COVID syndrome? : A critical review. Eur Arch Psychiatry Clin Neurosci 2024; 274:1797-1817. [PMID: 39545965 PMCID: PMC11579208 DOI: 10.1007/s00406-024-01911-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 09/16/2024] [Indexed: 11/17/2024]
Abstract
The SARS-CoV-2 pandemic has resulted in 762 million infections worldwide from 2020 to date, of which approximately ten percent are suffering from the effects after infection in 2019 (COVID-19) [1, 40]. In Germany, it is now assumed that at least one million people suffer from post-COVID condition with long-term consequences. These have been previously reported in diseases like Myalgic Encephalomyelitis (ME) and Chronic Fatigue Syndrome (CFS). Symptoms show a changing variability and recent surveys in the COVID context indicate that 10-30 % of outpatients, 50 to 70% of hospitalised patients suffer from sequelae. Recent data suggest that only 13% of all ill people were completely free of symptoms after recovery [3, 9]. Current hypotheses consider chronic inflammation, mitochondrial dysfunction, latent viral persistence, autoimmunity, changes of the human microbiome or multilocular sequelae in various organ system after infection. Hyperbaric oxygen therapy (HBOT) is applied since 1957 for heart surgery, scuba dive accidents, CO intoxication, air embolisms and infections with anaerobic pathogens. Under hyperbaric pressure, oxygen is physically dissolved in the blood in higher concentrations and reaches levels four times higher than under normobaric oxygen application. Moreover, the alternation of hyperoxia and normoxia induces a variety of processes at the cellular level, which improves oxygen supply in areas of locoregional hypoxia. Numerous target gene effects on new vessel formation, anti-inflammatory and anti-oedematous effects have been demonstrated [74]. The provision of intermittently high, local oxygen concentrations increases repair and regeneration processes and normalises the predominance of hyperinflammation. At present time only one prospective, randomized and placebo-controlled study exists with positive effects on global cognitive function, attention and executive function, psychiatric symptoms and pain interference. In conclusion, up to this date HBO is the only scientifically proven treatment in a prospective randomized controlled trial to be effective for cognitive improvement, regeneration of brain network and improvement of cardiac function. HBOT may have not only theoretical but also potential impact on targets of current pathophysiology of Post COVID condition, which warrants further scientific studies in patients.
Collapse
Affiliation(s)
- M T Pawlik
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany.
- Institute of Experimental Medicine, Christian-Albrechts-University of Kiel c/o German Naval Medical Institute, Kronshagen, Germany.
| | - G Rinneberg
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany
| | - A Koch
- Institute of Experimental Medicine, Christian-Albrechts-University of Kiel c/o German Naval Medical Institute, Kronshagen, Germany
| | - H Meyringer
- Department of Anesthesiology and Intensive Care Medicine, Caritas-Hospital St. Joseph, University of Regensburg, Regensburg, Germany
| | - T H Loew
- Department of Psychosomatic Medicine, University Hospital Regensburg, Regensburg, Germany
| | - A Kjellberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Solna, Sweden
- Perioperative Medicine and Intensive Care, Medical Unit Intensive Care and Thoracic surgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
2
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
3
|
Song IW, Washington M, Leynes C, Hsu J, Rayavara K, Bae Y, Haelterman N, Chen Y, Jiang MM, Drelich A, Tat V, Lanza DG, Lorenzo I, Heaney JD, Tseng CTK, Lee B, Marom R. Generation of a humanized mAce2 and a conditional hACE2 mouse models permissive to SARS-COV-2 infection. Mamm Genome 2024; 35:113-121. [PMID: 38488938 PMCID: PMC11955088 DOI: 10.1007/s00335-024-10033-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 02/02/2024] [Indexed: 03/17/2024]
Abstract
The Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) remains a public health concern and a subject of active research effort. Development of pre-clinical animal models is critical to study viral-host interaction, tissue tropism, disease mechanisms, therapeutic approaches, and long-term sequelae of infection. Here, we report two mouse models for studying SARS-CoV-2: A knock-in mAce2F83Y,H353K mouse that expresses a mouse-human hybrid form of the angiotensin-converting enzyme 2 (ACE2) receptor under the endogenous mouse Ace2 promoter, and a Rosa26 conditional knock-in mouse carrying the human ACE2 allele (Rosa26hACE2). Although the mAce2F83Y,H353K mice were susceptible to intranasal inoculation with SARS-CoV-2, they did not show gross phenotypic abnormalities. Next, we generated a Rosa26hACE2;CMV-Cre mouse line that ubiquitously expresses the human ACE2 receptor. By day 3 post infection with SARS-CoV-2, Rosa26hACE2;CMV-Cre mice showed significant weight loss, a variable degree of alveolar wall thickening and reduced survival rates. Viral load measurements confirmed inoculation in lung and brain tissues of infected Rosa26hACE2;CMV-Cre mice. The phenotypic spectrum displayed by our different mouse models translates to the broad range of clinical symptoms seen in the human patients and can serve as a resource for the community to model and explore both treatment strategies and long-term consequences of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- I-Wen Song
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Megan Washington
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Carolina Leynes
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Jason Hsu
- Department of Biochemistry, Cell and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kempaiah Rayavara
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Yangjin Bae
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Nele Haelterman
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Yuqing Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Ming-Ming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Aleksandra Drelich
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Vivian Tat
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Denise G Lanza
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Isabel Lorenzo
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Jason D Heaney
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Chien-Te Kent Tseng
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Ronit Marom
- Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA.
| |
Collapse
|
4
|
Drelich AK, Rayavara K, Hsu J, Saenkham-Huntsinger P, Judy BM, Tat V, Ksiazek TG, Peng BH, Tseng CTK. Characterization of Unique Pathological Features of COVID-Associated Coagulopathy: Studies with AC70 hACE2 Transgenic Mice Highly Permissive to SARS-CoV-2 Infection. PLoS Pathog 2024; 20:e1011777. [PMID: 38913740 PMCID: PMC11226087 DOI: 10.1371/journal.ppat.1011777] [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: 10/27/2023] [Revised: 07/05/2024] [Accepted: 06/10/2024] [Indexed: 06/26/2024] Open
Abstract
COVID-associated coagulopathy seemly plays a key role in post-acute sequelae of SARS- CoV-2 infection. However, the underlying pathophysiological mechanisms are poorly understood, largely due to the lack of suitable animal models that recapitulate key clinical and pathological symptoms. Here, we fully characterized AC70 line of human ACE2 transgenic (AC70 hACE2 Tg) mice for SARS-CoV-2 infection. We noted that this model is highly permissive to SARS-CoV-2 with values of 50% lethal dose and infectious dose as ~ 3 and ~ 0.5 TCID50 of SARS-CoV-2, respectively. Mice infected with 105 TCID50 of SARS-CoV-2 rapidly succumbed to infection with 100% mortality within 5 days. Lung and brain were the prime tissues harboring high viral titers, accompanied by histopathology. However, viral RNA and inflammatory mediators could be detectable in other organs, suggesting the nature of a systemic infection. Lethal challenge of AC70 hACE2 Tg mice caused acute onset of leukopenia, lymphopenia, along with an increased neutrophil-to-lymphocyte ratio (NLR). Importantly, infected animals recapitulated key features of COVID-19-associated coagulopathy. SARS-CoV-2 could induce the release of circulating neutrophil extracellular traps (NETs), along with activated platelet/endothelium marker. Immunohistochemical staining with anti-platelet factor-4 (PF4) antibody revealed profound platelet aggregates especially within blocked veins of the lungs. We showed that acute SARS-CoV-2 infection triggered a hypercoagulable state coexisting with ill-regulated fibrinolysis. Finally, we highlighted the potential role of Annexin A2 (ANXA2) in fibrinolytic failure. ANXA2 is a calcium-dependent phospholipid-binding protein that forms a heterotertrameric complexes localized at the extracellular membranes with two S100A10 small molecules acting as a co-receptor for tissue-plasminogen activator (t-PA), tightly involved in cell surface fibrinolysis. Thus, our results revealing elevated IgG type anti-ANXA2 antibody production, downregulated de novo ANXA2/S100A10 synthesis, and reduced ANXA2/S100A10 association in infected mice, this protein might serve as druggable targets for development of antithrombotic and/or anti-fibrinolytic agents to attenuate pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Aleksandra K. Drelich
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kempaiah Rayavara
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jason Hsu
- Department of Biochemistry, Cell and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Panatda Saenkham-Huntsinger
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Barbara M. Judy
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Vivian Tat
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Thomas G. Ksiazek
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
- World Reference Center for Emerging Viruses and Arboviruses, Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bi-Hung Peng
- Neurosciences, Cell Biology, and Anatomy, University of Texas Medical Branch Galveston, Texas, United States of America
| | - Chien-Te K. Tseng
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Biochemistry, Cell and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
5
|
Wang Y, Ye M, Zhang F, Freeman ZT, Yu H, Ye X, He Y. Ontology-based taxonomical analysis of experimentally verified natural and laboratory human coronavirus hosts and its implication for COVID-19 virus origination and transmission. PLoS One 2024; 19:e0295541. [PMID: 38252647 PMCID: PMC10802970 DOI: 10.1371/journal.pone.0295541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 11/26/2023] [Indexed: 01/24/2024] Open
Abstract
To fully understand COVID-19, it is critical to study all possible hosts of SARS-CoV-2 (the pathogen of COVID-19). In this work, we collected, annotated, and performed ontology-based taxonomical analysis of all the reported and verified hosts for all human coronaviruses including SARS-CoV, MERS-CoV, SARS-CoV-2, HCoV-229E, HCoV-NL63, HCoV-OC43, and HCoV-HKU1. A total of 37 natural hosts and 19 laboratory animal hosts of human coronaviruses were identified based on experimental evidence. Our analysis found that all the verified susceptible natural and laboratory animals belong to therian mammals. Specifically, these 37 natural therian hosts include one wildlife marsupial mammal (i.e., Virginia opossum) and 36 Eutheria mammals (a.k.a. placental mammals). The 19 laboratory animal hosts are also classified as therian mammals. The mouse models with genetically modified human ACE2 or DPP4 were more susceptible to virulent human coronaviruses with clear symptoms, suggesting the critical role of ACE2 and DPP4 to coronavirus virulence. Coronaviruses became more virulent and adaptive in the mouse hosts after a series of viral passages in the mice, providing clue to the possible coronavirus origination. The Huanan Seafood Wholesale Market animals identified early in the COVID-19 outbreak were also systematically analyzed as possible COVID-19 hosts. To support knowledge standardization and query, the annotated host knowledge was modeled and represented in the Coronavirus Infectious Disease Ontology (CIDO). Based on our and others' findings, we further propose a MOVIE model (i.e., Multiple-Organism viral Variations and Immune Evasion) to address how viral variations in therian animal hosts and the host immune evasion might have led to dynamic COVID-19 pandemic outcomes.
Collapse
Affiliation(s)
- Yang Wang
- Guizhou University School of Medicine, Guiyang, Guizhou, China
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People’s Hospital and NHC Key Laboratory of Immunological Diseases, People’s Hospital of Guizhou University, Guiyang, Guizhou, China
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Muhui Ye
- Chinese University of Hong Kong (Shenzhen), Shenzhen, Guangdong, China
| | - Fengwei Zhang
- Guizhou University School of Medicine, Guiyang, Guizhou, China
| | - Zachary Thomas Freeman
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Hong Yu
- Guizhou University School of Medicine, Guiyang, Guizhou, China
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People’s Hospital and NHC Key Laboratory of Immunological Diseases, People’s Hospital of Guizhou University, Guiyang, Guizhou, China
| | - Xianwei Ye
- Guizhou University School of Medicine, Guiyang, Guizhou, China
- Department of Respiratory and Critical Care Medicine, Guizhou Provincial People’s Hospital and NHC Key Laboratory of Immunological Diseases, People’s Hospital of Guizhou University, Guiyang, Guizhou, China
| | - Yongqun He
- Unit for Laboratory Animal Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI, United States of America
- Center for Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, United States of America
| |
Collapse
|
6
|
Babaeimarzangou SS, Zaker H, Soleimannezhadbari E, Gamchi NS, Kazeminia M, Tarighi S, Seyedian H, Tsatsakis A, Spandidos DA, Margina D. Vaccine development for zoonotic viral diseases caused by positive‑sense single‑stranded RNA viruses belonging to the Coronaviridae and Togaviridae families (Review). Exp Ther Med 2022; 25:42. [PMID: 36569444 PMCID: PMC9768462 DOI: 10.3892/etm.2022.11741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 11/10/2022] [Indexed: 12/02/2022] Open
Abstract
Outbreaks of zoonotic viral diseases pose a severe threat to public health and economies worldwide, with this currently being more prominent than it previously was human history. These emergency zoonotic diseases that originated and transmitted from vertebrates to humans have been estimated to account for approximately one billion cases of illness and have caused millions of deaths worldwide annually. The recent emergence of severe acute respiratory syndrome coronavirus-2 (coronavirus disease 2019) is an excellent example of the unpredictable public health threat causing a pandemic. The present review summarizes the literature data regarding the main vaccine developments in human clinical phase I, II and III trials against the zoonotic positive-sense single-stranded RNA viruses belonging to the Coronavirus and Alphavirus genera, including severe acute respiratory syndrome, Middle east respiratory syndrome, Venezuelan equine encephalitis virus, Semliki Forest virus, Ross River virus, Chikungunya virus and O'nyong-nyong virus. That there are neither vaccines nor effective antiviral drugs available against most of these viruses is undeniable. Therefore, new explosive outbreaks of these zoonotic viruses may surely be expected. The present comprehensive review provides an update on the status of vaccine development in different clinical trials against these viruses, as well as an overview of the present results of these trials.
Collapse
Affiliation(s)
- Seyed Sajjad Babaeimarzangou
- Division of Poultry Health and Diseases, Department of Clinical Sciences, Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Himasadat Zaker
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | | | - Naeimeh Shamsi Gamchi
- Histology and Microscopic Analysis Division, RASTA Specialized Research Institute (RSRI), West Azerbaijan Science and Technology Park (WASTP), Urmia 5756115322, Iran
| | - Masoud Kazeminia
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran 1417935840, Iran
| | - Shima Tarighi
- Veterinary Office of West Azerbaijan Province, Urmia 5717617695, Iran
| | - Homayon Seyedian
- Faculty of Veterinary Medicine, Urmia University, Urmia 5756151818, Iran
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Department of Medicine, University of Crete, 71307 Heraklion, Greece,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| | - Demetrios A. Spandidos
- Laboratory of Clinical Virology, School of Medicine, University of Crete, 71003 Heraklion, Greece
| | - Denisa Margina
- Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 020956 Bucharest, Romania,Correspondence to: Professor Denisa Margina, Department of Biochemistry, Faculty of Pharmacy, ‘Carol Davila’ University of Medicine and Pharmacy, 6 Traian Vuia Street, 020956 Bucharest, Romania
| |
Collapse
|
7
|
Ning T, Liu S, Xu J, Yang Y, Zhang N, Xie S, Min L, Zhang S, Zhu S, Wang Y. Potential intestinal infection and faecal-oral transmission of human coronaviruses. Rev Med Virol 2022; 32:e2363. [PMID: 35584273 PMCID: PMC9348496 DOI: 10.1002/rmv.2363] [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: 10/27/2021] [Revised: 02/25/2022] [Accepted: 05/06/2022] [Indexed: 01/08/2023]
Abstract
Human coronaviruses (HCoVs) were first described in 1960s for patients experiencing common cold. Since then, increasing number of HCoVs have been discovered, including those causing severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and the circulating coronavirus disease 2019 (COVID‐19), which can cause fatal respiratory disease in humans on infection. HCoVs are believed to spread mainly through respiratory droplets and close contact. However, studies have shown that a large proportion of patients with HCoV infection develop gastrointestinal (GI) symptoms, and many patients with confirmed HCoV infection have shown detectable viral RNA in their faecal samples. Furthermore, multiple in vitro and in vivo animal studies have provided direct evidence of intestinal HCoV infection. These data highlight the nature of HCoV GI infection and its potential faecal‐oral transmission. Here, we summarise the current findings on GI manifestations of HCoVs. We also discuss how HCoV GI infection might occur and the current evidence to establish the occurrence of faecal‐oral transmission.
Collapse
Affiliation(s)
- Tingting Ning
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Si Liu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Junxuan Xu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Yi Yang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Nan Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Sian Xie
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Li Min
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Shutian Zhang
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Shengtao Zhu
- Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, National Clinical Research Center for Digestive Disease, Beijing Digestive Disease Center, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Beijing, China
| | - Youchun Wang
- Division of HIV/AIDS and Sexually Transmitted Virus Vaccines, National Institutes for Food and Drug Control (NIFDC), Beijing, China
| |
Collapse
|
8
|
Yap TF, Hsu JC, Liu Z, Rayavara K, Tat V, Tseng CTK, Preston DJ. Efficacy and self-similarity of SARS-CoV-2 thermal decontamination. JOURNAL OF HAZARDOUS MATERIALS 2022; 429:127709. [PMID: 35086724 PMCID: PMC8572375 DOI: 10.1016/j.jhazmat.2021.127709] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/01/2021] [Accepted: 11/02/2021] [Indexed: 06/14/2023]
Abstract
Dry heat decontamination has been shown to effectively inactivate viruses without compromising the integrity of delicate personal protective equipment (PPE), allowing safe reuse and helping to alleviate shortages of PPE that have arisen due to COVID-19. Unfortunately, current thermal decontamination guidelines rely on empirical data which are often sparse, limited to a specific virus, and unable to provide fundamental insight into the underlying inactivation reaction. In this work, we experimentally quantified dry heat decontamination of SARS-CoV-2 on disposable masks and validated a model that treats the inactivation reaction as thermal degradation of macromolecules. Furthermore, upon nondimensionalization, all of the experimental data collapse onto a unified curve, revealing that the thermally driven decontamination process exhibits self-similar behavior. Our results show that heating surgical masks to 70 °C for 5 min inactivates over 99.9% of SARS-CoV-2. We also characterized the chemical and physical properties of disposable masks after heat treatment and did not observe degradation. The model presented in this work enables extrapolation of results beyond specific temperatures to provide guidelines for safe PPE decontamination. The modeling framework and self-similar behavior are expected to extend to most viruses-including yet-unencountered novel viruses-while accounting for a range of environmental conditions.
Collapse
Affiliation(s)
- Te Faye Yap
- Department of Mechanical Engineering, George R. Brown School of Engineering, Rice University, 6100 Main St., Houston, TX 77005, USA
| | - Jason C Hsu
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA
| | - Zhen Liu
- Department of Mechanical Engineering, George R. Brown School of Engineering, Rice University, 6100 Main St., Houston, TX 77005, USA
| | - Kempaiah Rayavara
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA
| | - Vivian Tat
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA
| | - Chien-Te K Tseng
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA; Center for Biodefense and Emerging Diseases, Galveston National Laboratory, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555, USA
| | - Daniel J Preston
- Department of Mechanical Engineering, George R. Brown School of Engineering, Rice University, 6100 Main St., Houston, TX 77005, USA.
| |
Collapse
|
9
|
Vlaskin MS. Review of air disinfection approaches and proposal for thermal inactivation of airborne viruses as a life-style and an instrument to fight pandemics. APPLIED THERMAL ENGINEERING 2022; 202:117855. [PMID: 34867067 PMCID: PMC8628600 DOI: 10.1016/j.applthermaleng.2021.117855] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/20/2021] [Accepted: 11/24/2021] [Indexed: 05/02/2023]
Abstract
COVID-19 (Coronavirus Disease 2019) pandemic highlighted the importance of air biosecurity because SARS-CoV-2 is mainly transmitted from person to person via airborne droplets. Preventing infectious droplets from entering the body is one of the best ways to protect against infection. This paper reviews the transmission patterns of airborne pathogens and air disinfection methods. A particular emphasis is put on studies devoted to the thermal inactivation of viruses. These reviews reveal that air heat treatment has not been seriously considered as a possible air disinfection approach. Simple calculations show that the energy input required for thermal disinfection of human's air daily consumption is almost the same as for daily water consumption (by heat treatment from room temperature to 100 °C). Moreover, it is possible to organize a continuous heat recovery from the air already heated during disinfection to the inlet air, thus significantly increasing the energy efficiency. Therefore, I propose a solution for the thermal inactivation of airborne pathogens based on air heating and its subsequent cooling in a heat exchanger with heat recovery. Such a solution could be used to create mobile personal and stationary indoor air disinfectors, as well as heating, ventilation, and air conditioning systems. Thermal disinfection of air to breathe might one day be part of people's daily life like thermal disinfection of drinking water. Aside from limiting infectious disease transmission, thermal inactivation might be the basis for developing inhaled vaccines using thermally inactivated whole pathogens.
Collapse
Affiliation(s)
- Mikhail S Vlaskin
- Joint Institute for High Temperatures of the Russian Academy of Sciences, 13/2 Izhorskaya St, Moscow 125412, Russia
| |
Collapse
|
10
|
Implications of testicular ACE2 and the renin-angiotensin system for SARS-CoV-2 on testis function. Nat Rev Urol 2022; 19:116-127. [PMID: 34837081 PMCID: PMC8622117 DOI: 10.1038/s41585-021-00542-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/26/2021] [Indexed: 12/16/2022]
Abstract
Although many studies have focused on SARS-CoV-2 infection in the lungs, comparatively little is known about the potential effects of the virus on male fertility. SARS-CoV-2 infection of target cells requires the presence of furin, angiotensin-converting enzyme 2 (ACE2) receptors, and transmembrane protease serine 2 (TMPRSS2). Thus, cells in the body that express these proteins might be highly susceptible to viral entry and downstream effects. Currently, reports regarding the expression of the viral entry proteins in the testes are conflicting; however, other members of the SARS-CoV family of viruses - such as SARS-CoV - have been suspected to cause testicular dysfunction and/or orchitis. SARS-CoV-2, which displays many similarities to SARS-CoV, could potentially cause similar adverse effects. Commonalities between SARS family members, taken in combination with sparse reports of testicular discomfort and altered hormone levels in patients with SARS-CoV-2, might indicate possible testicular dysfunction. Thus, SARS-CoV-2 infection has the potential for effects on testis somatic and germline cells and experimental approaches might be required to help identify potential short-term and long-term effects of SARS-CoV-2 on male fertility.
Collapse
|
11
|
Singh S, Kumar A, Sharma H. In-vitro and In-vivo Experimental Models for MERS-CoV, SARSCoV, and SARS-CoV-2 Viral Infection: A Compendious Review. Recent Pat Biotechnol 2022; 16:82-101. [PMID: 35068398 DOI: 10.2174/1872208316666220124101611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/01/2021] [Accepted: 12/06/2021] [Indexed: 06/14/2023]
Abstract
SARS-CoV-2 belongs to the Coronaviridae family of coronaviruses. This novel virus has predominantly affected a vast world population and was declared a pandemic outbreak. The clinical and scientific communities strive to develop and validate potential treatments and therapeutic measures. The comparative study of existing synthetic drugs, evaluation of safety aspects, and the devel opment of novel vaccines can be efficiently achieved by using suitable animal models of primary infection and validating translational findings in human cell lines and tissues. The current paper explores varied animal and cell/tissue models employed and recapitulate various critical issues of ailment manifestation in humans to develop and evaluate novel therapeutic countermeasures and even include some novel patent developed in this regard.
Collapse
Affiliation(s)
- Sonia Singh
- Institute of Pharmaceutical Research, GLA University, 17 km Stone, NH-2, Mathura-Delhi Road, Mathura, Chaumuhan, Uttar Pradesh-281406, India
| | - Aman Kumar
- Institute of Pharmaceutical Research, GLA University, 17 km Stone, NH-2, Mathura-Delhi Road, Mathura, Chaumuhan, Uttar Pradesh-281406, India
| | - Himanshu Sharma
- Department of Computer Engineering and Applications, GLA University, 17 km Stone, NH-2, Mathura-Delhi Road Mathura, Chaumuhan, Uttar Pradesh-281406, India
| |
Collapse
|
12
|
London V, Blitz MJ, Solmonovich R, Silver M, Minkoff H. Temporal Trend in SARS-CoV-2 Symptoms in Pregnant Women. Am J Perinatol 2021. [PMID: 34918329 DOI: 10.1055/s-0041-1739490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
OBJECTIVE The objective of this study was to examine temporal trends in the clinical presentation of patients diagnosed with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in pregnancy. STUDY DESIGN This is a retrospective cohort study of pregnant women who were universally screened for SARS-CoV-2 and tested positive. This multi-center study of admissions to labor and delivery units in New York City and Long Island included all SARS-CoV-2-infected pregnant women admitted to labor and delivery units between April 10th and June 4th 2020. Six Northwell Health hospitals and Maimonides Medical Center were included in the study. The main measures of the study included patient reports of COVID-19 symptoms: fever, cough, chest pain, shortness of breath, nausea, vomiting, and intensive care unit (ICU) admissions. The main outcome measure was the percentage of all infected women who reported any of the above symptoms. RESULTS In total, 427 infected pregnant women were included in the study. There was a statistically significant decline in the percentage of patients presenting with any symptoms over the course of the study. In addition, disease severity, symptoms of fever, cough, and chest pain/shortness of breath also significantly declined over time, and no ICU admissions were noted after the third week of April. CONCLUSIONS There was a temporal shift away from symptomatic presentation in pregnant women diagnosed with SARS-CoV-2 over the course of the first months of the epidemic in New York. Further studies are necessary to elucidate the cause of this change in presentation among pregnant women, to determine whether this trend is also observed in other patient populations. KEY POINTS · Retrospective cohort review of 427 SARS-CoV-2-infected pregnant women admitted to labor and delivery units.. · A significant decline in the percentage of patients presenting with symptoms over time was noted.. · Further studies are necessary to elucidate the cause of this change in presentation.. · Theories for the noted trend: viral evolution, decreased viral inoculums, and prolonged polymerase chain reaction positivity..
Collapse
Affiliation(s)
- Viktoriya London
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Maimonides Medical Center, Brooklyn, New York
| | - Matthew J Blitz
- Department of Obstetrics and Gynecology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York.,Division of Maternal-Fetal Medicine, Southside Hospital, Bay Shore, New York
| | - Rachel Solmonovich
- Department of Obstetrics and Gynecology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, New York
| | - Michael Silver
- Department of Obstetrics and Gynecology, SUNY Downstate Medical Center, Brooklyn, New York
| | - Howard Minkoff
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Maimonides Medical Center, Brooklyn, New York.,Department of Obstetrics and Gynecology, SUNY Downstate Medical Center, Brooklyn, New York
| |
Collapse
|
13
|
Saadi F, Pal D, Sarma JD. Spike Glycoprotein Is Central to Coronavirus Pathogenesis-Parallel Between m-CoV and SARS-CoV-2. Ann Neurosci 2021; 28:201-218. [PMID: 35341224 PMCID: PMC8948335 DOI: 10.1177/09727531211023755] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 03/24/2021] [Indexed: 01/04/2023] Open
Abstract
Coronaviruses (CoVs) are single-stranded, polyadenylated, enveloped RNA of positive polarity with a unique potential to alter host tropism. This has been exceptionally demonstrated by the emergence of deadly virus outbreaks of the past: Severe Acute Respiratory Syndrome (SARS-CoV) in 2003 and Middle East Respiratory Syndrome (MERS-CoV) in 2012. The 2019 outbreak by the new cross-species transmission of SARS-CoV-2 has put the world on alert. CoV infection is triggered by receptor recognition, membrane fusion, and successive viral entry mediated by the surface Spike (S) glycoprotein. S protein is one of the major antigenic determinants and the target for neutralizing antibodies. It is a valuable target in antiviral therapies because of its central role in cell-cell fusion, viral antigen spread, and host immune responses leading to immunopathogenesis. The receptor-binding domain of S protein has received greater attention as it initiates host attachment and contains major antigenic determinants. However, investigating the therapeutic potential of fusion peptide as a part of the fusion core complex assembled by the heptad repeats 1 and 2 (HR1 and HR2) is also warranted. Along with receptor attachment and entry, fusion mechanisms should also be explored for designing inhibitors as a therapeutic intervention. In this article, we review the S protein function and its role in mediating membrane fusion, spread, tropism, and its associated pathogenesis with notable therapeutic strategies focusing on results obtained from studies on a murine β-Coronavirus (m-CoV) and its associated disease process.
Collapse
Affiliation(s)
- Fareeha Saadi
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Kolkata, West Bengal, India
| | - Debnath Pal
- Department of Computational and Data Sciences, Indian Institute of Science, Bengaluru, Karnataka, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Kolkata, West Bengal, India
- Department of Ophthalmology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
14
|
Kaundal RK, Kalvala AK, Kumar A. Neurological Implications of COVID-19: Role of Redox Imbalance and Mitochondrial Dysfunction. Mol Neurobiol 2021; 58:4575-4587. [PMID: 34110602 PMCID: PMC8190166 DOI: 10.1007/s12035-021-02412-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Accepted: 04/29/2021] [Indexed: 12/20/2022]
Abstract
Severe acute respiratory syndrome coronavirus (SARS-CoV)-2 or COVID-19 has been declared as a pandemic disease by the World Health Organization (WHO). Globally, this disease affected 159 million of the population and reported ~ 3.3 million deaths to the current date (May 2021). There is no definitive treatment strategy that has been identified, although this disease has prevailed in its current form for the past 18 months. The main challenges in the (SARS-CoV)-2 infections are in identifying the heterogeneity in viral strains and the plausible mechanisms of viral infection to human tissues. In parallel to the investigations into the patho-mechanism of SARS-CoV-2 infection, understanding the fundamental processes underlying the clinical manifestations of COVID-19 is very crucial for designing effective therapies. Since neurological symptoms are very apparent in COVID-19 infected patients, here, we tried to emphasize the involvement of redox imbalance and subsequent mitochondrial dysfunction in the progression of the COVID-19 infection. It has been articulated that mitochondrial dysfunction is very apparent and also interlinked to neurological symptoms in COVID-19 infection. Overall, this article provides an in-depth overview of redox imbalance and mitochondrial dysfunction involvement in aggravating COVID-19 infection and its probable contribution to the neurological manifestation of the disease.
Collapse
Affiliation(s)
- Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Raebareli, Lucknow, India
- Icahn School of Medicine At Mount Sinai, 1470 Madison Ave, New York, NY, USA
| | - Anil K Kalvala
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, FL, North America, USA
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Kolkata, Kolkata, India.
| |
Collapse
|
15
|
Cajanding RJM. Comprehensive Review of Cardiovascular Involvement in COVID-19. AACN Adv Crit Care 2021; 32:169-187. [PMID: 33836538 DOI: 10.4037/aacnacc2021302] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
COVID-19 has emerged as one of the most devastating and clinically significant infectious diseases of the last decade. It has reached global pandemic status at an unprecedented pace and has placed significant demands on health care systems worldwide. Although COVID-19 primarily affects the lungs, epidemiologic reports have shown that the disease affects other vital organs of the body, including the heart, vasculature, kidneys, brain, and the hematopoietic system. Of importance is the emerging awareness of the effects of COVID-19 on the cardiovascular system. The current state of knowledge regarding cardiac involvement in COVID-19 is presented in this article, with particular focus on the cardiovascular manifestations and complications of COVID-19 infection. The mechanistic insights of disease causation and the relevant pathophysiology involved in COVID-19 as they affect the heart are explored and described. Relevant practice essentials and clinical management implications for patients with COVID-19 with a cardiac pathology are presented in light of recent evidence.
Collapse
Affiliation(s)
- Ruff Joseph Macale Cajanding
- Ruff Joseph Macale Cajanding is Charge Nurse, Adult Critical Care Unit, 6th Floor, King George V Building, St Bartholomew's Hospital, Barts Health NHS Trust, 2 King Edward Street, London EC1A 1HQ, United Kingdom
| |
Collapse
|
16
|
Karimzadeh S, Bhopal R, Nguyen Tien H. Review of infective dose, routes of transmission and outcome of COVID-19 caused by the SARS-COV-2: comparison with other respiratory viruses. Epidemiol Infect 2021; 149:e96. [PMID: 33849679 DOI: 10.20944/preprints202007.0613.v3] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/21/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is pandemic. Prevention and control strategies require an improved understanding of SARS-CoV-2 dynamics. We did a rapid review of the literature on SARS-CoV-2 viral dynamics with a focus on infective dose. We sought comparisons of SARS-CoV-2 with other respiratory viruses including SARS-CoV-1 and Middle East respiratory syndrome coronavirus. We examined laboratory animal and human studies. The literature on infective dose, transmission and routes of exposure was limited specially in humans, and varying endpoints were used for measurement of infection. Despite variability in animal studies, there was some evidence that increased dose at exposure correlated with higher viral load clinically, and severe symptoms. Higher viral load measures did not reflect coronavirus disease 2019 severity. Aerosol transmission seemed to raise the risk of more severe respiratory complications in animals. An accurate quantitative estimate of the infective dose of SARS-CoV-2 in humans is not currently feasible and needs further research. Our review suggests that it is small, perhaps about 100 particles. Further work is also required on the relationship between routes of transmission, infective dose, co-infection and outcomes.
Collapse
Affiliation(s)
- Sedighe Karimzadeh
- School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Raj Bhopal
- Usher Institute, University of Edinburgh, EdinburghEH3 9AG, UK
| | - Huy Nguyen Tien
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
17
|
Karimzadeh S, Bhopal R, Nguyen Tien H. Review of infective dose, routes of transmission and outcome of COVID-19 caused by the SARS-COV-2: comparison with other respiratory viruses. Epidemiol Infect 2021; 149:e96. [PMID: 33849679 PMCID: PMC8082124 DOI: 10.1017/s0950268821000790] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2020] [Revised: 03/13/2021] [Accepted: 04/09/2021] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is pandemic. Prevention and control strategies require an improved understanding of SARS-CoV-2 dynamics. We did a rapid review of the literature on SARS-CoV-2 viral dynamics with a focus on infective dose. We sought comparisons of SARS-CoV-2 with other respiratory viruses including SARS-CoV-1 and Middle East respiratory syndrome coronavirus. We examined laboratory animal and human studies. The literature on infective dose, transmission and routes of exposure was limited specially in humans, and varying endpoints were used for measurement of infection. Despite variability in animal studies, there was some evidence that increased dose at exposure correlated with higher viral load clinically, and severe symptoms. Higher viral load measures did not reflect coronavirus disease 2019 severity. Aerosol transmission seemed to raise the risk of more severe respiratory complications in animals. An accurate quantitative estimate of the infective dose of SARS-CoV-2 in humans is not currently feasible and needs further research. Our review suggests that it is small, perhaps about 100 particles. Further work is also required on the relationship between routes of transmission, infective dose, co-infection and outcomes.
Collapse
Affiliation(s)
- Sedighe Karimzadeh
- School of Medicine, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Raj Bhopal
- Usher Institute, University of Edinburgh, EdinburghEH3 9AG, UK
| | - Huy Nguyen Tien
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| |
Collapse
|
18
|
Molaei S, Dadkhah M, Asghariazar V, Karami C, Safarzadeh E. The immune response and immune evasion characteristics in SARS-CoV, MERS-CoV, and SARS-CoV-2: Vaccine design strategies. Int Immunopharmacol 2021; 92:107051. [PMID: 33429331 PMCID: PMC7522676 DOI: 10.1016/j.intimp.2020.107051] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/24/2020] [Accepted: 09/24/2020] [Indexed: 01/25/2023]
Abstract
The worldwide outbreak of SARS-CoV-2, severe acute respiratory syndrome coronavirus 2 as a novel human coronavirus, was the worrying news at the beginning of 2020. Since its emergence complicated more than 870,000 individuals and led to more than 43,000 deaths worldwide. Considering to the potential threat of a pandemic and transmission severity of it, there is an urgent need to evaluate and realize this new virus's structure and behavior and the immunopathology of this disease to find potential therapeutic protocols and to design and develop effective vaccines. This disease is able to agitate the response of the immune system in the infected patients, so ARDS, as a common consequence of immunopathological events for infections with Middle East respiratory syndrome coronavirus (MERS-CoV), SARS-CoV, and SARS-CoV-2, could be the main reason for death. Here, we summarized the immune response and immune evasion characteristics in SARS-CoV, MERS-CoV, and SARS-CoV-2 and therapeutic and prophylactic strategies with a focus on vaccine development and its challenges.
Collapse
Affiliation(s)
- Soheila Molaei
- Deputy of Research & Technology, Ardabil University of Medical Sciences, Ardabil, Iran; Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Masoomeh Dadkhah
- Pharmaceutical Sciences Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Pharmacology, School of Pharmacy, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Vahid Asghariazar
- Deputy of Research & Technology, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Chiman Karami
- Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Department of Microbiology, Parasitology, and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
19
|
Yong SJ. Persistent Brainstem Dysfunction in Long-COVID: A Hypothesis. ACS Chem Neurosci 2021; 12:573-580. [PMID: 33538586 PMCID: PMC7874499 DOI: 10.1021/acschemneuro.0c00793] [Citation(s) in RCA: 132] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 02/01/2021] [Indexed: 02/07/2023] Open
Abstract
Long-COVID is a postviral illness that can affect survivors of COVID-19, regardless of initial disease severity or age. Symptoms of long-COVID include fatigue, dyspnea, gastrointestinal and cardiac problems, cognitive impairments, myalgia, and others. While the possible causes of long-COVID include long-term tissue damage, viral persistence, and chronic inflammation, the review proposes, perhaps for the first time, that persistent brainstem dysfunction may also be involved. This hypothesis can be split into two parts. The first is the brainstem tropism and damage in COVID-19. As the brainstem has a relatively high expression of ACE2 receptor compared with other brain regions, SARS-CoV-2 may exhibit tropism therein. Evidence also exists that neuropilin-1, a co-receptor of SARS-CoV-2, may be expressed in the brainstem. Indeed, autopsy studies have found SARS-CoV-2 RNA and proteins in the brainstem. The brainstem is also highly prone to damage from pathological immune or vascular activation, which has also been observed in autopsy of COVID-19 cases. The second part concerns functions of the brainstem that overlap with symptoms of long-COVID. The brainstem contains numerous distinct nuclei and subparts that regulate the respiratory, cardiovascular, gastrointestinal, and neurological processes, which can be linked to long-COVID. As neurons do not readily regenerate, brainstem dysfunction may be long-lasting and, thus, is long-COVID. Indeed, brainstem dysfunction has been implicated in other similar disorders, such as chronic pain and migraine and myalgic encephalomyelitis or chronic fatigue syndrome.
Collapse
Affiliation(s)
- Shin Jie Yong
- Department of Biological
Sciences, Sunway University, Petaling Jaya, Selangor 47500, Malaysia
| |
Collapse
|
20
|
Zhou P, Li Z, Xie L, An D, Fan Y, Wang X, Li Y, Liu X, Wu J, Li G, Li Q. Research progress and challenges to coronavirus vaccine development. J Med Virol 2021; 93:741-754. [PMID: 32936465 DOI: 10.1002/jmv.26517] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 07/29/2020] [Accepted: 08/06/2020] [Indexed: 01/07/2023]
Abstract
Coronaviruses (CoVs) are nonsegmented, single-stranded, positive-sense RNA viruses highly pathogenic to humans. Some CoVs are known to cause respiratory and intestinal diseases, posing a threat to the global public health. Against this backdrop, it is of critical importance to develop safe and effective vaccines against these CoVs. This review discusses human vaccine candidates in any stage of development and explores the viral characteristics, molecular epidemiology, and immunology associated with CoV vaccine development. At present, there are many obstacles and challenges to vaccine research and development, including the lack of knowledge about virus transmission, pathogenesis, and immune response, absence of the most appropriate animal models.
Collapse
Affiliation(s)
- Peiwen Zhou
- Department of Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Zonghui Li
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Linqing Xie
- Department of Guangzhou Cyanvaccine Biotechnology Co, Ltd, Guangzhou, China
| | - Dong An
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Yaohua Fan
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiao Wang
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yiwei Li
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xiaohong Liu
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianguo Wu
- Department of Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Geng Li
- Department of Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- Department of Laboratory Animal Center, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qin Li
- Department of Guangzhou Cyanvaccine Biotechnology Co, Ltd, Guangzhou, China
| |
Collapse
|
21
|
Middle East Respiratory Syndrome Coronavirus Gene 5 Modulates Pathogenesis in Mice. J Virol 2021; 95:JVI.01172-20. [PMID: 33144319 DOI: 10.1128/jvi.01172-20] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 07/08/2020] [Indexed: 12/15/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) causes a highly lethal pneumonia that emerged in 2012. There is limited information on MERS-CoV pathogenesis, as data from patients are scarce and the generation of animal models reproducing MERS clinical manifestations has been challenging. Human dipeptidyl peptidase 4 knock-in (hDPP4-KI) mice and a mouse-adapted MERS-CoV strain (MERSMA-6-1-2) were recently described. hDPP4-KI mice infected with MERSMA-6-1-2 show pathological signs of respiratory disease, high viral titers in the lung, and death. In this work, a mouse-adapted MERS-CoV infectious cDNA was engineered by introducing nonsynonymous mutations contained in the MERSMA-6-1-2 genome into a MERS-CoV infectious cDNA, leading to a recombinant mouse-adapted virus (rMERS-MA) that was virulent in hDDP4-KI mice. MERS-CoV adaptation to cell culture or mouse lungs led to mutations and deletions in genus-specific gene 5 that prevented full-length protein expression. In contrast, analysis of 476 MERS-CoV field isolates showed that gene 5 is highly stable in vivo in both humans and camels. To study the role of protein 5, two additional viruses were engineered expressing a full-length gene 5 (rMERS-MA-5FL) or containing a complete gene 5 deletion (rMERS-MA-Δ5). rMERS-MA-5FL virus was unstable, as deletions appeared during passage in different tissue culture cells, highlighting MERS-CoV instability. The virulence of rMERS-MA-Δ5 was analyzed in a sublethal hDPP4-KI mouse model. Unexpectedly, all mice died after infection with rMERS-MA-Δ5, in contrast to those infected with the parental virus, which contains a 17-nucleotide (nt) deletion and a stop codon in protein 5 at position 108. Expression of interferon and proinflammatory cytokines was delayed and dysregulated in the lungs of rMERS-MA-Δ5-infected mice. Overall, these data indicated that the rMERS-MA-Δ5 virus was more virulent than the parental one and suggest that the residual gene 5 sequence present in the mouse-adapted parental virus had a function in ameliorating severe MERS-CoV pathogenesis.IMPORTANCE Middle East respiratory syndrome coronavirus (MERS-CoV) is a zoonotic virus causing human infections with high mortality rate (∼35%). Animal models together with reverse-genetics systems are essential to understand MERS-CoV pathogenesis. We developed a reverse-genetics system for a mouse-adapted MERS-CoV that reproduces the virus behavior observed in humans. This system is highly useful to investigate the role of specific viral genes in pathogenesis. In addition, we described a virus lacking gene 5 expression that is more virulent than the parental one. The data provide novel functions in IFN modulation for gene 5 in the context of viral infection and will help to develop novel antiviral strategies.
Collapse
|
22
|
Liang Z, Zhu H, Wang X, Jing B, Li Z, Xia X, Sun H, Yang Y, Zhang W, Shi L, Zeng H, Sun B. Adjuvants for Coronavirus Vaccines. Front Immunol 2020; 11:589833. [PMID: 33240278 PMCID: PMC7677582 DOI: 10.3389/fimmu.2020.589833] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/14/2020] [Indexed: 12/20/2022] Open
Abstract
Vaccine development utilizing various platforms is one of the strategies that has been proposed to address the coronavirus disease 2019 (COVID-19) pandemic. Adjuvants are critical components of both subunit and certain inactivated vaccines because they induce specific immune responses that are more robust and long-lasting. A review of the history of coronavirus vaccine development demonstrates that only a few adjuvants, including aluminum salts, emulsions, and TLR agonists, have been formulated for the severe acute respiratory syndrome-associated coronavirus (SARS-CoV), Middle East respiratory syndrome-related coronavirus (MERS-CoV), and currently the SARS-CoV-2 vaccines in experimental and pre-clinical studies. However, there is still a lack of evidence regarding the effects of the adjuvants tested in coronavirus vaccines. This paper presents an overview of adjuvants that have been formulated in reported coronavirus vaccine studies, which should assist with the design and selection of adjuvants with optimal efficacy and safety profiles for COVID-19 vaccines.
Collapse
Affiliation(s)
- Zhihui Liang
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, China
- School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Haoru Zhu
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, China
- School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Xin Wang
- School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Bo Jing
- School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Zifan Li
- School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Xinyu Xia
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, China
- School of Chemical Engineering, Dalian University of Technology, Dalian, China
| | - Hongwu Sun
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Yun Yang
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Weiting Zhang
- NCPC Genetech Biotechnology Co., Ltd., Shijiazhuang, China
| | - Li Shi
- Basic Research Department, Shanghai Zerun Biotechnology Co., Ltd., Shanghai, China
| | - Hao Zeng
- National Engineering Research Center of Immunological Products, Department of Microbiology and Biochemical Pharmacy, College of Pharmacy and Laboratory Medicine, Third Military Medical University, Chongqing, China
| | - Bingbing Sun
- State Key Laboratory of Fine Chemicals, Dalian University of Technology, Dalian, China
- School of Chemical Engineering, Dalian University of Technology, Dalian, China
| |
Collapse
|
23
|
Gandhi M, Beyrer C, Goosby E. Masks Do More Than Protect Others During COVID-19: Reducing the Inoculum of SARS-CoV-2 to Protect the Wearer. J Gen Intern Med 2020; 35:3063-3066. [PMID: 32737790 PMCID: PMC7393808 DOI: 10.1007/s11606-020-06067-8] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 07/15/2020] [Indexed: 12/17/2022]
Abstract
Although the benefit of population-level public facial masking to protect others during the COVID-19 pandemic has received a great deal of attention, we discuss for one of the first times the hypothesis that universal masking reduces the "inoculum" or dose of the virus for the mask-wearer, leading to more mild and asymptomatic infection manifestations. Masks, depending on type, filter out the majority of viral particles, but not all. We first discuss the near-century-old literature around the viral inoculum and severity of disease (conceptualized as the LD50 or lethal dose of the virus). We include examples of rising rates of asymptomatic infection with population-level masking, including in closed settings (e.g., cruise ships) with and without universal masking. Asymptomatic infections may be harmful for spread but could actually be beneficial if they lead to higher rates of exposure. Exposing society to SARS-CoV-2 without the unacceptable consequences of severe illness with public masking could lead to greater community-level immunity and slower spread as we await a vaccine. This theory of viral inoculum and mild or asymptomatic disease with SARS-CoV-2 in light of population-level masking has received little attention so this is one of the first perspectives to discuss the evidence supporting this theory.
Collapse
Affiliation(s)
- Monica Gandhi
- Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco (UCSF) , San Francisco, CA, USA.
| | - Chris Beyrer
- Desmond M. Tutu Professor of Public Health and Human Rights, Johns Hopkins Bloomberg School of Public Health , Baltimore, MD, USA
| | - Eric Goosby
- Department of Medicine, Division of HIV, Infectious Diseases and Global Medicine, University of California, San Francisco (UCSF) , San Francisco, CA, USA
| |
Collapse
|
24
|
Mittal R, Meneveau C, Wu W. A mathematical framework for estimating risk of airborne transmission of COVID-19 with application to face mask use and social distancing. PHYSICS OF FLUIDS (WOODBURY, N.Y. : 1994) 2020; 32:101903. [PMID: 33100806 PMCID: PMC7583361 DOI: 10.1063/5.0025476] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 09/17/2020] [Indexed: 05/02/2023]
Abstract
A mathematical model for estimating the risk of airborne transmission of a respiratory infection such as COVID-19 is presented. The model employs basic concepts from fluid dynamics and incorporates the known scope of factors involved in the airborne transmission of such diseases. Simplicity in the mathematical form of the model is by design so that it can serve not only as a common basis for scientific inquiry across disciplinary boundaries but it can also be understandable by a broad audience outside science and academia. The caveats and limitations of the model are discussed in detail. The model is used to assess the protection from transmission afforded by face coverings made from a variety of fabrics. The reduction in the transmission risk associated with increased physical distance between the host and susceptible is also quantified by coupling the model with available and new large eddy simulation data on scalar dispersion in canonical flows. Finally, the effect of the level of physical activity (or exercise intensity) of the host and the susceptible in enhancing the transmission risk is also assessed.
Collapse
Affiliation(s)
- Rajat Mittal
- Author to whom correspondence should be addressed:
| | - Charles Meneveau
- Mechanical Engineering, Johns Hopkins
University, 3400 N. Charles St., Baltimore, Maryland 21218,
USA
| | - Wen Wu
- Mechanical Engineering, University of
Mississippi, 209C Carrier Hall, Oxford, Mississippi 38677,
USA
| |
Collapse
|
25
|
Murta V, Villarreal A, Ramos AJ. Severe Acute Respiratory Syndrome Coronavirus 2 Impact on the Central Nervous System: Are Astrocytes and Microglia Main Players or Merely Bystanders? ASN Neuro 2020; 12:1759091420954960. [PMID: 32878468 PMCID: PMC7476346 DOI: 10.1177/1759091420954960] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
With confirmed coronavirus disease 2019 (COVID-19) cases surpassing the 18 million mark around the globe, there is an imperative need to gain comprehensive understanding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Although the main clinical manifestations of COVID-19 are associated with respiratory or intestinal symptoms, reports of neurological signs and symptoms are increasing. The etiology of these neurological manifestations remains obscure, and probably involves several direct pathways, not excluding the direct entry of the virus to the central nervous system (CNS) through the olfactory epithelium, circumventricular organs, or disrupted blood–brain barrier. Furthermore, neuroinflammation might occur in response to the strong systemic cytokine storm described for COVID-19, or due to dysregulation of the CNS rennin-angiotensin system. Descriptions of neurological manifestations in patients in the previous coronavirus (CoV) outbreaks have been numerous for the SARS-CoV and lesser for Middle East respiratory syndrome coronavirus (MERS-CoV). Strong evidence from patients and experimental models suggests that some human variants of CoV have the ability to reach the CNS and that neurons, astrocytes, and/or microglia can be target cells for CoV. A growing body of evidence shows that astrocytes and microglia have a major role in neuroinflammation, responding to local CNS inflammation and/or to disbalanced peripheral inflammation. This is another potential mechanism for SARS-CoV-2 damage to the CNS. In this comprehensive review, we will summarize the known neurological manifestations of SARS-CoV-2, SARS-CoV and MERS-CoV; explore the potential role for astrocytes and microglia in the infection and neuroinflammation; and compare them with the previously described human and animal CoV that showed neurotropism to propose possible underlying mechanisms.
Collapse
Affiliation(s)
- Veronica Murta
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, UBA-CONICET, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Alejandro Villarreal
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, UBA-CONICET, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Alberto J Ramos
- Laboratorio de Neuropatología Molecular, Instituto de Biología Celular y Neurociencia "Prof. E. De Robertis," Facultad de Medicina, UBA-CONICET, Universidad de Buenos Aires, Ciudad de Buenos Aires, Argentina
| |
Collapse
|
26
|
Zhang N, Shang J, Li C, Zhou K, Du L. An overview of Middle East respiratory syndrome coronavirus vaccines in preclinical studies. Expert Rev Vaccines 2020; 19:817-829. [PMID: 32842811 DOI: 10.1080/14760584.2020.1813574] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
INTRODUCTION Middle East respiratory syndrome coronavirus (MERS-CoV) causes high mortality in humans. No vaccines are approved for use in humans; therefore, a consistent effort to develop safe and effective MERS vaccines is needed. AREAS COVERED This review describes the structure of MERS-CoV and the function of its proteins, summarizes MERS vaccine candidates under preclinical study (based on spike and non-spike structural proteins, inactivated virus, and live-attenuated virus), and highlights potential problems that could prevent these vaccines entering clinical trials. It provides guidance for the development of safe and effective MERS-CoV vaccines. EXPERT OPINION Although many MERS-CoV vaccines have been developed, most remain at the preclinical stage. Some vaccines demonstrate immunogenicity and efficacy in animal models, while others have potential adverse effects or low efficacy against high-dose or divergent virus strains. Novel strategies are needed to design safe and effective MERS vaccines to induce broad-spectrum immune responses and improve protective efficacy against multiple strains of MERS-CoV and MERS-like coronaviruses with pandemic potential. More funds should be invested to move vaccine candidates into human clinical trials.
Collapse
Affiliation(s)
- Naru Zhang
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College , Hangzhou, China
| | - Jian Shang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota , Saint Paul, MN, USA
| | - Chaoqun Li
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College , Hangzhou, China
| | - Kehui Zhou
- Department of Clinical Medicine, School of Medicine, Zhejiang University City College , Hangzhou, China
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center , New York, NY, USA
| |
Collapse
|
27
|
Cataldi M, Pignataro G, Taglialatela M. Neurobiology of coronaviruses: Potential relevance for COVID-19. Neurobiol Dis 2020; 143:105007. [PMID: 32622086 PMCID: PMC7329662 DOI: 10.1016/j.nbd.2020.105007] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/25/2020] [Accepted: 06/27/2020] [Indexed: 12/18/2022] Open
Abstract
In the first two decades of the 21st century, there have been three outbreaks of severe respiratory infections caused by highly pathogenic coronaviruses (CoVs) around the world: the severe acute respiratory syndrome (SARS) by the SARS-CoV in 2002-2003, the Middle East respiratory syndrome (MERS) by the MERS-CoV in June 2012, and Coronavirus Disease 2019 (COVID-19) by the SARS-CoV-2 presently affecting most countries In all of these, fatalities are a consequence of a multiorgan dysregulation caused by pulmonary, renal, cardiac, and circulatory damage; however, COVID patients may show significant neurological signs and symptoms such as headache, nausea, vomiting, and sensory disturbances, the most prominent being anosmia and ageusia. The neuroinvasive potential of CoVs might be responsible for at least part of these symptoms and may contribute to the respiratory failure observed in affected patients. Therefore, in the present manuscript, we have reviewed the available preclinical evidence on the mechanisms and consequences of CoVs-induced CNS damage, and highlighted the potential role of CoVs in determining or aggravating acute and long-term neurological diseases in infected individuals. We consider that a widespread awareness of the significant neurotropism of CoVs might contribute to an earlier recognition of the signs and symptoms of viral-induced CNS damage. Moreover, a better understanding of the cellular and molecular mechanisms by which CoVs affect CNS function and cause CNS damage could help in planning new strategies for prognostic evaluation and targeted therapeutic intervention.
Collapse
Affiliation(s)
| | | | - Maurizio Taglialatela
- Division of Pharmacology, Department of Neuroscience, University of Naples "Federico II", 80131 Naples, Italy.
| |
Collapse
|
28
|
Study of an Air Curtain in the Context of Individual Protection from Exposure to Coronavirus (SARS-CoV-2) Contained in Cough-Generated Fluid Particles. PHYSICS 2020. [DOI: 10.3390/physics2030018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The ongoing respiratory COVID-19 pandemic has heavily impacted the social and private lives of the majority of the global population. This infection is primarily transmitted via virus-laden fluid particles (i.e., droplets and aerosols) that are formed in the respiratory tract of infected individuals and expelled from the mouth in the course of breathing, talking, coughing, and sneezing. To mitigate the risk of virus transmission, in many places of the world, the public has been asked or even obliged to use face covers. It is plausible that in the years ahead we will see the use of face masks, face shields and respirators become a normal practice in our life. However, wearing face covers is uncomfortable in some situations, like, for example, in summer heat, while staying on beaches or at hotel swimming pools, doing exercises in gyms, etc. Also, most types of face cover become contaminated with time and need to be periodically replaced or disinfected. These nuisances are caused by the fact that face covers are based on material barriers, which prevent inward and outward propagation of aerosol and droplets containing the pathogen. Here, we study a non-material based protection barrier created by a flow of well directed down stream of air across the front of the open face. The protection is driven by dragging virus-laden particles inside the width of the air flow and hence, as a consequence, displacing them away from their primary trajectories. Applying well established gas-particle flow formalism, we analyzed the dynamics of aerosols and droplets at different regimes of the flow laying over the bodies of the fluid particles. The analysis allowed us to establish the rates of velocity gain of the fluid particles of dimensions relevant for the pathogen transmissions, while they are crossing the width of the air barrier. On the basis of this analysis, we provide a comprehensive study of the protection effectiveness of the air barrier for a susceptible individual located indoor, in an infected environment. Our study shows that such, potentially portable, air curtains can effectively provide both inward and outward protection and serve as an effective personal protective equipment (PPE) mitigating human to human transmission of virus infection like COVID-19.
Collapse
|
29
|
Singh A, Singh RS, Sarma P, Batra G, Joshi R, Kaur H, Sharma AR, Prakash A, Medhi B. A Comprehensive Review of Animal Models for Coronaviruses: SARS-CoV-2, SARS-CoV, and MERS-CoV. Virol Sin 2020; 35:290-304. [PMID: 32607866 PMCID: PMC7324485 DOI: 10.1007/s12250-020-00252-z] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
The recent outbreak of coronavirus disease (COVID-19) caused by the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has already affected a large population of the world. SARS-CoV-2 belongs to the same family of severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV). COVID-19 has a complex pathology involving severe acute respiratory infection, hyper-immune response, and coagulopathy. At present, there is no therapeutic drug or vaccine approved for the disease. There is an urgent need for an ideal animal model that can reflect clinical symptoms and underlying etiopathogenesis similar to COVID-19 patients which can be further used for evaluation of underlying mechanisms, potential vaccines, and therapeutic strategies. The current review provides a paramount insight into the available animal models of SARS-CoV-2, SARS-CoV, and MERS-CoV for the management of the diseases.
Collapse
Affiliation(s)
- Ashutosh Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Rahul Soloman Singh
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Phulen Sarma
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Gitika Batra
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Rupa Joshi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Hardeep Kaur
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Amit Raj Sharma
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Ajay Prakash
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, 160012, India.
| |
Collapse
|
30
|
Hashem AM, Algaissi A, Agrawal AS, Al-Amri SS, Alhabbab RY, Sohrab SS, S Almasoud A, Alharbi NK, Peng BH, Russell M, Li X, Tseng CTK. A Highly Immunogenic, Protective, and Safe Adenovirus-Based Vaccine Expressing Middle East Respiratory Syndrome Coronavirus S1-CD40L Fusion Protein in a Transgenic Human Dipeptidyl Peptidase 4 Mouse Model. J Infect Dis 2020; 220:1558-1567. [PMID: 30911758 PMCID: PMC7107499 DOI: 10.1093/infdis/jiz137] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/21/2019] [Indexed: 12/02/2022] Open
Abstract
Background Infection control measures have played a major role in limiting human/camel-to-human transmission of Middle East respiratory syndrome coronavirus (MERS-CoV); however, development of effective and safe human or camel vaccines is warranted. Methods We extended and optimized our previous recombinant adenovirus 5 (rAd5)–based vaccine platform characterized by in vivo amplified and CD40-mediated specific responses to generate MERS-CoV S1 subunit-based vaccine. We generated rAd5 constructs expressing CD40-targeted S1 fusion protein (rAd5-S1/F/CD40L), untargeted S1 (rAd5-S1), and Green Fluorescent Protein (rAd5-GFP), and evaluated their efficacy and safety in human dipeptidyl peptidase 4 transgenic (hDPP4 Tg+) mice. Results Immunization of hDPP4 Tg+ mice with a single dose of rAd5-S1/F/CD40L elicited as robust and significant specific immunoglobulin G and neutralizing antibodies as those induced with 2 doses of rAd5-S1. After MERS-CoV challenge, both vaccines conferred complete protection against morbidity and mortality, as evidenced by significantly undetectable/reduced pulmonary viral loads compared to the control group. However, rAd5-S1– but not rAd5-S1/F/CD40L–immunized mice exhibited marked pulmonary perivascular hemorrhage post–MERS-CoV challenge despite the observed protection. Conclusions Incorporation of CD40L into rAd5-based MERS-CoV S1 vaccine targeting molecule and molecular adjuvants not only enhances immunogenicity and efficacy but also prevents inadvertent pulmonary pathology after viral challenge, thereby offering a promising strategy to enhance safety and potency of vaccines.
Collapse
Affiliation(s)
- Anwar M Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine.,Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, Saudi Arabia.,Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah Algaissi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston.,Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University
| | | | - Sawsan S Al-Amri
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, Saudi Arabia.,Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rowa Y Alhabbab
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, Saudi Arabia.,Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah
| | - Sayed S Sohrab
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrahman S Almasoud
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Naif Khalaf Alharbi
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Bi-Hung Peng
- Department of Neurosciences, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston
| | - Marsha Russell
- Center for Vaccine Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa, Ontario
| | - Xuguang Li
- Center for Vaccine Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa, Ontario
| | - Chien-Te K Tseng
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston.,Center of Biodefense and Emerging Disease, University of Texas Medical Branch, Galveston
| |
Collapse
|
31
|
Ogier M, Andéol G, Sagui E, Dal Bo G. How to detect and track chronic neurologic sequelae of COVID-19? Use of auditory brainstem responses and neuroimaging for long-term patient follow-up. Brain Behav Immun Health 2020; 5:100081. [PMID: 32427134 PMCID: PMC7227537 DOI: 10.1016/j.bbih.2020.100081] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 02/08/2023] Open
Abstract
This review intends to provide an overview of the current knowledge on neurologic sequelae of COVID-19 and their possible etiology, and, based on available data, proposes possible improvements in current medical care procedures. We conducted a thorough review of the scientific literature on neurologic manifestations of COVID-19, the neuroinvasive propensity of known coronaviruses (CoV) and their possible effects on brain structural and functional integrity. It appears that around one third of COVID-19 patients admitted to intensive care units (ICU) for respiratory difficulties exhibit neurologic symptoms. This may be due to progressive brain damage and dysfunction triggered by severe hypoxia and hypoxemia, heightened inflammation and SARS-CoV-2 dissemination into brain parenchyma, as suggested by current reports and analyses of previous CoV outbreaks. Viral invasion of the brain may particularly target and alter brainstem and thalamic functions and, consequently, result in sensorimotor dysfunctions and psychiatric disorders. Moreover, data collected from other structurally homologous CoV suggest that SARS-CoV-2 infection may lead to brain cell degeneration and demyelination similar to multiple sclerosis (MS). Hence, current evidence warrants further evaluation and long-term follow-up of possible neurologic sequelae in COVID-19 patients. It may be particularly relevant to evaluate brainstem integrity in recovered patients, as it is suspected that this cerebral area may particularly be dysfunctional following SARS-CoV-2 infection. Because CoV infection can potentially lead to chronic neuroinflammation and progressive demyelination, neuroimaging features and signs of MS may also be evaluated in the long term in recovered COVID-19 patients.
Collapse
Affiliation(s)
- Michael Ogier
- French Armed Forces Biomedical Research Institute, 1 place Valérie André, 91220, Brétigny sur Orge, France
| | - Guillaume Andéol
- French Armed Forces Biomedical Research Institute, 1 place Valérie André, 91220, Brétigny sur Orge, France
| | - Emmanuel Sagui
- French Armed Forces Biomedical Research Institute, 1 place Valérie André, 91220, Brétigny sur Orge, France
- European Hospital of Marseille, 6 rue Désirée Clary, 13003, Marseille, France
| | - Gregory Dal Bo
- French Armed Forces Biomedical Research Institute, 1 place Valérie André, 91220, Brétigny sur Orge, France
| |
Collapse
|
32
|
Rodon J, Okba NMA, Te N, van Dieren B, Bosch BJ, Bensaid A, Segalés J, Haagmans BL, Vergara-Alert J. Blocking transmission of Middle East respiratory syndrome coronavirus (MERS-CoV) in llamas by vaccination with a recombinant spike protein. Emerg Microbes Infect 2020; 8:1593-1603. [PMID: 31711379 PMCID: PMC6853226 DOI: 10.1080/22221751.2019.1685912] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The ongoing Middle East respiratory syndrome coronavirus (MERS-CoV) outbreaks pose a worldwide public health threat. Blocking MERS-CoV zoonotic transmission from dromedary camels, the animal reservoir, could potentially reduce the number of primary human cases. Here we report MERS-CoV transmission from experimentally infected llamas to naïve animals. Directly inoculated llamas shed virus for at least 6 days and could infect all in-contact naïve animals 4–5 days after exposure. With the aim to block virus transmission, we examined the efficacy of a recombinant spike S1-protein vaccine. In contrast to naïve animals, in-contact vaccinated llamas did not shed infectious virus upon exposure to directly inoculated llamas, consistent with the induction of strong virus neutralizing antibody responses. Our data provide further evidence that vaccination of the reservoir host may impede MERS-CoV zoonotic transmission to humans.
Collapse
Affiliation(s)
- Jordi Rodon
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Bellaterra (Cerdanyola del Vallès), Spain
| | - Nisreen M A Okba
- Department of Viroscience, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Nigeer Te
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Bellaterra (Cerdanyola del Vallès), Spain
| | - Brenda van Dieren
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Berend-Jan Bosch
- Virology Division, Department of Infectious Diseases & Immunology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Albert Bensaid
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Bellaterra (Cerdanyola del Vallès), Spain
| | - Joaquim Segalés
- UAB, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Bellaterra (Cerdanyola del Vallès), Spain.,Departament de Sanitat i Anatomia Animals, Facultat de Veterinària, UAB, Bellaterra (Cerdanyola del Vallès), Spain
| | - Bart L Haagmans
- Department of Viroscience, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Júlia Vergara-Alert
- IRTA, Centre de Recerca en Sanitat Animal (CReSA, IRTA-UAB), Bellaterra (Cerdanyola del Vallès), Spain
| |
Collapse
|
33
|
Algaissi A, Agrawal AS, Han S, Peng BH, Luo C, Li F, Chan TS, Couch RB, Tseng CTK. Elevated Human Dipeptidyl Peptidase 4 Expression Reduces the Susceptibility of hDPP4 Transgenic Mice to Middle East Respiratory Syndrome Coronavirus Infection and Disease. J Infect Dis 2019; 219:829-835. [PMID: 30256968 PMCID: PMC6376904 DOI: 10.1093/infdis/jiy574] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 09/20/2018] [Indexed: 01/06/2023] Open
Abstract
Background The ongoing Middle East respiratory syndrome coronavirus (MERS-CoV) infections pose threats to public health worldwide, making an understanding of MERS pathogenesis and development of effective medical countermeasures (MCMs) urgent. Methods We used homozygous (+/+) and heterozygous (+/−) human dipeptidyl peptidase 4 (hDPP4) transgenic mice to study the effect of hDPP4 on MERS-CoV infection. Specifically, we determined values of 50% lethal dose (LD50) of MERS-CoV for the 2 strains of mice, compared and correlated their levels of soluble (s)hDPP4 expression to susceptibility, and explored recombinant (r)shDPP4 as an effective MCM for MERS infection. Results hDPP4+/+ mice were unexpectedly more resistant than hDPP4+/− mice to MERS-CoV infection, as judged by increased LD50, reduced lung viral infection, attenuated morbidity and mortality, and reduced histopathology. Additionally, the resistance to MERS-CoV infection directly correlated with increased serum shDPP4 and serum virus neutralizing activity. Finally, administration of rshDPP4 led to reduced lung virus titer and histopathology. Conclusions Our studies suggest that the serum shDPP4 levels play a role in MERS pathogenesis and demonstrate a potential of rshDPP4 as a treatment option for MERS. Additionally, it offers a validated pair of Tg mice strains for characterizing the effect of shDPP4 on MERS pathogenesis.
Collapse
Affiliation(s)
- Abdullah Algaissi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston.,Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Saudi Arabia
| | - Anurodh S Agrawal
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston
| | - Song Han
- Department of Molecular Diagnostics, University of Texas Medical Branch, Galveston
| | - Bi-Hung Peng
- Department of Neurosciences, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston
| | - Chuming Luo
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul
| | - Fang Li
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul
| | - Teh-Sheng Chan
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston
| | - Robert B Couch
- Department of Internal Medicine, Division of Infectious Disease, University of Texas Medical Branch, Galveston
| | - Chien-Te K Tseng
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston.,Center for Biodefense and Emerging Infectious Disease, University of Texas Medical Branch, Galveston
| |
Collapse
|
34
|
Hashem AM, Algaissi A, Agrawal AS, Al-Amri SS, Alhabbab RY, Sohrab SS, S Almasoud A, Alharbi NK, Peng BH, Russell M, Li X, Tseng CTK. A Highly Immunogenic, Protective, and Safe Adenovirus-Based Vaccine Expressing Middle East Respiratory Syndrome Coronavirus S1-CD40L Fusion Protein in a Transgenic Human Dipeptidyl Peptidase 4 Mouse Model. J Infect Dis 2019; 220:1558-1567. [PMID: 30911758 DOI: 10.1093/infdis/jiz13710.1093/infdis/jiz137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Accepted: 03/21/2019] [Indexed: 05/22/2023] Open
Abstract
BACKGROUND Infection control measures have played a major role in limiting human/camel-to-human transmission of Middle East respiratory syndrome coronavirus (MERS-CoV); however, development of effective and safe human or camel vaccines is warranted. METHODS We extended and optimized our previous recombinant adenovirus 5 (rAd5)-based vaccine platform characterized by in vivo amplified and CD40-mediated specific responses to generate MERS-CoV S1 subunit-based vaccine. We generated rAd5 constructs expressing CD40-targeted S1 fusion protein (rAd5-S1/F/CD40L), untargeted S1 (rAd5-S1), and Green Fluorescent Protein (rAd5-GFP), and evaluated their efficacy and safety in human dipeptidyl peptidase 4 transgenic (hDPP4 Tg+) mice. RESULTS Immunization of hDPP4 Tg+ mice with a single dose of rAd5-S1/F/CD40L elicited as robust and significant specific immunoglobulin G and neutralizing antibodies as those induced with 2 doses of rAd5-S1. After MERS-CoV challenge, both vaccines conferred complete protection against morbidity and mortality, as evidenced by significantly undetectable/reduced pulmonary viral loads compared to the control group. However, rAd5-S1- but not rAd5-S1/F/CD40L-immunized mice exhibited marked pulmonary perivascular hemorrhage post-MERS-CoV challenge despite the observed protection. CONCLUSIONS Incorporation of CD40L into rAd5-based MERS-CoV S1 vaccine targeting molecule and molecular adjuvants not only enhances immunogenicity and efficacy but also prevents inadvertent pulmonary pathology after viral challenge, thereby offering a promising strategy to enhance safety and potency of vaccines.
Collapse
MESH Headings
- Adenoviruses, Human/genetics
- Adjuvants, Immunologic/genetics
- Adjuvants, Immunologic/pharmacology
- Animals
- Antibodies, Neutralizing/blood
- Antibodies, Viral/blood
- CD40 Ligand/genetics
- CD40 Ligand/pharmacology
- Coronavirus Infections/immunology
- Coronavirus Infections/prevention & control
- Dipeptidyl Peptidase 4/genetics
- Dipeptidyl Peptidase 4/metabolism
- Drug Carriers
- Genetic Vectors
- Immunoglobulin G/blood
- Lung/virology
- Mice
- Mice, Transgenic
- Middle East Respiratory Syndrome Coronavirus/immunology
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/immunology
- Recombinant Fusion Proteins/pharmacology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
- Survival Analysis
- Vaccines, Subunit/administration & dosage
- Vaccines, Subunit/genetics
- Vaccines, Subunit/immunology
- Vaccines, Synthetic/administration & dosage
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
- Viral Load
- Viral Vaccines/administration & dosage
- Viral Vaccines/genetics
- Viral Vaccines/immunology
Collapse
Affiliation(s)
- Anwar M Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdullah Algaissi
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University
| | | | - Sawsan S Al-Amri
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Rowa Y Alhabbab
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah
| | - Sayed S Sohrab
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Abdulrahman S Almasoud
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Naif Khalaf Alharbi
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Bi-Hung Peng
- Department of Neurosciences, Cell Biology, and Anatomy, University of Texas Medical Branch, Galveston
| | - Marsha Russell
- Center for Vaccine Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa, Ontario
| | - Xuguang Li
- Center for Vaccine Evaluation, Biologics and Genetic Therapies Directorate, Health Canada, Ottawa, Ontario
| | - Chien-Te K Tseng
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston
- Center of Biodefense and Emerging Disease, University of Texas Medical Branch, Galveston
| |
Collapse
|
35
|
Sarkar S, Heise MT. Mouse Models as Resources for Studying Infectious Diseases. Clin Ther 2019; 41:1912-1922. [PMID: 31540729 PMCID: PMC7112552 DOI: 10.1016/j.clinthera.2019.08.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 08/05/2019] [Accepted: 08/09/2019] [Indexed: 12/17/2022]
Abstract
Mouse models are important tools both for studying the pathogenesis of infectious diseases and for the preclinical evaluation of vaccines and therapies against a wide variety of human pathogens. The use of genetically defined inbred mouse strains, humanized mice, and gene knockout mice has allowed the research community to explore how pathogens cause disease, define the role of specific host genes in either controlling or promoting disease, and identify potential targets for the prevention or treatment of a wide range of infectious agents. This review discusses several of the most commonly used mouse model systems, as well as new resources such as the Collaborative Cross as models for studying infectious diseases.
Collapse
Affiliation(s)
- Sanjay Sarkar
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Mark T Heise
- Department of Genetics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
| |
Collapse
|
36
|
Algaissi A, Agrawal AS, Hashem AM, Tseng CTK. Quantification of the Middle East Respiratory Syndrome-Coronavirus RNA in Tissues by Quantitative Real-Time RT-PCR. Methods Mol Biol 2019; 2099:99-106. [PMID: 31883090 PMCID: PMC7122982 DOI: 10.1007/978-1-0716-0211-9_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Since the emergence of the Middle East respiratory syndrome-coronavirus (MERS-CoV) in 2012, more than 2280 confirmed human infections and 800 associated deaths had been reported to the World Health Organization. MERS-CoV is a single-stranded RNA virus that belongs to the Coronaviridae family. MERS-CoV infection leads to a variety of clinical outcomes in humans ranging from asymptomatic and mild infection to severe acute lung injury and multi-organ failure and death. To study the pathogenesis of MERS-CoV infection and development of medical countermeasures (MCMs) for MERS, a number of genetically modified mouse models have been developed, including various versions of transgenic mice expressing the human DPP4 viral receptor. Tracking and quantifying viral infection, among others, in permissive hosts is a key endpoint for studying MERS pathogenesis and evaluating the efficacy of selected MCMs developed for MERS. In addition to quantifying infectious progeny virus which requires high-containment biosafety level (BSL)-3 laboratory, here we outlined an established real-time quantitative RT-PCR (RT-qPCR)-based procedure to unequivocally quantify MERS-CoV-specific RNAs within the lungs of infected human DPP4 (hDPP4, transgenic (hDPP4 Tg) mice under a standard BSL-2 laboratory.
Collapse
Affiliation(s)
- Abdullah Algaissi
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
- Department of Medical Laboratories Technology, College of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Anurodh S Agrawal
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Anwar M Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia.
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.
| | - Chien-Te K Tseng
- Departments of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
- Center for Biodefense and Emerging Infectious Disease, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
37
|
Lin LC, Huang C, Yao B, Lin J, Agrawal A, Algaissi A, Peng B, Liu Y, Huang P, Juang R, Chang Y, Tseng C, Chen H, Hu CJ. Viromimetic STING Agonist-Loaded Hollow Polymeric Nanoparticles for Safe and Effective Vaccination against Middle East Respiratory Syndrome Coronavirus. ADVANCED FUNCTIONAL MATERIALS 2019; 29:1807616. [PMID: 32313544 PMCID: PMC7161765 DOI: 10.1002/adfm.201807616] [Citation(s) in RCA: 108] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/17/2019] [Indexed: 05/04/2023]
Abstract
The continued threat of emerging, highly lethal infectious pathogens such as Middle East respiratory syndrome coronavirus (MERS-CoV) calls for the development of novel vaccine technology that offers safe and effective prophylactic measures. Here, a novel nanoparticle vaccine is developed to deliver subunit viral antigens and STING agonists in a virus-like fashion. STING agonists are first encapsulated into capsid-like hollow polymeric nanoparticles, which show multiple favorable attributes, including a pH-responsive release profile, prominent local immune activation, and reduced systemic reactogenicity. Upon subsequent antigen conjugation, the nanoparticles carry morphological semblance to native virions and facilitate codelivery of antigens and STING agonists to draining lymph nodes and immune cells for immune potentiation. Nanoparticle vaccine effectiveness is supported by the elicitation of potent neutralization antibody and antigen-specific T cell responses in mice immunized with a MERS-CoV nanoparticle vaccine candidate. Using a MERS-CoV-permissive transgenic mouse model, it is shown that mice immunized with this nanoparticle-based MERS-CoV vaccine are protected against a lethal challenge of MERS-CoV without triggering undesirable eosinophilic immunopathology. Together, the biocompatible hollow nanoparticle described herein provides an excellent strategy for delivering both subunit vaccine candidates and novel adjuvants, enabling accelerated development of effective and safe vaccines against emerging viral pathogens.
Collapse
Affiliation(s)
| | - Chen‐Yu Huang
- Department of Veterinary MedicineNational Taiwan UniversityTaipei10617Taiwan
| | - Bing‐Yu Yao
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
| | - Jung‐Chen Lin
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
| | - Anurodh Agrawal
- Department of Microbiology and ImmunologyThe University of Texas Medical BranchGalvestonTX77555USA
| | - Abdullah Algaissi
- Department of Microbiology and ImmunologyThe University of Texas Medical BranchGalvestonTX77555USA
- Department of Medical Laboratories TechnologyJazan UniversityJazan45142Saudi Arabia
| | - Bi‐Hung Peng
- Department of Neurosciences, Cell Biology & AnatomyThe University of Texas Medical BranchGalvestonTX77555USA
| | - Yu‐Han Liu
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
| | - Ping‐Han Huang
- Department of Veterinary MedicineNational Taiwan UniversityTaipei10617Taiwan
| | - Rong‐Huay Juang
- Department of Biochemical Science and TechnologyNational Taiwan UniversityTaipei10617Taiwan
| | - Yuan‐Chih Chang
- Institute of Cellular and Organismic BiologyAcademia SinicaTaipei11529Taiwan
| | - Chien‐Te Tseng
- Department of Microbiology and ImmunologyThe University of Texas Medical BranchGalvestonTX77555USA
- Center for Biodefense and Emerging DiseaseThe University of Texas Medical BranchGalvestonTX77555USA
| | - Hui‐Wen Chen
- Department of Veterinary MedicineNational Taiwan UniversityTaipei10617Taiwan
| | - Che‐Ming Jack Hu
- Institute of Biomedical SciencesAcademia SinicaTaipei11529Taiwan
| |
Collapse
|
38
|
Lin LCW, Huang CY, Yao BY, Lin JC, Agrawal A, Algaissi A, Peng BH, Liu YH, Huang PH, Juang RH, Chang YC, Tseng CT, Chen HW, Hu CMJ. Viromimetic STING Agonist-Loaded Hollow Polymeric Nanoparticles for Safe and Effective Vaccination against Middle East Respiratory Syndrome Coronavirus. ADVANCED FUNCTIONAL MATERIALS 2019; 29:1807616. [PMID: 32313544 DOI: 10.1002/adfm.201807676] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/17/2019] [Indexed: 05/22/2023]
Abstract
The continued threat of emerging, highly lethal infectious pathogens such as Middle East respiratory syndrome coronavirus (MERS-CoV) calls for the development of novel vaccine technology that offers safe and effective prophylactic measures. Here, a novel nanoparticle vaccine is developed to deliver subunit viral antigens and STING agonists in a virus-like fashion. STING agonists are first encapsulated into capsid-like hollow polymeric nanoparticles, which show multiple favorable attributes, including a pH-responsive release profile, prominent local immune activation, and reduced systemic reactogenicity. Upon subsequent antigen conjugation, the nanoparticles carry morphological semblance to native virions and facilitate codelivery of antigens and STING agonists to draining lymph nodes and immune cells for immune potentiation. Nanoparticle vaccine effectiveness is supported by the elicitation of potent neutralization antibody and antigen-specific T cell responses in mice immunized with a MERS-CoV nanoparticle vaccine candidate. Using a MERS-CoV-permissive transgenic mouse model, it is shown that mice immunized with this nanoparticle-based MERS-CoV vaccine are protected against a lethal challenge of MERS-CoV without triggering undesirable eosinophilic immunopathology. Together, the biocompatible hollow nanoparticle described herein provides an excellent strategy for delivering both subunit vaccine candidates and novel adjuvants, enabling accelerated development of effective and safe vaccines against emerging viral pathogens.
Collapse
Affiliation(s)
| | - Chen-Yu Huang
- Department of Veterinary Medicine National Taiwan University Taipei 10617 Taiwan
| | - Bing-Yu Yao
- Institute of Biomedical Sciences Academia Sinica Taipei 11529 Taiwan
| | - Jung-Chen Lin
- Institute of Biomedical Sciences Academia Sinica Taipei 11529 Taiwan
| | - Anurodh Agrawal
- Department of Microbiology and Immunology The University of Texas Medical Branch Galveston TX 77555 USA
| | - Abdullah Algaissi
- Department of Microbiology and Immunology The University of Texas Medical Branch Galveston TX 77555 USA
- Department of Medical Laboratories Technology Jazan University Jazan 45142 Saudi Arabia
| | - Bi-Hung Peng
- Department of Neurosciences, Cell Biology & Anatomy The University of Texas Medical Branch Galveston TX 77555 USA
| | - Yu-Han Liu
- Institute of Biomedical Sciences Academia Sinica Taipei 11529 Taiwan
| | - Ping-Han Huang
- Department of Veterinary Medicine National Taiwan University Taipei 10617 Taiwan
| | - Rong-Huay Juang
- Department of Biochemical Science and Technology National Taiwan University Taipei 10617 Taiwan
| | - Yuan-Chih Chang
- Institute of Cellular and Organismic Biology Academia Sinica Taipei 11529 Taiwan
| | - Chien-Te Tseng
- Department of Microbiology and Immunology The University of Texas Medical Branch Galveston TX 77555 USA
- Center for Biodefense and Emerging Disease The University of Texas Medical Branch Galveston TX 77555 USA
| | - Hui-Wen Chen
- Department of Veterinary Medicine National Taiwan University Taipei 10617 Taiwan
| | - Che-Ming Jack Hu
- Institute of Biomedical Sciences Academia Sinica Taipei 11529 Taiwan
| |
Collapse
|
39
|
Skariyachan S, Challapilli SB, Packirisamy S, Kumargowda ST, Sridhar VS. Recent Aspects on the Pathogenesis Mechanism, Animal Models and Novel Therapeutic Interventions for Middle East Respiratory Syndrome Coronavirus Infections. Front Microbiol 2019; 10:569. [PMID: 30984127 PMCID: PMC6448012 DOI: 10.3389/fmicb.2019.00569] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2018] [Accepted: 03/05/2019] [Indexed: 12/17/2022] Open
Abstract
Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is an emerging zoonotic virus considered as one of the major public threat with a total number of 2 298 laboratory-confirmed cases and 811 associated deaths reported by World Health Organization as of January 2019. The transmission of the virus was expected to be from the camels found in Middle Eastern countries via the animal and human interaction. The genome structure provided information about the pathogenicity and associated virulent factors present in the virus. Recent studies suggested that there were limited insight available on the development of novel therapeutic strategies to induce immunity against the virus. The severities of MERS-CoV infection highlight the necessity of effective approaches for the development of various therapeutic remedies. Thus, the present review comprehensively and critically illustrates the recent aspects on the epidemiology of the virus, the structural and functional features of the viral genome, viral entry and transmission, major mechanisms of pathogenesis and associated virulent factors, current animal models, detection methods and novel strategies for the development of vaccines against MERS-CoV. The review further illustrates the molecular and computational virtual screening platforms which provide insights for the identification of putative drug targets and novel lead molecules toward the development of therapeutic remedies.
Collapse
Affiliation(s)
- Sinosh Skariyachan
- R&D Centre, Department of Biotechnology, Dayananda Sagar College of Engineering, Bengaluru, India
| | | | | | | | | |
Collapse
|
40
|
Schindewolf C, Menachery VD. Middle East Respiratory Syndrome Vaccine Candidates: Cautious Optimism. Viruses 2019; 11:E74. [PMID: 30658390 PMCID: PMC6356267 DOI: 10.3390/v11010074] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/10/2019] [Accepted: 01/12/2019] [Indexed: 12/28/2022] Open
Abstract
Efforts towards developing a vaccine for Middle East respiratory syndrome coronavirus (MERS-CoV) have yielded promising results. Utilizing a variety of platforms, several vaccine approaches have shown efficacy in animal models and begun to enter clinical trials. In this review, we summarize the current progress towards a MERS-CoV vaccine and highlight potential roadblocks identified from previous attempts to generate coronavirus vaccines.
Collapse
Affiliation(s)
- Craig Schindewolf
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555 TX, USA.
| | - Vineet D Menachery
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, 77555 TX, USA.
| |
Collapse
|
41
|
Wang C, Hua C, Xia S, Li W, Lu L, Jiang S. Combining a Fusion Inhibitory Peptide Targeting the MERS-CoV S2 Protein HR1 Domain and a Neutralizing Antibody Specific for the S1 Protein Receptor-Binding Domain (RBD) Showed Potent Synergism against Pseudotyped MERS-CoV with or without Mutations in RBD. Viruses 2019; 11:v11010031. [PMID: 30621343 PMCID: PMC6356712 DOI: 10.3390/v11010031] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Revised: 12/30/2018] [Accepted: 01/02/2019] [Indexed: 01/19/2023] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) has continuously posed a threat to public health worldwide, yet no therapeutics or vaccines are currently available to prevent or treat MERS-CoV infection. We previously identified a fusion inhibitory peptide (HR2P-M2) targeting the MERS-CoV S2 protein HR1 domain and a highly potent neutralizing monoclonal antibody (m336) specific to the S1 spike protein receptor-binding domain (RBD). However, m336 was found to have reduced efficacy against MERS-CoV strains with mutations in RBD, and HR2P-M2 showed low potency, thus limiting the clinical application of each when administered separately. However, we herein report that the combination of m336 and HR2P-M2 exhibited potent synergism in inhibiting MERS-CoV S protein-mediated cell–cell fusion and infection by MERS-CoV pseudoviruses with or without mutations in the RBD, resulting in the enhancement of antiviral activity in contrast to either one administered alone. Thus, this combinatorial strategy could be used in clinics for the urgent treatment of MERS-CoV-infected patients.
Collapse
Affiliation(s)
- Cong Wang
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China.
| | - Chen Hua
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China.
| | - Shuai Xia
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China.
| | - Weihua Li
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai 200032, China.
| | - Lu Lu
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China.
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences and Shanghai Public Health Clinical Center, Fudan University, Shanghai 200032, China.
- NHC Key Laboratory of Reproduction Regulation (Shanghai Institute of Planned Parenthood Research), Fudan University, Shanghai 200032, China.
| |
Collapse
|
42
|
Liang R, Wang L, Zhang N, Deng X, Su M, Su Y, Hu L, He C, Ying T, Jiang S, Yu F. Development of Small-Molecule MERS-CoV Inhibitors. Viruses 2018; 10:v10120721. [PMID: 30562987 PMCID: PMC6316138 DOI: 10.3390/v10120721] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2018] [Revised: 12/11/2018] [Accepted: 12/12/2018] [Indexed: 12/27/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) with potential to cause global pandemics remains a threat to the public health, security, and economy. In this review, we focus on advances in the research and development of small-molecule MERS-CoV inhibitors targeting different stages of the MERS-CoV life cycle, aiming to prevent or treat MERS-CoV infection.
Collapse
Affiliation(s)
- Ruiying Liang
- College of Life and Science, Hebei Agricultural University, Baoding 071001, China.
| | - Lili Wang
- Research Center of Chinese Jujube, Hebei Agricultural University, Baoding 071001, China.
| | - Naru Zhang
- Department of Clinical Medicine, Faculty of Medicine, Zhejiang University City College, Hangzhou 310015, China.
| | - Xiaoqian Deng
- College of Life and Science, Hebei Agricultural University, Baoding 071001, China.
| | - Meng Su
- College of Life and Science, Hebei Agricultural University, Baoding 071001, China.
| | - Yudan Su
- College of Life and Science, Hebei Agricultural University, Baoding 071001, China.
| | - Lanfang Hu
- College of Life and Science, Hebei Agricultural University, Baoding 071001, China.
| | - Chen He
- College of Life and Science, Hebei Agricultural University, Baoding 071001, China.
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China.
| | - Fei Yu
- College of Life and Science, Hebei Agricultural University, Baoding 071001, China.
| |
Collapse
|
43
|
Al-Omari A, Rabaan AA, Salih S, Al-Tawfiq JA, Memish ZA. MERS coronavirus outbreak: Implications for emerging viral infections. Diagn Microbiol Infect Dis 2018; 93:265-285. [PMID: 30413355 PMCID: PMC7127703 DOI: 10.1016/j.diagmicrobio.2018.10.011] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Revised: 09/28/2018] [Accepted: 10/11/2018] [Indexed: 02/08/2023]
Abstract
In September 2012, a novel coronavirus was isolated from a patient who died in Saudi Arabia after presenting with acute respiratory distress and acute kidney injury. Analysis revealed the disease to be due to a novel virus which was named Middle East Respiratory Coronavirus (MERS-CoV). There have been several MERS-CoV hospital outbreaks in KSA, continuing to the present day, and the disease has a mortality rate in excess of 35%. Since 2012, the World Health Organization has been informed of 2220 laboratory-confirmed cases resulting in at least 790 deaths. Cases have since arisen in 27 countries, including an outbreak in the Republic of Korea in 2015 in which 36 people died, but more than 80% of cases have occurred in Saudi Arabia.. Human-to-human transmission of MERS-CoV, particularly in healthcare settings, initially caused a ‘media panic’, however human-to-human transmission appears to require close contact and thus far the virus has not achieved epidemic potential. Zoonotic transmission is of significant importance and evidence is growing implicating the dromedary camel as the major animal host in spread of disease to humans. MERS-CoV is now included on the WHO list of priority blueprint diseases for which there which is an urgent need for accelerated research and development as they have the potential to cause a public health emergency while there is an absence of efficacious drugs and/or vaccines. In this review we highlight epidemiological, clinical, and infection control aspects of MERS-CoV as informed by the Saudi experience. Attention is given to recommended treatments and progress towards vaccine development. 2220 laboratory-confirmed cases of MERS-CoV resulting in at least 790 deaths since 2012 MERS-CoV is on the WHO list of priority blueprint diseases Zoonotic and human-to-human transmission modes need further clarification. No specific therapy has yet been approved. There is a need for well-controlled clinical trials on potential direct therapies.
Collapse
Affiliation(s)
- Awad Al-Omari
- Critical Care and Infection Control Department, Dr. Sulaiman Al-Habib Medical Group, and Al-Faisal University, Riyadh, Saudi Arabia
| | - Ali A Rabaan
- Molecular Diagnostic Laboratory, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia.
| | - Samer Salih
- Internal Medicine Department, Dr.Sulaiman Al-Habib Medical Group, Riyadh, Saudi Arabia
| | - Jaffar A Al-Tawfiq
- Medical Department, Johns Hopkins Aramco Healthcare, Dhahran, Saudi Arabia, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ziad A Memish
- College of Medicine, Al-Faisal University, Riyadh, Saudi Arabia
| |
Collapse
|
44
|
Fan C, Wu X, Liu Q, Li Q, Liu S, Lu J, Yang Y, Cao Y, Huang W, Liang C, Ying T, Jiang S, Wang Y. A Human DPP4-Knockin Mouse's Susceptibility to Infection by Authentic and Pseudotyped MERS-CoV. Viruses 2018; 10:448. [PMID: 30142928 PMCID: PMC6164841 DOI: 10.3390/v10090448] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2018] [Revised: 08/13/2018] [Accepted: 08/17/2018] [Indexed: 01/01/2023] Open
Abstract
Infection by the Middle East respiratory syndrome coronavirus (MERS-CoV) causes respiratory illness and has a high mortality rate (~35%). The requirement for the virus to be manipulated in a biosafety level three (BSL-3) facility has impeded development of urgently-needed antiviral agents. Here, we established anovel mouse model by inserting human dipeptidyl peptidase 4 (hDPP4) into the Rosa26 locus using CRISPR/Cas9, resulting in global expression of the transgene in a genetically stable mouse line. The mice were highly susceptible to infection by MERS-CoV clinical strain hCoV-EMC, which induced severe diffuse pulmonary disease in the animals, and could also be infected by an optimized pseudotyped MERS-CoV. Administration of the neutralizing monoclonal antibodies, H111-1 and m336, as well as a fusion inhibitor peptide, HR2P-M2, protected mice from challenge with authentic and pseudotyped MERS-CoV. These results confirmed that the hDPP4-knockin mouse is a novel model for studies of MERS-CoV pathogenesis and anti-MERS-CoV antiviral agents in BSL-3 and BSL-2facilities, respectively.
Collapse
Affiliation(s)
- Changfa Fan
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Xi Wu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Qiang Liu
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Qianqian Li
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Susu Liu
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Jianjun Lu
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety Evaluation, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China.
| | - Yanwei Yang
- National Center for Safety Evaluation of Drugs, Institute for Food and Drug Safety Evaluation, National Institutes for Food and Drug Control, A8 Hongda Middle Street, Beijing Economic-Technological Development Area, Beijing 100176, China.
| | - Yuan Cao
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Weijin Huang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Chunnan Liang
- Division of Animal Model Research, Institute for Laboratory Animal Resources, National Institutes for Food and Drug Control, Beijing 100050, China.
| | - Tianlei Ying
- Key Laboratory of Medical Molecular Virology of the Ministries of Education and Health, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology of the Ministries of Education and Health, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Youchun Wang
- Division of HIV/AIDS and Sex-Transmitted Virus Vaccines, National Institutes for Food and Drug Control, Beijing 100050, China.
| |
Collapse
|
45
|
Abstract
INTRODUCTION Six years have passed since Middle East respiratory syndrome (MERS) coronavirus (MERS-CoV), a newly emerging infectious virus, was first reported in 2012. Although MERS-CoV has had a consistently high mortality rate in humans, no vaccines have been approved to prevent MERS-CoV infection in humans. MERS-CoV spike (S) protein is a key target for development of MERS vaccines. AREAS COVERED In this review, we illustrate the structure and function of S protein as a vaccine target, describe available animal models for evaluating MERS vaccines, and summarize recent progress on MERS-CoV S-based vaccines, focusing on their ability to elicit antibody and/or cellular immune responses, neutralizing antibodies, and protection against MERS-CoV infection in different models. Prospects for future MERS-CoV S-based vaccines are discussed. EXPERT COMMENTARY The majority of MERS vaccines under development are based on MERS-CoV S protein, including full-length S, S1, and receptor-binding domain (RBD). While it is essential to evaluate the safety of full-length S and S1-based MERS vaccines, further improvement of the efficacy of RBD-based vaccines using novel strategies would be necessary. Overall, this review provides informative guidance for designing and developing safe and effective MERS vaccines based on viral S protein.
Collapse
Affiliation(s)
- Yusen Zhou
- a State Key Laboratory of Pathogen and Biosecurity , Beijing Institute of Microbiology and Epidemiology , Beijing , China
| | - Shibo Jiang
- b Lindsley F. Kimball Research Institute, New York Blood Center , New York , NY , USA.,c Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, Shanghai Medical College and Institute of Medical Microbiology , Fudan University , Shanghai , China
| | - Lanying Du
- b Lindsley F. Kimball Research Institute, New York Blood Center , New York , NY , USA
| |
Collapse
|
46
|
Production of a Monoclonal Antibody Targeting the M Protein of MERS-CoV for Detection of MERS-CoV Using a Synthetic Peptide Epitope Formulated with a CpG-DNA-Liposome Complex. Int J Pept Res Ther 2018; 25:819-826. [PMID: 32214929 PMCID: PMC7087630 DOI: 10.1007/s10989-018-9731-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2018] [Indexed: 01/08/2023]
Abstract
The Middle East respiratory syndrome-related coronavirus (MERS-CoV) contains four major structural proteins, the spike glycoprotein, nucleocapsid phosphoprotein, membrane (M) glycoprotein and small envelope glycoprotein. The M protein of MERS-CoV has a role in the morphogenesis or assembly of the virus and inhibits type I interferon expression in infected cells. Here, we produced a monoclonal antibody specific against the M protein of MERS-CoV by injecting BALB/c mice with a complex containing the epitope peptide and CpG–DNA encapsulated with a phosphatidyl-β-oleoyl-γ-palmitoyl ethanolamine (DOPE):cholesterol hemisuccinate (CHEMS). The monoclonal antibody was reactive to the epitope peptide of the M protein of MERS-CoV which was confirmed by western blotting and immunoprecipitations. Indirect immunofluorescence assay and confocal image analysis showed that the monoclonal antibody binds specifically to the M protein of MERS-CoV in the virus-infected cells. Further studies using this monoclonal antibody may provide important information on the function of the M protein and its future application in diagnostics.
Collapse
|
47
|
Nyon MP, Du L, Tseng CTK, Seid CA, Pollet J, Naceanceno KS, Agrawal A, Algaissi A, Peng BH, Tai W, Jiang S, Bottazzi ME, Strych U, Hotez PJ. Engineering a stable CHO cell line for the expression of a MERS-coronavirus vaccine antigen. Vaccine 2018; 36:1853-1862. [PMID: 29496347 PMCID: PMC5860679 DOI: 10.1016/j.vaccine.2018.02.065] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Revised: 02/12/2018] [Accepted: 02/16/2018] [Indexed: 12/21/2022]
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) has infected at least 2040 patients and caused 712 deaths since its first appearance in 2012, yet neither pathogen-specific therapeutics nor approved vaccines are available. To address this need, we are developing a subunit recombinant protein vaccine comprising residues 377–588 of the MERS-CoV spike protein receptor-binding domain (RBD), which, when formulated with the AddaVax adjuvant, it induces a significant neutralizing antibody response and protection against MERS-CoV challenge in vaccinated animals. To prepare for the manufacture and first-in-human testing of the vaccine, we have developed a process to stably produce the recombinant MERS S377-588 protein in Chinese hamster ovary (CHO) cells. To accomplish this, we transfected an adherent dihydrofolate reductase-deficient CHO cell line (adCHO) with a plasmid encoding S377-588 fused with the human IgG Fc fragment (S377-588-Fc). We then demonstrated the interleukin-2 signal peptide-directed secretion of the recombinant protein into extracellular milieu. Using a gradually increasing methotrexate (MTX) concentration to 5 μM, we increased protein yield by a factor of 40. The adCHO-expressed S377-588-Fc recombinant protein demonstrated functionality and binding specificity identical to those of the protein from transiently transfected HEK293T cells. In addition, hCD26/dipeptidyl peptidase-4 (DPP4) transgenic mice vaccinated with AddaVax-adjuvanted S377-588-Fc could produce neutralizing antibodies against MERS-CoV and survived for at least 21 days after challenge with live MERS-CoV with no evidence of immunological toxicity or eosinophilic immune enhancement. To prepare for large scale-manufacture of the vaccine antigen, we have further developed a high-yield monoclonal suspension CHO cell line.
Collapse
Affiliation(s)
- Mun Peak Nyon
- Tropical Infectious Diseases Research and Education Centre (TIDREC), University of Malaya, Kuala Lumpur, Malaysia; Texas Children's Hospital Center for Vaccine Development, USA; Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Chien-Te Kent Tseng
- Department of Microbiology and Immunology & Center of Biodefense and Emerging Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Christopher A Seid
- Texas Children's Hospital Center for Vaccine Development, USA; Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Jeroen Pollet
- Texas Children's Hospital Center for Vaccine Development, USA; Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Kevin S Naceanceno
- Texas Children's Hospital Center for Vaccine Development, USA; Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Anurodh Agrawal
- Department of Microbiology and Immunology & Center of Biodefense and Emerging Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Abdullah Algaissi
- Department of Microbiology and Immunology & Center of Biodefense and Emerging Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Bi-Hung Peng
- Department of Microbiology and Immunology & Center of Biodefense and Emerging Diseases, University of Texas Medical Branch, Galveston, TX, USA
| | - Wanbo Tai
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY, USA; Key Laboratory of Medical Molecular Virology of MOE/MOH, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Maria Elena Bottazzi
- Texas Children's Hospital Center for Vaccine Development, USA; Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA.
| | - Ulrich Strych
- Texas Children's Hospital Center for Vaccine Development, USA; Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Peter J Hotez
- Texas Children's Hospital Center for Vaccine Development, USA; Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
48
|
Excler JL, Delvecchio CJ, Wiley RE, Williams M, Yoon IK, Modjarrad K, Boujelal M, Moorthy VS, Hersi AS, Kim JH. Toward Developing a Preventive MERS-CoV Vaccine-Report from a Workshop Organized by the Saudi Arabia Ministry of Health and the International Vaccine Institute, Riyadh, Saudi Arabia, November 14-15, 2015. Emerg Infect Dis 2018; 22. [PMID: 27439020 PMCID: PMC4982192 DOI: 10.3201/eid2208.160229] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Middle East respiratory syndrome (MERS) remains a serious international public health threat. With the goal of accelerating the development of countermeasures against MERS coronavirus (MERS-CoV), funding agencies, nongovernmental organizations, and researchers across the world assembled in Riyadh, Saudi Arabia, on November 14-15, 2015, to discuss vaccine development challenges. The meeting was spearheaded by the Saudi Ministry of Health and co-organized by the International Vaccine Institute, South Korea. Accelerating the development of a preventive vaccine requires a better understanding of MERS epidemiology, transmission, and pathogenesis in humans and animals. A combination of rodent and nonhuman primate models should be considered in evaluating and developing preventive and therapeutic vaccine candidates. Dromedary camels should be considered for the development of veterinary vaccines. Several vaccine technology platforms targeting the MERS-CoV spike protein were discussed. Mechanisms to maximize investment, provide robust data, and affect public health are urgently needed.
Collapse
|
49
|
McKimm-Breschkin JL, Jiang S, Hui DS, Beigel JH, Govorkova EA, Lee N. Prevention and treatment of respiratory viral infections: Presentations on antivirals, traditional therapies and host-directed interventions at the 5th ISIRV Antiviral Group conference. Antiviral Res 2018; 149:118-142. [PMID: 29162476 PMCID: PMC7133686 DOI: 10.1016/j.antiviral.2017.11.013] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022]
Abstract
The International Society for Influenza and other Respiratory Virus Diseases held its 5th Antiviral Group (isirv-AVG) Conference in Shanghai, China, in conjunction with the Shanghai Public Health Center and Fudan University from 14-16 June 2017. The three-day programme encompassed presentations on some of the clinical features, management, immune responses and virology of respiratory infections, including influenza A(H1N1)pdm09 and A(H7N9) viruses, MERS-CoV, SARS-CoV, adenovirus Type 80, enterovirus D68, metapneumovirus and respiratory syncytial virus (RSV). Updates were presented on several therapeutics currently in clinical trials, including influenza polymerase inhibitors pimodivir/JNJ6362387, S033188, favipiravir, monoclonal antibodies MHAA45449A and VIS410, and host directed strategies for influenza including nitazoxanide, and polymerase ALS-008112 and fusion inhibitors AK0529, GS-5806 for RSV. Updates were also given on the use of the currently licensed neuraminidase inhibitors. Given the location in China, there were also presentations on the use of Traditional Chinese Medicines. Following on from the previous conference, there were ongoing discussions on appropriate endpoints for severe influenza in clinical trials from regulators and clinicians, an issue which remains unresolved. The aim of this conference summary is to provide information for not only conference participants, but a detailed referenced review of the current status of clinical trials, and pre-clinical development of therapeutics and vaccines for influenza and other respiratory diseases for a broader audience.
Collapse
Affiliation(s)
| | - Shibo Jiang
- College of Basic Medical Sciences, Fudan University, Shanghai, China; Lindsley F. Kimball Research Institute, New York Blood Center, NY, USA
| | - David S Hui
- Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong Special Administrative Region
| | - John H Beigel
- Leidos Biomedical Research, Inc., Support to National Institute of Allergy and Infectious Diseases, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Elena A Govorkova
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, USA
| | - Nelson Lee
- Faculty of Medicine and Dentistry, University of Alberta, Canada
| |
Collapse
|
50
|
Cho H, Excler JL, Kim JH, Yoon IK. Development of Middle East Respiratory Syndrome Coronavirus vaccines - advances and challenges. Hum Vaccin Immunother 2017; 14:304-313. [PMID: 29048984 DOI: 10.1080/21645515.2017.1389362] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is an emerging pathogen with the potential to pose a threat to global public health. Sporadic cases and outbreaks continue to be reported in the Middle East, and case fatality rates remain high at approximately 36% globally. No specific preventive or therapeutic countermeasures currently exist. A safe and effective vaccine could play an important role in protecting against the threat from MERS-CoV. This review discusses human vaccine candidates currently under development, and explores viral characteristics, molecular epidemiology and immunology relevant to MERS-CoV vaccine development. At present, a DNA vaccine candidate has begun a human clinical trial, while two vector-based candidates will very soon begin human trials. Protein-based vaccines are still at pre-clinical stage. Challenges to successful development include incomplete understanding of viral transmission, pathogenesis and immune response (in particular at the mucosal level), no optimal animal challenge models, lack of standardized immunological assays, and insufficient sustainable funding.
Collapse
Affiliation(s)
- Heeyoun Cho
- a Department of Clinical Development and Regulatory , International Vaccine Institute , Seoul , Republic of Korea
| | - Jean-Louis Excler
- a Department of Clinical Development and Regulatory , International Vaccine Institute , Seoul , Republic of Korea
| | - Jerome H Kim
- a Department of Clinical Development and Regulatory , International Vaccine Institute , Seoul , Republic of Korea
| | - In-Kyu Yoon
- a Department of Clinical Development and Regulatory , International Vaccine Institute , Seoul , Republic of Korea
| |
Collapse
|