1
|
Alshehri A, Mir NA, Miled N. Detection of Middle East Respiratory Syndrome Coronavirus-Specific RNA and Anti-MERS-Receptor-Binding Domain Antibodies in Camel Milk from Different Regions of Saudi Arabia. Viral Immunol 2022; 35:673-680. [PMID: 36534466 DOI: 10.1089/vim.2022.0045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Middle East respiratory syndrome coronavirus (MERS-CoV) causes viral pneumonia disease in humans. The close contact with camels and drinking milk may cause MERS-CoV transfer to humans. This study was designed to detect the existence of MERS-CoV in camel milk samples collected from healthy animals of various barns located around Saudi Arabia. Camel milk samples were examined for MERS-CoV RNA by real time-quantitative polymerase chain reaction, also enzyme-linked immunosorbent assay was performed to detect IgG antibodies directed against Middle East respiratory syndrome receptor-binding domain. Among 83 camel milk samples tested, the result showed that seven samples (8.4%) were positive for MERS-CoV RNA, whereas 40.9% of camel milk samples had antibodies directed against MERS-CoV. The findings indicate that some regions (East and South part) are characterized by a high incidence of viral antibodies. The Southwestern region displayed the lowest infection rates. Among the camel breeds, the lowest positivity for detection of MERS-CoV RNA and IgG antibodies was found in Sahilia. This could be related to a higher resistance to viral infection of the breed Sahilia and/or to the geographical origin of the camels sampled in the study. This needs to be more explored to reduce spread of infection and also to understand the underlying reasons. The presence of viral RNA in camel milk samples warrants for measures to prevent possible foodborne transmission of MERS-CoV through milk consumption.
Collapse
Affiliation(s)
- Aiydh Alshehri
- Department of Microbiology, Riyadh Municipality Central Area Labs, Riyadh, Saudi Arabia.,Departmet of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia
| | - Naiman Ali Mir
- Department of Microbiology, Riyadh Municipality Central Area Labs, Riyadh, Saudi Arabia.,Department of Microbiology, Mumtaz Degree and P.G. College, Hyderabad, India
| | - Nabil Miled
- Departmet of Biological Sciences, College of Science, University of Jeddah, Jeddah, Saudi Arabia.,Life Sciences Research Unit, Center for Sciences and Medical Research, The University of Jeddah, Jeddah, Saudi Arabia.,Functional Genomics and Plant Physiology Research Unit, Higher Institute of Biotechnology of Sfax, Sfax University, Sfax, Tunisia
| |
Collapse
|
2
|
Alharbi NK, Aljamaan F, Aljami HA, Alenazi MW, Albalawi H, Almasoud A, Alharthi FJ, Azhar EI, Barhoumi T, Bosaeed M, Gilbert SC, Hashem AM. Immunogenicity of High-Dose MVA-Based MERS Vaccine Candidate in Mice and Camels. Vaccines (Basel) 2022; 10:vaccines10081330. [PMID: 36016218 PMCID: PMC9413082 DOI: 10.3390/vaccines10081330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/16/2022] Open
Abstract
The Middle East respiratory syndrome coronavirus (MERS-CoV) is a zoonotic pathogen that can transmit from dromedary camels to humans, causing severe pneumonia, with a 35% mortality rate. Vaccine candidates have been developed and tested in mice, camels, and humans. Previously, we developed a vaccine based on the modified vaccinia virus Ankara (MVA) viral vector, encoding a full-length spike protein of MERS-CoV, MVA-MERS. Here, we report the immunogenicity of high-dose MVA-MERS in prime–boost vaccinations in mice and camels. Methods: Three groups of mice were immunised with MVA wild-type (MVA-wt) and MVA-MERS (MVA-wt/MVA-MERS), MVA-MERS/MVA-wt, or MVA-MERS/MVA-MERS. Camels were immunised with two doses of PBS, MVA-wt, or MVA-MERS. Antibody (Ab) responses were evaluated using ELISA and MERS pseudovirus neutralisation assays. Results: Two high doses of MVA-MERS induced strong Ab responses in both mice and camels, including neutralising antibodies. Anti-MVA Ab responses did not affect the immune responses to the vaccine antigen (MERS-CoV spike). Conclusions: MVA-MERS vaccine, administered in a homologous prime–boost regimen, induced high levels of neutralising anti-MERS-CoV antibodies in mice and camels. This could be considered for further development and evaluation as a dromedary vaccine to reduce MERS-CoV transmission to humans.
Collapse
Affiliation(s)
- Naif Khalaf Alharbi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
- Correspondence:
| | - Fahad Aljamaan
- Animal Facilities, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Haya A. Aljami
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Mohammed W. Alenazi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Hind Albalawi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Abdulrahman Almasoud
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Fatima J. Alharthi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
| | - Esam I. Azhar
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 22254, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| | - Tlili Barhoumi
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
| | - Mohammad Bosaeed
- Vaccine Development Unit, King Abdullah International Medical Research Center, Riyadh 11481, Saudi Arabia
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh 14611, Saudi Arabia
- Department of Medicine, King Abdulaziz Medical City, Riyadh 12746, Saudi Arabia
| | | | - Anwar M. Hashem
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah 22254, Saudi Arabia
- Vaccines and Immunotherapy Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 22254, Saudi Arabia
| |
Collapse
|
3
|
Faustino R, Faria M, Teixeira M, Palavra F, Sargento P, do Céu Costa M. Systematic review and meta-analysis of the prevalence of coronavirus: One health approach for a global strategy. One Health 2022; 14:100383. [PMID: 35399617 PMCID: PMC8979611 DOI: 10.1016/j.onehlt.2022.100383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/26/2022] [Accepted: 03/27/2022] [Indexed: 01/02/2023] Open
Abstract
Coronaviruses have been responsible for major epidemic crises in 2003 with SARS-CoV-1, in 2012 with MERS-CoV and in 2019 with SARS-CoV-2 (COVID-19), causing serious atypical pneumonia in humans. We intend, with this systematic analysis and meta-analysis, to clarify the prevalence of the various strains of coronavirus in different animal species. For this purpose, we carried out an electronic survey using Pubmed's Veterinary Science search tool to conduct a systematic assessment of published studies reporting the prevalence of different strains of coronavirus in different animal species between 2015 and 2020. We conducted different analysis to assess sensitivity, publication bias, and heterogeneity, using random effect. The final meta-analysis included 42 studies for systematic review and 29 in the meta-analysis. For the geographic regions with a prevalence greater than or equal to 0.20 (Forest plot overall; prevalence = 0.20, p < 0.01, Q = 10,476.22 and I2 = 100%), the most commonly detected viruses were: enteric coronavirus (ECoV), pigeon-dominant coronavirus, (PdCoV), Avian coronavirus M41, Avian coronavirus C46, Avian coronavirus A99, Avian coronavirus JMK, MERS-CoV, Bovine coronavirus, Ro-BatCoV GCCDC1, Alphacoronavirus, Betacoronavirus, Deltacoronavirus, Gamacoronavirus and human coronaviruses (HCoVs). The wide presence of different strains of coronavirus in different animal species on all continents demonstrates the great biodiversity and ubiquity of these viruses. The most recent epidemiological crises caused by coronavirus demonstrates our unpreparedness to anticipate and mitigate emerging risks, as well as the need to implement new epidemiological surveillance programs for viruses. Combined with the need to create advanced training courses in One Health, this is paramount in order to ensure greater effectiveness in fighting the next pandemics.
Collapse
|
4
|
Cheng X, Cao Q, Liao SS. An overview of literature on COVID-19, MERS and SARS: Using text mining and latent Dirichlet allocation. J Inf Sci 2022; 48:304-320. [PMID: 38603038 PMCID: PMC7464068 DOI: 10.1177/0165551520954674] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The unprecedented outbreak of COVID-19 is one of the most serious global threats to public health in this century. During this crisis, specialists in information science could play key roles to support the efforts of scientists in the health and medical community for combatting COVID-19. In this article, we demonstrate that information specialists can support health and medical community by applying text mining technique with latent Dirichlet allocation procedure to perform an overview of a mass of coronavirus literature. This overview presents the generic research themes of the coronavirus diseases: COVID-19, MERS and SARS, reveals the representative literature per main research theme and displays a network visualisation to explore the overlapping, similarity and difference among these themes. The overview can help the health and medical communities to extract useful information and interrelationships from coronavirus-related studies.
Collapse
Affiliation(s)
- Xian Cheng
- Business School, Sichuan University, China
| | - Qiang Cao
- Department of Information Systems, City University of Hong Kong, China
| | | |
Collapse
|
5
|
Woodford J, Sagara I, Dicko A, Zeguime A, Doucoure M, Kwan J, Zaidi I, Doritchamou J, Snow-Smith M, Alani N, Renn J, Kosik I, Holly J, Yewdell J, Esposito D, Sadtler K, Duffy P. Severe Acute Respiratory Syndrome Coronavirus 2 Seroassay Performance and Optimization in a Population With High Background Reactivity in Mali. J Infect Dis 2021; 224:2001-2009. [PMID: 34612499 PMCID: PMC8522418 DOI: 10.1093/infdis/jiab498] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 10/04/2021] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND False positivity may hinder the utility of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) serological tests in sub-Saharan Africa. METHODS From 312 Malian samples collected before 2020, we measured antibodies to the commonly tested SARS-CoV-2 antigens and 4 other betacoronaviruses by enzyme-linked immunosorbent assay (ELISA). In a subset of samples, we assessed antibodies to a panel of Plasmodium falciparum antigens by suspension bead array and functional antiviral activity by SARS-CoV-2 pseudovirus neutralization assay. We then evaluated the performance of an ELISA using SARS-CoV-2 spike protein and receptor-binding domain developed in the United States using Malian positive and negative control samples. To optimize test performance, we compared single- and 2-antigen approaches using existing assay cutoffs and population-specific cutoffs. RESULTS Background reactivity to SARS-CoV-2 antigens was common in prepandemic Malian samples. The SARS-CoV-2 reactivity varied between communities, increased with age, and correlated negligibly/weakly with other betacoronavirus and P falciparum antibodies. No prepandemic samples demonstrated functional activity. Regardless of the cutoffs applied, test specificity improved using a 2-antigen approach. Test performance was optimal using a 2-antigen assay with population-specific cutoffs (sensitivity, 73.9% [95% confidence interval {CI}, 51.6-89.8]; specificity, 99.4% [95% CI, 97.7-99.9]). CONCLUSIONS We have addressed the problem of SARS-CoV-2 seroassay performance in Africa by using a 2-antigen assay with cutoffs defined by performance in the target population.
Collapse
Affiliation(s)
- John Woodford
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Issaka Sagara
- Malaria Research and Training Center/University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Alassane Dicko
- Malaria Research and Training Center/University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Amatigue Zeguime
- Malaria Research and Training Center/University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - M’Bouye Doucoure
- Malaria Research and Training Center/University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Jennifer Kwan
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Irfan Zaidi
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Justin Doritchamou
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Maryonne Snow-Smith
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Nada Alani
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jonathan Renn
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ivan Kosik
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jaroslav Holly
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jonathan Yewdell
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Dominic Esposito
- Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland, USA
| | - Kaitlyn Sadtler
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Patrick Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
6
|
Woodford J, Sagara I, Dicko A, Zeguime A, Doucoure M, Kwan J, Zaidi I, Doritchamou J, Snow-Smith M, Alani N, Renn J, Kosik I, Holly J, Yewdell J, Esposito D, Sadtler K, Duffy P. Severe Acute Respiratory Syndrome Coronavirus 2 Seroassay Performance and Optimization in a Population With High Background Reactivity in Mali. J Infect Dis 2021. [PMID: 34612499 DOI: 10.1101/2021.03.08.21252784v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023] Open
Abstract
BACKGROUND False positivity may hinder the utility of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) serological tests in sub-Saharan Africa. METHODS From 312 Malian samples collected before 2020, we measured antibodies to the commonly tested SARS-CoV-2 antigens and 4 other betacoronaviruses by enzyme-linked immunosorbent assay (ELISA). In a subset of samples, we assessed antibodies to a panel of Plasmodium falciparum antigens by suspension bead array and functional antiviral activity by SARS-CoV-2 pseudovirus neutralization assay. We then evaluated the performance of an ELISA using SARS-CoV-2 spike protein and receptor-binding domain developed in the United States using Malian positive and negative control samples. To optimize test performance, we compared single- and 2-antigen approaches using existing assay cutoffs and population-specific cutoffs. RESULTS Background reactivity to SARS-CoV-2 antigens was common in prepandemic Malian samples. The SARS-CoV-2 reactivity varied between communities, increased with age, and correlated negligibly/weakly with other betacoronavirus and P falciparum antibodies. No prepandemic samples demonstrated functional activity. Regardless of the cutoffs applied, test specificity improved using a 2-antigen approach. Test performance was optimal using a 2-antigen assay with population-specific cutoffs (sensitivity, 73.9% [95% confidence interval {CI}, 51.6-89.8]; specificity, 99.4% [95% CI, 97.7-99.9]). CONCLUSIONS We have addressed the problem of SARS-CoV-2 seroassay performance in Africa by using a 2-antigen assay with cutoffs defined by performance in the target population.
Collapse
Affiliation(s)
- John Woodford
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Issaka Sagara
- Malaria Research and Training Center/University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Alassane Dicko
- Malaria Research and Training Center/University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Amatigue Zeguime
- Malaria Research and Training Center/University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - M'Bouye Doucoure
- Malaria Research and Training Center/University of Science, Techniques, and Technologies of Bamako, Bamako, Mali
| | - Jennifer Kwan
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Irfan Zaidi
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Justin Doritchamou
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Maryonne Snow-Smith
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Nada Alani
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jonathan Renn
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Ivan Kosik
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jaroslav Holly
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Jonathan Yewdell
- Laboratory of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Dominic Esposito
- Frederick National Laboratory for Cancer Research, National Institutes of Health, Frederick, Maryland, USA
| | - Kaitlyn Sadtler
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | - Patrick Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
7
|
Holloway P, Gibson M, van Doremalen N, Nash S, Holloway T, Letko M, Cardwell JM, Al Omari B, Al-Majali A, Abu-Basha E, Mangtani P, Munster VJ, Guitian J. Risk Factors for Middle East Respiratory Syndrome Coronavirus Infection among Camel Populations, Southern Jordan, 2014-2018. Emerg Infect Dis 2021; 27:2301-2311. [PMID: 34423762 PMCID: PMC8386791 DOI: 10.3201/eid2709.203508] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
After the first detection of Middle East respiratory syndrome coronavirus (MERS-CoV) in camels in Jordan in 2013, we conducted 2 consecutive surveys in 2014–2015 and 2017–2018 investigating risk factors for MERS-CoV infection among camel populations in southern Jordan. Multivariate analysis to control for confounding demonstrated that borrowing of camels, particularly males, for breeding purposes was associated with increased MERS-CoV seroprevalence among receiving herds, suggesting a potential route of viral transmission between herds. Increasing age, herd size, and use of water troughs within herds were also associated with increased seroprevalence. Closed herd management practices were found to be protective. Future vaccination strategies among camel populations in Jordan could potentially prioritize breeding males, which are likely to be shared between herds. In addition, targeted management interventions with the potential to reduce transmission between herds should be considered; voluntary closed herd schemes offer a possible route to achieving disease-free herds.
Collapse
|
8
|
Immunotherapeutic Efficacy of IgY Antibodies Targeting the Full-Length Spike Protein in an Animal Model of Middle East Respiratory Syndrome Coronavirus Infection. PHARMACEUTICALS (BASEL, SWITZERLAND) 2021. [PMID: 34073502 DOI: 10.3390/ph14060511.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Identified in 2012, the Middle East respiratory syndrome coronavirus (MERS-CoV) causes severe and often fatal acute respiratory illness in humans. No approved prophylactic or therapeutic interventions are currently available. In this study, we developed chicken egg yolk antibodies (IgY Abs) specific to the MERS-CoV spike (S) protein and evaluated their neutralizing efficiency against MERS-CoV infection. S-specific IgY Abs were produced by injecting chickens with the purified recombinant S protein of MERS-CoV at a high titer (4.4 mg/mL per egg yolk) at week 7 post immunization. Western blotting and immune-dot blot assays demonstrated specific binding to the MERS-CoV S protein. In vitro neutralization of the generated IgY Abs against MERS-CoV was evaluated and showed a 50% neutralizing concentration of 51.42 μg/mL. In vivo testing using a human-transgenic mouse model showed a reduction of viral antigen positive cells in treated mice, compared to the adjuvant-only controls. Moreover, the lung cells of the treated mice showed significantly reduced inflammation, compared to the controls. Our results show efficient neutralization of MERS-CoV infection both in vitro and in vivo using S-specific IgY Abs. Clinical trials are needed to evaluate the efficiency of the IgY Abs in camels and humans.
Collapse
|
9
|
El-Kafrawy SA, Abbas AT, Sohrab SS, Tabll AA, Hassan AM, Iwata-Yoshikawa N, Nagata N, Azhar EI. Immunotherapeutic Efficacy of IgY Antibodies Targeting the Full-Length Spike Protein in an Animal Model of Middle East Respiratory Syndrome Coronavirus Infection. Pharmaceuticals (Basel) 2021; 14:511. [PMID: 34073502 PMCID: PMC8229159 DOI: 10.3390/ph14060511] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 02/07/2023] Open
Abstract
Identified in 2012, the Middle East respiratory syndrome coronavirus (MERS-CoV) causes severe and often fatal acute respiratory illness in humans. No approved prophylactic or therapeutic interventions are currently available. In this study, we developed chicken egg yolk antibodies (IgY Abs) specific to the MERS-CoV spike (S) protein and evaluated their neutralizing efficiency against MERS-CoV infection. S-specific IgY Abs were produced by injecting chickens with the purified recombinant S protein of MERS-CoV at a high titer (4.4 mg/mL per egg yolk) at week 7 post immunization. Western blotting and immune-dot blot assays demonstrated specific binding to the MERS-CoV S protein. In vitro neutralization of the generated IgY Abs against MERS-CoV was evaluated and showed a 50% neutralizing concentration of 51.42 μg/mL. In vivo testing using a human-transgenic mouse model showed a reduction of viral antigen positive cells in treated mice, compared to the adjuvant-only controls. Moreover, the lung cells of the treated mice showed significantly reduced inflammation, compared to the controls. Our results show efficient neutralization of MERS-CoV infection both in vitro and in vivo using S-specific IgY Abs. Clinical trials are needed to evaluate the efficiency of the IgY Abs in camels and humans.
Collapse
Affiliation(s)
- Sherif A. El-Kafrawy
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.)
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Aymn T. Abbas
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.)
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Biotechnology Research Laboratories, Gastroenterology, Surgery Centre, Mansoura University, Mansoura 35516, Egypt
| | - Sayed S. Sohrab
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.)
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ashraf A. Tabll
- Microbial Biotechnology Department, Genetic Engineering and Biotechnology Division, National Research Centre, Dokki 12622, Egypt;
- Department of Immunology, Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo 11517, Egypt
| | - Ahmed M. Hassan
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.)
| | - Naoko Iwata-Yoshikawa
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (N.I.-Y.); (N.N.)
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (N.I.-Y.); (N.N.)
| | - Esam I. Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.)
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
10
|
Lenshin SV, Romashin AV, Vyshemirsky OI, Lvov DK, Alkhovsky SV. [Bats of the subtropical climate zone of the Krasnodar Territory of Russia as a possible reservoir of zoonotic viral infections]. Vopr Virusol 2021; 66:112-122. [PMID: 33993681 DOI: 10.36233/0507-4088-41] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 05/15/2021] [Indexed: 11/05/2022]
Abstract
Emerging and reemerging infections pose a grave global health threat. The emergence of the SARS-CoV-2 virus and the resulting COVID-19 pandemic have demonstrated the importance of studying of zoonotic viruses directly in natural foci. For SARS-like coronaviruses, as well as for many other zoonotic pathogens (including hemorrhagic fevers and rabies agents), the main reservoir are horseshoe bats (Rhinolophus spp.), which are widely distributed in Eurasia and Africa. Their range also covers the southern regions of Russia, including the North Caucasus and Crimea. Large colonies of these animals are located on the territory of Sochi National Park (SNP; subtropical zone of Krasnodar Territory, Greater Sochi region, North Caucasus). In total, according to long-term observations, up to 23 species of bats were registered here, including the great (Rh. ferrumequinum), the lesser (Rh. hipposideros), and the Mediterranean (Rh. euryale) horseshoe bats.This review provides information on zoonotic viruses associated with species of bats distributed in the subtropical zone of Krasnodar Territory of Russia, and analyzes their possible role as a natural reservoir of emerging and reemerging infections. Studying the circulation of zoonotic viruses in bats is an important element of monitoring viral populations in natural foci.
Collapse
Affiliation(s)
- S V Lenshin
- FSBRI «Research Institute of Medical Primatology» of the Ministry of Higher Education and Science of Russia
| | - A V Romashin
- FSBI «Sochi National Park» of the Ministry of Natural Resources and Environment of Russia
| | - O I Vyshemirsky
- FSBRI «Research Institute of Medical Primatology» of the Ministry of Higher Education and Science of Russia
| | - D K Lvov
- FSBI «National Research Centre for Epidemiology and Microbiology named after honorary academician N.F. Gamaleya» of the Ministry of Health of Russia
| | - S V Alkhovsky
- FSBI «National Research Centre for Epidemiology and Microbiology named after honorary academician N.F. Gamaleya» of the Ministry of Health of Russia
| |
Collapse
|
11
|
Sagara I, Woodford J, Dicko A, Zeguime A, Doucoure M, Kwan J, Zaidi I, Doritchamou J, Snow-Smith M, Alani N, Renn J, Kosik I, Holly J, Yewdell J, Esposito D, Sadtler K, Duffy P. SARS-CoV-2 seroassay optimization and performance in a population with high background reactivity in Mali. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.03.08.21252784. [PMID: 33758883 PMCID: PMC7987042 DOI: 10.1101/2021.03.08.21252784] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Serological tests are an indispensable tool to understand the epidemiology of the SARS-CoV-2 pandemic, particularly in areas where molecular diagnostics are limited. Poor assay performance may hinder the utility of these tests, including high rates of false-positivity previously reported in sub-Saharan Africa. From 312 Malian samples collected prior to 2020, we measured antibodies to the commonly tested SARS-CoV-2 antigens and four other betacoronaviruses by ELISA, and assessed functional cross-reactivity in a subset by SARS-CoV-2 pseudovirus neutralization assay. We then evaluated the performance of an ELISA developed in the US, using two-antigen SARS-CoV-2 spike protein and receptor-binding domain. To optimize test performance, we compared single and two-antigen approaches using existing assay cutoffs and population-specific cutoffs for Malian control samples (positive and negative). Background reactivity to SARS-CoV-2 antigens was common in pre-pandemic samples compared to US controls (43.4% (135/311) for spike protein, 22.8% (71/312) for RBD, and 33.9% (79/233) for nucleocapsid protein). SARS-CoV-2 reactivity correlated weakly with other betacoronavirus reactivity, varied between Malian communities, and increased with age. No pre-pandemic samples demonstrated functional activity. Regardless of the cutoffs applied, specificity improved using a two-antigen approach. Test performance was optimal using a two-antigen assay with population-specific cutoffs derived from ROC curve analysis [Sensitivity: 73.9% (51.6-89.8), Specificity: 99.4% (97.7-99.9)]. In the setting of high background reactivity, such as sub-Saharan Africa, SARS-CoV-2 serological assays need careful qualification is to characterize the epidemiology of disease, prevent unnecessary harm, and allocate resources for targeted control measures.
Collapse
|
12
|
Mohapatra RK, Pintilie L, Kandi V, Sarangi AK, Das D, Sahu R, Perekhoda L. The recent challenges of highly contagious COVID-19, causing respiratory infections: Symptoms, diagnosis, transmission, possible vaccines, animal models, and immunotherapy. Chem Biol Drug Des 2020. [PMID: 32654267 DOI: 10.1111/cbdd.v96.510.1111/cbdd.13761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/30/2023]
Abstract
COVID-19 is highly contagious pathogenic viral infection initiated from Wuhan seafood wholesale market of China on December 2019 and spread rapidly around the whole world due to onward transmission. This recent outbreak of novel coronavirus (CoV) was believed to be originated from bats and causing respiratory infections such as common cold, dry cough, fever, headache, dyspnea, pneumonia, and finally Severe Acute Respiratory Syndrome (SARS) in humans. For this widespread zoonotic virus, human-to-human transmission has resulted in nearly 83 lakh cases in 213 countries and territories with 4,50,686 deaths as on 19 June 2020. This review presents a report on the origin, transmission, symptoms, diagnosis, possible vaccines, animal models, and immunotherapy for this novel virus and will provide ample references for the researchers toward the ongoing development of therapeutic agents and vaccines and also preventing the spread of this disease.
Collapse
Affiliation(s)
- Ranjan K Mohapatra
- Department of Chemistry, Government College of Engineering, Keonjhar, Odisha, India
| | - Lucia Pintilie
- Department of Synthesis of Bioactive Substances and Pharmaceutical Technologies, National Institute for Chemical and Pharmaceutical Research and Development, Bucharest, Romania
| | - Venkataramana Kandi
- Department of Microbiology, Pratima Institute of Medical Sciences, Karimnagar, Hyderabad, India
| | - Ashish K Sarangi
- Department of Chemistry, School of Applied Sciences, Centurion University of Technology and Management, Odisha, India
| | - Debadutta Das
- Department of Chemistry, Sukanti Degree College, Subarnapur, Odisha, India
| | - Raghaba Sahu
- College of Pharmacy, Seoul National University, Seoul, South Korea
| | - Lina Perekhoda
- Department of medicinal chemistry, National University of Pharmacy, Kharkiv, Ukraine
| |
Collapse
|
13
|
Mohapatra RK, Pintilie L, Kandi V, Sarangi AK, Das D, Sahu R, Perekhoda L. The recent challenges of highly contagious COVID-19, causing respiratory infections: Symptoms, diagnosis, transmission, possible vaccines, animal models, and immunotherapy. Chem Biol Drug Des 2020; 96:1187-1208. [PMID: 32654267 PMCID: PMC7405220 DOI: 10.1111/cbdd.13761] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/20/2020] [Accepted: 06/28/2020] [Indexed: 01/07/2023]
Abstract
COVID-19 is highly contagious pathogenic viral infection initiated from Wuhan seafood wholesale market of China on December 2019 and spread rapidly around the whole world due to onward transmission. This recent outbreak of novel coronavirus (CoV) was believed to be originated from bats and causing respiratory infections such as common cold, dry cough, fever, headache, dyspnea, pneumonia, and finally Severe Acute Respiratory Syndrome (SARS) in humans. For this widespread zoonotic virus, human-to-human transmission has resulted in nearly 83 lakh cases in 213 countries and territories with 4,50,686 deaths as on 19 June 2020. This review presents a report on the origin, transmission, symptoms, diagnosis, possible vaccines, animal models, and immunotherapy for this novel virus and will provide ample references for the researchers toward the ongoing development of therapeutic agents and vaccines and also preventing the spread of this disease.
Collapse
Affiliation(s)
| | - Lucia Pintilie
- Department of Synthesis of Bioactive Substances and Pharmaceutical TechnologiesNational Institute for Chemical and Pharmaceutical Research and DevelopmentBucharestRomania
| | - Venkataramana Kandi
- Department of MicrobiologyPratima Institute of Medical SciencesKarimnagarHyderabadIndia
| | - Ashish K. Sarangi
- Department of ChemistrySchool of Applied Sciences, Centurion University of Technology and ManagementOdishaIndia
| | - Debadutta Das
- Department of ChemistrySukanti Degree CollegeSubarnapurOdishaIndia
| | - Raghaba Sahu
- College of PharmacySeoul National UniversitySeoulSouth Korea
| | - Lina Perekhoda
- Department of medicinal chemistryNational University of PharmacyKharkivUkraine
| |
Collapse
|
14
|
Abbas AT, El-Kafrawy SA, Sohrab SS, Tabll AA, Hassan AM, Iwata-Yoshikawa N, Nagata N, Azhar EI. Anti-S1 MERS-COV IgY Specific Antibodies Decreases Lung Inflammation and Viral Antigen Positive Cells in the Human Transgenic Mouse Model. Vaccines (Basel) 2020; 8:634. [PMID: 33139631 PMCID: PMC7712919 DOI: 10.3390/vaccines8040634] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/21/2020] [Accepted: 10/28/2020] [Indexed: 02/07/2023] Open
Abstract
The Middle East respiratory syndrome coronavirus (MERS-CoV) was identified in 2012 and causes severe and often fatal acute respiratory illness in humans. No approved prophylactic and therapeutic interventions are currently available. In this study, we have developed egg yolk antibodies (immunoglobulin Y (IgY)) specific for MERS-CoV spike protein (S1) in order to evaluate their neutralizing efficiency against MERS-CoV infection. S1-specific immunoglobulins were produced by injecting chickens with purified recombinant S1 protein of MERS-CoV at a high titer (5.7 mg/mL egg yolk) at week 7 post immunization. Western blotting and immune-dot blot assays demonstrated that the IgY antibody specifically bound to the MERS-CoV S1 protein. Anti-S1 antibodies were also able to recognize MERS-COV inside cells, as demonstrated by an immunofluorescence assay. Plaque reduction and microneutralization assays showed the neutralization of MERS-COV in Vero cells by anti-S1 IgY antibodies and non-significantly reduced virus titers in the lungs of MERS-CoV-infected mice during early infection, with a nonsignificant decrease in weight loss. However, a statistically significant (p = 0.0196) quantitative reduction in viral antigen expression and marked reduction in inflammation were observed in lung tissue. Collectively, our data suggest that the anti-MERS-CoV S1 IgY could serve as a potential candidate for the passive treatment of MERS-CoV infection.
Collapse
Affiliation(s)
- Aymn T. Abbas
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.); (E.I.A.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Biotechnology Research Laboratories, Gastroenterology, Surgery Centre, Mansoura University, Mansoura 35511, Egypt
| | - Sherif A. El-Kafrawy
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.); (E.I.A.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Department of Clinical Pathology, National Liver Institute, Menoufiya University, Shebin El-Kom 32511, Egypt
| | - Sayed Sartaj Sohrab
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.); (E.I.A.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Ashraf A. Tabll
- Genetic Engineering and Biotechnology Division, Microbial Biotechnology Department (Biomedical Technology Group), National Research Centre, Dokki 12622, Egypt;
- Department of Immunology, Egypt Center for Research and Regenerative Medicine (ECRRM), Cairo 11517, Egypt
| | - Ahmed M. Hassan
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.); (E.I.A.)
| | - Naoko Iwata-Yoshikawa
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (N.I.-Y.); (N.N.)
| | - Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo 208-0011, Japan; (N.I.-Y.); (N.N.)
| | - Esam I. Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (S.A.E.-K.); (S.S.S.); (A.M.H.); (E.I.A.)
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| |
Collapse
|
15
|
Anti-S1 MERS-COV IgY Specific Antibodies Decreases Lung Inflammation and Viral Antigen Positive Cells in the Human Transgenic Mouse Model. Vaccines (Basel) 2020. [PMID: 33139631 DOI: 10.3390/vaccines8040634.] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The Middle East respiratory syndrome coronavirus (MERS-CoV) was identified in 2012 and causes severe and often fatal acute respiratory illness in humans. No approved prophylactic and therapeutic interventions are currently available. In this study, we have developed egg yolk antibodies (immunoglobulin Y (IgY)) specific for MERS-CoV spike protein (S1) in order to evaluate their neutralizing efficiency against MERS-CoV infection. S1-specific immunoglobulins were produced by injecting chickens with purified recombinant S1 protein of MERS-CoV at a high titer (5.7 mg/mL egg yolk) at week 7 post immunization. Western blotting and immune-dot blot assays demonstrated that the IgY antibody specifically bound to the MERS-CoV S1 protein. Anti-S1 antibodies were also able to recognize MERS-COV inside cells, as demonstrated by an immunofluorescence assay. Plaque reduction and microneutralization assays showed the neutralization of MERS-COV in Vero cells by anti-S1 IgY antibodies and non-significantly reduced virus titers in the lungs of MERS-CoV-infected mice during early infection, with a nonsignificant decrease in weight loss. However, a statistically significant (p = 0.0196) quantitative reduction in viral antigen expression and marked reduction in inflammation were observed in lung tissue. Collectively, our data suggest that the anti-MERS-CoV S1 IgY could serve as a potential candidate for the passive treatment of MERS-CoV infection.
Collapse
|
16
|
Tinto B, Salinas S, Dicko A, Kagone TS, Traore I, de Rekeneire N, Bicaba BW, Hien H, Meda N, van de Perre P, Kania D, Simonin Y. Spreading of SARS-CoV-2 in West Africa and assessment of risk factors. Epidemiol Infect 2020; 148:e213. [PMID: 32921332 PMCID: PMC7506176 DOI: 10.1017/s0950268820002149] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/21/2020] [Accepted: 08/28/2020] [Indexed: 01/08/2023] Open
Abstract
Although the African continent is, for the moment, less impacted than the rest of the world, it still faces the risk of a spread of COVID-19. In this study, we have conducted a systematic review of the information available in the literature in order to provide an overview of the epidemiological and clinical features of COVID-19 pandemic in West Africa and of the impact of risk factors such as comorbidities, climatic conditions and demography on the pandemic. Burkina Faso is used as a case study to better describe the situation in West Africa. The epidemiological situation of COVID-19 in West Africa is marked by a continuous increase in the numbers of confirmed cases. This geographic area had on 29 July 2020, 131 049 confirmed cases by polymerase chain reaction, 88 305 recoveries and 2102 deaths. Several factors may influence the SARS-CoV-2 circulation in Africa: (i) comorbidities: diabetes mellitus and high blood pressure could lead to an increase in the number of severe cases of SARS-CoV-2; (ii) climatic factors: the high temperatures could be a factor contributing to slow the spread of the virus and (iii) demography: the West Africa population is very young and this could be a factor limiting the occurrence of severe forms of SARS-CoV-2 infection. Although the spread of the SARS-CoV-2 epidemic in West Africa is relatively slow compared to European countries, vigilance must remain. Difficulties in access to diagnostic tests, lack of hospital equipment, but also the large number of people working in the informal sector (such as trading, businesses, transport and restoration) makes it difficult to apply preventive measures, namely physical distancing and containment.
Collapse
Affiliation(s)
- B. Tinto
- Laboratoire National de Référence des Fièvres Hémorragiques Virales, Centre MURAZ, Institut National de Santé Publique (INSP), Bobo-Dioulasso, Burkina Faso
| | - S. Salinas
- Pathogenesis and Control of Chronic Infections, Université de Montpellier, INSERM, EFS, Montpellier, France
| | - A. Dicko
- Laboratoire Central de Référence, INSP, Ouagadougou, Burkina Faso
| | - T. S. Kagone
- Laboratoire National de Référence des Fièvres Hémorragiques Virales, Centre MURAZ, Institut National de Santé Publique (INSP), Bobo-Dioulasso, Burkina Faso
| | - I. Traore
- Laboratoire National de Référence des Fièvres Hémorragiques Virales, Centre MURAZ, Institut National de Santé Publique (INSP), Bobo-Dioulasso, Burkina Faso
| | - N. de Rekeneire
- Centre Muraz, INSP, Bobo-Dioulasso, Burkina Faso
- Expertise France, Paris, France
| | - B. W. Bicaba
- Centre des Opérations de Réponse aux Urgences Sanitaires (CORUS), INSP, Ouagadougou, Burkina Faso
| | - H. Hien
- INSP, Ouagadougou, Burkina Faso
| | - N. Meda
- UFR/SDS, Université Ouaga I Professeur Joseph KI-ZERBO, Ouagadougou, Burkina Faso
| | - P. van de Perre
- Pathogenesis and Control of Chronic Infections, Université de Montpellier, INSERM, EFS, Montpellier, France
| | - D. Kania
- Laboratoire National de Référence des Fièvres Hémorragiques Virales, Centre MURAZ, Institut National de Santé Publique (INSP), Bobo-Dioulasso, Burkina Faso
| | - Y. Simonin
- Pathogenesis and Control of Chronic Infections, Université de Montpellier, INSERM, EFS, Montpellier, France
| |
Collapse
|
17
|
Gautam A, Kaphle K, Shrestha B, Phuyal S. Susceptibility to SARS, MERS, and COVID-19 from animal health perspective. Open Vet J 2020; 10:164-177. [PMID: 32821661 PMCID: PMC7419072 DOI: 10.4314/ovj.v10i2.6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 05/01/2020] [Indexed: 12/11/2022] Open
Abstract
Viruses are having great time as they seem to have bogged humans down. Severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), and novel coronavirus (COVID-19) are the three major coronaviruses of present-day global human and animal health concern. COVID-19 caused by SARS-CoV-2 is identified as the newest disease, presumably of bat origin. Different theories on the evolution of viruses are in circulation, yet there is no denying the fact that the animal source is the skeleton. The whole world is witnessing the terror of the COVID-19 pandemic that is following the same path of SARS and MERS, and seems to be more severe. In addition to humans, several species of animals are reported to have been infected with these life-threatening viruses. The possible routes of transmission and their zoonotic potentialities are the subjects of intense research. This review article aims to overview the link of all these three deadly coronaviruses among animals along with their phylogenic evolution and cross-species transmission. This is essential since animals as pets or food are said to pose some risk, and their better understanding is a must in order to prepare a possible plan for future havoc in both human and animal health. Although COVID-19 is causing a human health hazard globally, its reporting in animals are limited compared to SARS and MERS. Non-human primates and carnivores are most susceptible to SARS-coronavirus and SARS-CoV-2, respectively, whereas the dromedary camel is susceptible to MERS-coronavirus. Phylogenetically, the trio viruses are reported to have originated from bats and have special capacity to undergo mutation and genomic recombination in order to infect humans through its reservoir or replication host. However, it is difficult to analyze how the genomic pattern of coronaviruses occurs. Thus, increased possibility of new virus-variants infecting humans and animals in the upcoming days seems to be the biggest challenge for the future of the world. One health approach is portrayed as our best way ahead, and understanding the animal dimension will go a long way in formulating such preparedness plans.
Collapse
Affiliation(s)
- Aasish Gautam
- Institute of Agriculture and Animal Science, Tribhuvan University, Rupandehi, Nepal
| | - Krishna Kaphle
- Veterinary Teaching Hospital, Institute of Agriculture and Animal Science, Tribhuvan University, Rupandehi, Nepal
| | - Birendra Shrestha
- Institute of Agriculture and Animal Science, Tribhuvan University, Rupandehi, Nepal
| | - Samiksha Phuyal
- Institute of Agriculture and Animal Science, Tribhuvan University, Rupandehi, Nepal
| |
Collapse
|
18
|
Mostafa A, Kandeil A, Shehata M, El Shesheny R, Samy AM, Kayali G, Ali MA. Middle East Respiratory Syndrome Coronavirus (MERS-CoV): State of the Science. Microorganisms 2020; 8:E991. [PMID: 32630780 PMCID: PMC7409282 DOI: 10.3390/microorganisms8070991] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 06/29/2020] [Accepted: 06/30/2020] [Indexed: 02/07/2023] Open
Abstract
Coronaviruses belong to a large family of viruses that can cause disease outbreaks ranging from the common cold to acute respiratory syndrome. Since 2003, three zoonotic members of this family evolved to cross species barriers infecting humans and resulting in relatively high case fatality rates (CFR). Compared to Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV, CFR = 10%) and pandemic Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2, CFR = 6%), the Middle East Respiratory Syndrome Coronavirus (MERS-CoV) has scored the highest CFR (approximately 35%). In this review, we systematically summarize the current state of scientific knowledge about MERS-CoV, including virology and origin, epidemiology, zoonotic mode of transmission, and potential therapeutic or prophylactic intervention modalities.
Collapse
Affiliation(s)
- Ahmed Mostafa
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo 12622, Egypt; (A.M.); (A.K.); (M.S.); (R.E.S.)
| | - Ahmed Kandeil
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo 12622, Egypt; (A.M.); (A.K.); (M.S.); (R.E.S.)
| | - Mahmoud Shehata
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo 12622, Egypt; (A.M.); (A.K.); (M.S.); (R.E.S.)
| | - Rabeh El Shesheny
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo 12622, Egypt; (A.M.); (A.K.); (M.S.); (R.E.S.)
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Abdallah M. Samy
- Entomology Department, Faculty of Science, Ain Shams University, Abbassia, Cairo 11566, Egypt;
| | - Ghazi Kayali
- Department of Epidemiology, Human Genetics, and Environmental Sciences, University of Texas, Houston, TX 77030, USA
- Human Link, Baabda 1109, Lebanon
| | - Mohamed A. Ali
- Center of Scientific Excellence for Influenza Viruses, Environmental Research Division, National Research Centre (NRC), Cairo 12622, Egypt; (A.M.); (A.K.); (M.S.); (R.E.S.)
| |
Collapse
|
19
|
Alharbi NK, Ibrahim OH, Alhafufi A, Kasem S, Aldowerij A, Albrahim R, Abu-Obaidah A, Alkarar A, Bayoumi FA, Almansour AM, Aldubaib M, Al-Abdely HM, Balkhy HH, Qasim I. Challenge infection model for MERS-CoV based on naturally infected camels. Virol J 2020; 17:77. [PMID: 32552831 PMCID: PMC7298446 DOI: 10.1186/s12985-020-01347-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Accepted: 06/02/2020] [Indexed: 02/07/2023] Open
Abstract
Background Middle East Respiratory Syndrome coronavirus (MERS-CoV) is an emerging virus that infects humans and camels with no approved antiviral therapy or vaccine. Some vaccines are in development for camels as a one-health intervention where vaccinating camels is proposed to reduce human viral exposure. This intervention will require an understanding of the prior exposure of camels to the virus and appropriate vaccine efficacy studies in camels. Methods We conducted a cross sectional seroprevalence study in young dromedary camels to determine the rate of MERS-CoV seropositivity in young camels. Next, we utilised naturally infected camels as a natural challenge model that can be used by co-housing these camels with healthy naive camels in a ratio of 1 to 2. This model is aimed to support studies on natural virus transmission as well as evaluating drug and vaccine efficacy. Results We found that 90% of the screened camels have pre-existing antibodies for MERS-CoV. In addition, the challenge model resulted in MERS-CoV transmission within 48 h with infections that continued for 14 days post challenge. Conclusions Our finding suggests that the majority of young dromedary camels in Saudi Arabia are seropositive and that naturally infected camels can serve as a challenge model to assess transmission, therapeutics, and vaccine efficacy.
Collapse
Affiliation(s)
- Naif Khalaf Alharbi
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia. .,King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| | - Osman H Ibrahim
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Ali Alhafufi
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Samy Kasem
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia.,Department of Virology, Faculty of Veterinary Medicine, Kafrelsheikh University, El Geish Street, Kafrelsheikh, 33516, Egypt
| | - Ali Aldowerij
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Raed Albrahim
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Ali Abu-Obaidah
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Ali Alkarar
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | | | | | - Musaad Aldubaib
- College of Agriculture and Veterinary Medicine, Qassim University, Qassim, Saudi Arabia
| | - Hail M Al-Abdely
- Ministry of Health, Riyadh, Saudi Arabia.,Internal Medicine Department, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Hanan H Balkhy
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.,King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.,Department of Infection Prevention and Control, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
| | - Ibrahim Qasim
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| |
Collapse
|
20
|
Global status of Middle East respiratory syndrome coronavirus in dromedary camels: a systematic review. Epidemiol Infect 2020; 147:e84. [PMID: 30869000 PMCID: PMC6518605 DOI: 10.1017/s095026881800345x] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Dromedary camels have been shown to be the main reservoir for human Middle East respiratory syndrome (MERS) infections. This systematic review aims to compile and analyse all published data on MERS-coronavirus (CoV) in the global camel population to provide an overview of current knowledge on the distribution, spread and risk factors of infections in dromedary camels. We included original research articles containing laboratory evidence of MERS-CoV infections in dromedary camels in the field from 2013 to April 2018. In general, camels only show minor clinical signs of disease after being infected with MERS-CoV. Serological evidence of MERS-CoV in camels has been found in 20 countries, with molecular evidence for virus circulation in 13 countries. The seroprevalence of MERS-CoV antibodies increases with age in camels, while the prevalence of viral shedding as determined by MERS-CoV RNA detection in nasal swabs decreases. In several studies, camels that were sampled at animal markets or quarantine facilities were seropositive more often than camels at farms as well as imported camels vs. locally bred camels. Some studies show a relatively higher seroprevalence and viral detection during the cooler winter months. Knowledge of the animal reservoir of MERS-CoV is essential to develop intervention and control measures to prevent human infections.
Collapse
|
21
|
Ramshaw RE, Letourneau ID, Hong AY, Hon J, Morgan JD, Osborne JCP, Shirude S, Van Kerkhove MD, Hay SI, Pigott DM. A database of geopositioned Middle East Respiratory Syndrome Coronavirus occurrences. Sci Data 2019; 6:318. [PMID: 31836720 PMCID: PMC6911100 DOI: 10.1038/s41597-019-0330-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 11/15/2019] [Indexed: 12/21/2022] Open
Abstract
As a World Health Organization Research and Development Blueprint priority pathogen, there is a need to better understand the geographic distribution of Middle East Respiratory Syndrome Coronavirus (MERS-CoV) and its potential to infect mammals and humans. This database documents cases of MERS-CoV globally, with specific attention paid to zoonotic transmission. An initial literature search was conducted in PubMed, Web of Science, and Scopus; after screening articles according to the inclusion/exclusion criteria, a total of 208 sources were selected for extraction and geo-positioning. Each MERS-CoV occurrence was assigned one of the following classifications based upon published contextual information: index, unspecified, secondary, mammal, environmental, or imported. In total, this database is comprised of 861 unique geo-positioned MERS-CoV occurrences. The purpose of this article is to share a collated MERS-CoV database and extraction protocol that can be utilized in future mapping efforts for both MERS-CoV and other infectious diseases. More broadly, it may also provide useful data for the development of targeted MERS-CoV surveillance, which would prove invaluable in preventing future zoonotic spillover. Measurement(s) | Middle East Respiratory Syndrome • geographic location | Technology Type(s) | digital curation | Factor Type(s) | geographic distribution of Middle East Respiratory Syndrome Coronavirus (MERS-CoV) • year | Sample Characteristic - Organism | Middle East respiratory syndrome-related coronavirus | Sample Characteristic - Location | Earth (planet) |
Machine-accessible metadata file describing the reported data: 10.6084/m9.figshare.11108801
Collapse
Affiliation(s)
- Rebecca E Ramshaw
- Institute for Health Metrics and Evaluation, University of Washington, 2301 5th Ave., Suite 600, Seattle, WA, United States
| | - Ian D Letourneau
- Institute for Health Metrics and Evaluation, University of Washington, 2301 5th Ave., Suite 600, Seattle, WA, United States
| | - Amy Y Hong
- Bloomberg School of Public Health, Johns Hopkins University, 615N Wolfe St, Baltimore, MD, 21205, United States
| | - Julia Hon
- Institute for Health Metrics and Evaluation, University of Washington, 2301 5th Ave., Suite 600, Seattle, WA, United States
| | - Julia D Morgan
- Institute for Health Metrics and Evaluation, University of Washington, 2301 5th Ave., Suite 600, Seattle, WA, United States
| | - Joshua C P Osborne
- Institute for Health Metrics and Evaluation, University of Washington, 2301 5th Ave., Suite 600, Seattle, WA, United States
| | - Shreya Shirude
- Institute for Health Metrics and Evaluation, University of Washington, 2301 5th Ave., Suite 600, Seattle, WA, United States
| | - Maria D Van Kerkhove
- Department of Infectious Hazards Management, Health Emergencies Programme, World Health Organization, Avenue Appia 20, 1211, Geneva, Switzerland
| | - Simon I Hay
- Institute for Health Metrics and Evaluation, University of Washington, 2301 5th Ave., Suite 600, Seattle, WA, United States.,Department of Health Metrics Sciences, School of Medicine, University of Washington, 2301 5th Ave., Suite 600, Seattle, WA, United States
| | - David M Pigott
- Institute for Health Metrics and Evaluation, University of Washington, 2301 5th Ave., Suite 600, Seattle, WA, United States. .,Department of Health Metrics Sciences, School of Medicine, University of Washington, 2301 5th Ave., Suite 600, Seattle, WA, United States.
| |
Collapse
|
22
|
Alharbi NK, Qasim I, Almasoud A, Aljami HA, Alenazi MW, Alhafufi A, Aldibasi OS, Hashem AM, Kasem S, Albrahim R, Aldubaib M, Almansour A, Temperton NJ, Kupke A, Becker S, Abu-Obaidah A, Alkarar A, Yoon IK, Azhar E, Lambe T, Bayoumi F, Aldowerij A, Ibrahim OH, Gilbert SC, Balkhy HH. Humoral Immunogenicity and Efficacy of a Single Dose of ChAdOx1 MERS Vaccine Candidate in Dromedary Camels. Sci Rep 2019; 9:16292. [PMID: 31705137 PMCID: PMC6841732 DOI: 10.1038/s41598-019-52730-4] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/16/2019] [Indexed: 11/08/2022] Open
Abstract
MERS-CoV seronegative and seropositive camels received a single intramuscular dose of ChAdOx1 MERS, a replication-deficient adenoviral vectored vaccine expressing MERS-CoV spike protein, with further groups receiving control vaccinations. Infectious camels with active naturally acquired MERS-CoV infection, were co-housed with the vaccinated camels at a ratio of 1:2 (infected:vaccinated); nasal discharge and virus titres were monitored for 14 days. Overall, the vaccination reduced virus shedding and nasal discharge (p = 0.0059 and p = 0.0274, respectively). Antibody responses in seropositive camels were enhancedby the vaccine; these camels had a higher average age than seronegative. Older seronegative camels responded more strongly to vaccination than younger animals; and neutralising antibodies were detected in nasal swabs. Further work is required to optimise vaccine regimens for younger seronegative camels.
Collapse
Affiliation(s)
- Naif Khalaf Alharbi
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia.
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia.
| | - Ibrahim Qasim
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Abdulrahman Almasoud
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haya A Aljami
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Mohamed W Alenazi
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Ali Alhafufi
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Omar S Aldibasi
- Department of Bioinformatics and Biostatistics, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - 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
| | - Samy Kasem
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
- Department of Virology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, El Geish Street, 33516, Egypt
| | - Raed Albrahim
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Musaad Aldubaib
- College of Agriculture and Veterinary Medicine, Qassim University, Qassim, Saudi Arabia
| | - Ali Almansour
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Nigel J Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, Kent, ME4 4TB, United Kingdom
| | - Alexandra Kupke
- Institute of Virology, Philipps University of Marburg, Marburg, Germany
- German Center for Infection Research (DZIF), Partner Site Gieβen-Marburg-Langen, Germany
| | - Stephan Becker
- Institute of Virology, Philipps University of Marburg, Marburg, Germany
- German Center for Infection Research (DZIF), Partner Site Gieβen-Marburg-Langen, Germany
| | - Ali Abu-Obaidah
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Ali Alkarar
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - In-Kyu Yoon
- International Vaccine Institute, Seoul, South Korea
| | - Esam Azhar
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Teresa Lambe
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Faisal Bayoumi
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Ali Aldowerij
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Osman H Ibrahim
- Ministry of Environment, Water and Agriculture (MEWA), Riyadh, Saudi Arabia
| | - Sarah C Gilbert
- The Jenner Institute, University of Oxford, Oxford, OX3 7DQ, UK
| | - Hanan H Balkhy
- Department of Infectious Disease Research, King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
- King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- Department of Infection Prevention and Control, Ministry of National Guard - Health Affairs, Riyadh, Saudi Arabia
| |
Collapse
|
23
|
Detection of MERS-CoV antigen on formalin-fixed paraffin-embedded nasal tissue of alpacas by immunohistochemistry using human monoclonal antibodies directed against different epitopes of the spike protein. Vet Immunol Immunopathol 2019; 218:109939. [PMID: 31526954 PMCID: PMC7112921 DOI: 10.1016/j.vetimm.2019.109939] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Revised: 09/02/2019] [Accepted: 09/08/2019] [Indexed: 01/27/2023]
Abstract
Middle East respiratory syndrome (MERS) represents an important respiratory disease accompanied by lethal outcome in one third of human patients. In recent years, several investigators developed protective antibodies which could be used as prophylaxis in prospective human epidemics. In the current study, eight human monoclonal antibodies (mAbs) with neutralizing and non-neutralizing capabilities, directed against different epitopes of the MERS-coronavirus (MERS-CoV) spike (MERS-S) protein, were investigated with regard to their ability to immunohistochemically detect respective epitopes on formalin-fixed paraffin-embedded (FFPE) nasal tissue sections of MERS-CoV experimentally infected alpacas. The most intense immunoreaction was detected using a neutralizing antibody directed against the receptor binding domain S1B of the MERS-S protein, which produced an immunosignal in the cytoplasm of ciliated respiratory epithelium and along the apical membranous region. A similar staining was obtained by two other mAbs which recognize the sialic acid-binding domain and the ectodomain of the membrane fusion subunit S2, respectively. Five mAbs lacked immunoreactivity for MERS-CoV antigen on FFPE tissue, even though they belong, at least in part, to the same epitope group. In summary, three tested human mAbs demonstrated capacity for detection of MERS-CoV antigen on FFPE samples and may be implemented in double or triple immunohistochemical methods.
Collapse
|
24
|
Some One Health based control strategies for the Middle East respiratory syndrome coronavirus. One Health 2019; 8:100102. [PMID: 31485476 PMCID: PMC6715958 DOI: 10.1016/j.onehlt.2019.100102] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 08/19/2019] [Accepted: 08/19/2019] [Indexed: 02/08/2023] Open
Abstract
The Middle East respiratory syndrome coronavirus (MERS-CoV) presents an ideal example for developing One Health concepts. Dromedary camels are the principal reservoir for the virus. Infected camels shed the virus in body secretions, particularly nasal discharges. MERS-CoV has the potential to remain active in the environment for some time under optimum conditions of temperature and humidity. This shedding sustains the virus in endemic communities and thus contact with camels is considered a major risk factor for human infection. Reducing virus shedding from camels will have a great positive impact on reducing the human risk of infection. Our main objective is to highlight the potential aspects of reducing virus shedding from camels to the environment, thereby reducing the possibility of human infection. We will focus on the potential roles of camel markets, camel shows, importation, transportation and grazing in the amplification and shedding of the virus, providing some novel concepts for the control approaches for the MERS-CoV.
Collapse
|
25
|
Dighe A, Jombart T, Van Kerkhove MD, Ferguson N. A systematic review of MERS-CoV seroprevalence and RNA prevalence in dromedary camels: Implications for animal vaccination. Epidemics 2019; 29:100350. [PMID: 31201040 PMCID: PMC6899506 DOI: 10.1016/j.epidem.2019.100350] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/29/2019] [Accepted: 06/03/2019] [Indexed: 12/17/2022] Open
Abstract
Most adult dromedaries in Africa and the Middle East have been infected with MERS-CoV. Seroprevalence increases with age, while active infection is more common in calves. Prevalence is higher at sites where different dromedary populations mix. Further study is needed to determine if prevalence of infection varies seasonally.
Human infection with Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is driven by recurring dromedary-to-human spill-over events, leading decision-makers to consider dromedary vaccination. Dromedary vaccine candidates in the development pipeline are showing hopeful results, but gaps in our understanding of the epidemiology of MERS-CoV in dromedaries must be addressed to design and evaluate potential vaccination strategies. We aim to bring together existing measures of MERS-CoV infection in dromedary camels to assess the distribution of infection, highlighting knowledge gaps and implications for animal vaccination. We systematically reviewed the published literature on MEDLINE, EMBASE and Web of Science that reported seroprevalence and/or prevalence of active MERS-CoV infection in dromedary camels from both cross-sectional and longitudinal studies. 60 studies met our eligibility criteria. Qualitative syntheses determined that MERS-CoV seroprevalence increased with age up to 80–100% in adult dromedaries supporting geographically widespread endemicity of MERS-CoV in dromedaries in both the Arabian Peninsula and countries exporting dromedaries from Africa. The high prevalence of active infection measured in juveniles and at sites where dromedary populations mix should guide further investigation – particularly of dromedary movement – and inform vaccination strategy design and evaluation through mathematical modelling.
Collapse
Affiliation(s)
- Amy Dighe
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Medical School Building, St Mary's Hospital, Norfolk Place, London, W2 1PG, United Kingdom.
| | - Thibaut Jombart
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Medical School Building, St Mary's Hospital, Norfolk Place, London, W2 1PG, United Kingdom; Department of Infectious Disease Epidemiology, London School of Hygiene & Tropical Medicine, Keppel St, Bloomsbury, London, WC1E 7HT, United Kingdom; UK Public Health Rapid Support Team, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, United Kingdom.
| | - Maria D Van Kerkhove
- Department of Global Infectious Hazards Management, Health Emergencies Program, World Health Organization, Avenue Appia 20, CH-1211, Geneva, Switzerland.
| | - Neil Ferguson
- MRC Centre for Global Infectious Disease Analysis, Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, Medical School Building, St Mary's Hospital, Norfolk Place, London, W2 1PG, United Kingdom.
| |
Collapse
|
26
|
Interferon Regulatory Factor 3-Mediated Signaling Limits Middle-East Respiratory Syndrome (MERS) Coronavirus Propagation in Cells from an Insectivorous Bat. Viruses 2019; 11:v11020152. [PMID: 30781790 PMCID: PMC6410008 DOI: 10.3390/v11020152] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Revised: 02/08/2019] [Accepted: 02/11/2019] [Indexed: 12/14/2022] Open
Abstract
Insectivorous bats are speculated to be ancestral hosts of Middle-East respiratory syndrome (MERS) coronavirus (CoV). MERS-CoV causes disease in humans with thirty-five percent fatality, and has evolved proteins that counteract human antiviral responses. Since bats experimentally infected with MERS-CoV do not develop signs of disease, we tested the hypothesis that MERS-CoV would replicate less efficiently in bat cells than in human cells because of its inability to subvert antiviral responses in bat cells. We infected human and bat (Eptesicus fuscus) cells with MERS-CoV and observed that the virus grew to higher titers in human cells. MERS-CoV also effectively suppressed the antiviral interferon beta (IFNβ) response in human cells, unlike in bat cells. To determine if IRF3, a critical mediator of the interferon response, also regulated the response in bats, we examined the response of IRF3 to poly(I:C), a synthetic analogue of viral double-stranded RNA. We observed that bat IRF3 responded to poly(I:C) by nuclear translocation and post-translational modifications, hallmarks of IRF3 activation. Suppression of IRF3 by small-interfering RNA (siRNA) demonstrated that IRF3 was critical for poly(I:C) and MERS-CoV induced induction of IFNβ in bat cells. Our study demonstrates that innate antiviral signaling in E. fuscus bat cells is resistant to MERS-CoV-mediated subversion.
Collapse
|
27
|
Kelly-Cirino C, Mazzola LT, Chua A, Oxenford CJ, Van Kerkhove MD. An updated roadmap for MERS-CoV research and product development: focus on diagnostics. BMJ Glob Health 2019; 4:e001105. [PMID: 30815285 PMCID: PMC6361340 DOI: 10.1136/bmjgh-2018-001105] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Revised: 10/13/2018] [Accepted: 10/23/2018] [Indexed: 01/12/2023] Open
Abstract
Diagnostics play a central role in the early detection and control of outbreaks and can enable a more nuanced understanding of the disease kinetics and risk factors for the Middle East respiratory syndrome-coronavirus (MERS-CoV), one of the high-priority pathogens identified by the WHO. In this review we identified sources for molecular and serological diagnostic tests used in MERS-CoV detection, case management and outbreak investigations, as well as surveillance for humans and animals (camels), and summarised the performance of currently available tests, diagnostic needs, and associated challenges for diagnostic test development and implementation. A more detailed understanding of the kinetics of infection of MERS-CoV is needed in order to optimise the use of existing assays. Notably, MERS-CoV point-of-care tests are needed in order to optimise supportive care and to minimise transmission risk. However, for new test development, sourcing clinical material continues to be a major challenge to achieving assay validation. Harmonisation and standardisation of laboratory methods are essential for surveillance and for a rapid and effective international response to emerging diseases. Routine external quality assessment, along with well-characterised and up-to-date proficiency panels, would provide insight into MERS-CoV diagnostic performance worldwide. A defined set of Target Product Profiles for diagnostic technologies will be developed by WHO to address these gaps in MERS-CoV outbreak management.
Collapse
Affiliation(s)
| | | | - Arlene Chua
- Department of Information, Evidence and Research, WHO, Geneva, Switzerland.,Medecins Sans Frontières, Geneva, Switzerland
| | | | | |
Collapse
|
28
|
Zhou Y, Yang Y, Huang J, Jiang S, Du L. Advances in MERS-CoV Vaccines and Therapeutics Based on the Receptor-Binding Domain. Viruses 2019; 11:v11010060. [PMID: 30646569 PMCID: PMC6357101 DOI: 10.3390/v11010060] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/28/2022] Open
Abstract
Middle East respiratory syndrome (MERS) coronavirus (MERS-CoV) is an infectious virus that was first reported in 2012. The MERS-CoV genome encodes four major structural proteins, among which the spike (S) protein has a key role in viral infection and pathogenesis. The receptor-binding domain (RBD) of the S protein contains a critical neutralizing domain and is an important target for development of MERS vaccines and therapeutics. In this review, we describe the relevant features of the MERS-CoV S-protein RBD, summarize recent advances in the development of MERS-CoV RBD-based vaccines and therapeutic antibodies, and illustrate potential challenges and strategies to further improve their efficacy.
Collapse
Affiliation(s)
- Yusen Zhou
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing 100071, China.
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China.
| | - Yang Yang
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06520, USA.
| | - Jingwei Huang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA.
| | - Shibo Jiang
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA.
| | - Lanying Du
- Lindsley F. Kimball Research Institute, New York Blood Center, New York, NY 10065, USA.
| |
Collapse
|
29
|
Bats and Coronaviruses. Viruses 2019; 11:v11010041. [PMID: 30634396 PMCID: PMC6356540 DOI: 10.3390/v11010041] [Citation(s) in RCA: 283] [Impact Index Per Article: 47.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 01/05/2019] [Accepted: 01/07/2019] [Indexed: 11/17/2022] Open
Abstract
Bats are speculated to be reservoirs of several emerging viruses including coronaviruses (CoVs) that cause serious disease in humans and agricultural animals. These include CoVs that cause severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS), porcine epidemic diarrhea (PED) and severe acute diarrhea syndrome (SADS). Bats that are naturally infected or experimentally infected do not demonstrate clinical signs of disease. These observations have allowed researchers to speculate that bats are the likely reservoirs or ancestral hosts for several CoVs. In this review, we follow the CoV outbreaks that are speculated to have originated in bats. We review studies that have allowed researchers to identify unique adaptation in bats that may allow them to harbor CoVs without severe disease. We speculate about future studies that are critical to identify how bats can harbor multiple strains of CoVs and factors that enable these viruses to “jump” from bats to other mammals. We hope that this review will enable readers to identify gaps in knowledge that currently exist and initiate a dialogue amongst bat researchers to share resources to overcome present limitations.
Collapse
|
30
|
Ommeh S, Zhang W, Zohaib A, Chen J, Zhang H, Hu B, Ge XY, Yang XL, Masika M, Obanda V, Luo Y, Li S, Waruhiu C, Li B, Zhu Y, Ouma D, Odendo V, Wang LF, Anderson DE, Lichoti J, Mungube E, Gakuya F, Zhou P, Ngeiywa KJ, Yan B, Agwanda B, Shi ZL. Genetic Evidence of Middle East Respiratory Syndrome Coronavirus (MERS-Cov) and Widespread Seroprevalence among Camels in Kenya. Virol Sin 2018; 33:484-492. [PMID: 30570714 PMCID: PMC6335226 DOI: 10.1007/s12250-018-0076-4] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2018] [Accepted: 11/27/2018] [Indexed: 11/25/2022] Open
Abstract
We describe the first genome isolation of Middle East respiratory syndrome coronavirus (MERS-CoV) in Kenya. This fatal zoonotic pathogen was first described in the Kingdom of Saudi Arabia in 2012. Epidemiological and molecular evidence revealed zoonotic transmission from camels to humans and between humans. Currently, MERS-CoV is classified by the WHO as having high pandemic potential requiring greater surveillance. Previous studies of MERS-CoV in Kenya mainly focused on site-specific and archived camel and human serum samples for antibodies. We conducted active nationwide cross-sectional surveillance of camels and humans in Kenya, targeting both nasal swabs and plasma samples from 1,163 camels and 486 humans collected from January 2016 to June 2018. A total of 792 camel plasma samples were positive by ELISA. Seroprevalence increased with age, and the highest prevalence was observed in adult camels (82.37%, 95% confidence interval (CI) 79.50–84.91). More female camels were significantly seropositive (74.28%, 95% CI 71.14–77.19) than male camels (P < 0.001) (53.74%, 95% CI 48.48–58.90). Only 11 camel nasal swabs were positive for MERS-CoV by reverse transcription-quantitative PCR. Phylogenetic analysis of whole genome sequences showed that Kenyan MERS-CoV clustered within sub-clade C2, which is associated with the African clade, but did not contain signature deletions of orf4b in African viruses. None of the human plasma screened contained neutralizing antibodies against MERS-CoV. This study confirms the geographically widespread occurrence of MERS-CoV in Kenyan camels. Further one-health surveillance approaches in camels, wildlife, and human populations are needed.
Collapse
Affiliation(s)
- Sheila Ommeh
- Institute for Biotechnology Research, Jomo Kenyatta University of Agriculture and Technology, Nairobi, 62000-00200, Kenya
| | - Wei Zhang
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ali Zohaib
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Jing Chen
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Huajun Zhang
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Ben Hu
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xing-Yi Ge
- College of Biology, Hunan University, Changsha, 410006, China
| | - Xing-Lou Yang
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Moses Masika
- Department of Medical Microbiology, University of Nairobi, Nairobi, 30197-00100, Kenya
| | - Vincent Obanda
- Veterinary Services Department, Kenya Wildlife Service, Nairobi, 40241-00100, Kenya
| | - Yun Luo
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Shan Li
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Cecilia Waruhiu
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Bei Li
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Yan Zhu
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Desterio Ouma
- Veterinary Research Institute, Kenya Agriculture and Livestock Research Organization, Nairobi, 57811-00200, Kenya
| | - Vincent Odendo
- Veterinary Research Institute, Kenya Agriculture and Livestock Research Organization, Nairobi, 57811-00200, Kenya
| | - Lin-Fa Wang
- Veterinary Services Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Danielle E Anderson
- Veterinary Services Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore, 169857, Singapore
| | - Jacqueline Lichoti
- Directorate of Veterinary Services, State Department of Livestock, Ministry of Agriculture, Livestock Fisheries and Irrigation, Nairobi, 34188-00100, Kenya
| | - Erick Mungube
- Veterinary Research Institute, Kenya Agriculture and Livestock Research Organization, Nairobi, 57811-00200, Kenya
| | - Francis Gakuya
- Veterinary Services Department, Kenya Wildlife Service, Nairobi, 40241-00100, Kenya
| | - Peng Zhou
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Kisa-Juma Ngeiywa
- Directorate of Veterinary Services, State Department of Livestock, Ministry of Agriculture, Livestock Fisheries and Irrigation, Nairobi, 34188-00100, Kenya.,Kenya Camel Association, Nairobi, 30095-00100, Kenya
| | - Bing Yan
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Bernard Agwanda
- Department of Zoology, National Museums of Kenya, Nairobi, 40658-00100, Kenya.
| | - Zheng-Li Shi
- CAS Key Laboratory for Special Pathogens and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430071, China.
| |
Collapse
|
31
|
MERS: Progress on the global response, remaining challenges and the way forward. Antiviral Res 2018; 159:35-44. [PMID: 30236531 PMCID: PMC7113883 DOI: 10.1016/j.antiviral.2018.09.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 09/04/2018] [Indexed: 01/04/2023]
Abstract
This article summarizes progress in research on Middle East Respiratory Syndrome (MERS) since a FAO-OIE-WHO Global Technical Meeting held at WHO Headquarters in Geneva on 25-27 September 2017. The meeting reviewed the latest scientific findings and identified and prioritized the global activities necessary to prevent, manage and control the disease. Critical needs for research and technical guidance identified during the meeting have been used to update the WHO R&D MERS-CoV Roadmap for diagnostics, therapeutics and vaccines and a broader public health research agenda. Since the 2017 meeting, progress has been made on several key actions in animal populations, at the animal/human interface and in human populations. This report also summarizes the latest scientific studies on MERS since 2017, including data from more than 50 research studies examining the presence of MERS-CoV infection in dromedary camels.
Collapse
|
32
|
Countrywide Survey for MERS-Coronavirus Antibodies in Dromedaries and Humans in Pakistan. Virol Sin 2018; 33:410-417. [PMID: 30311100 PMCID: PMC6235758 DOI: 10.1007/s12250-018-0051-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Accepted: 09/03/2018] [Indexed: 12/11/2022] Open
Abstract
Middle East Respiratory Syndrome Coronavirus (MERS-CoV) is a zoonotic pathogen capable of causing severe respiratory disease in humans. Although dromedary camels are considered as a major reservoir host, the MERS-CoV infection dynamics in camels are not fully understood. Through surveillance in Pakistan, nasal (n = 776) and serum (n = 1050) samples were collected from camels between November 2015 and February 2018. Samples were collected from animal markets, free-roaming herds and abattoirs. An in-house ELISA was developed to detect IgG against MERS-CoV. A total of 794 camels were found seropositive for MERS-CoV. Prevalence increased with the age and the highest seroprevalence was recorded in camels aged > 10 years (81.37%) followed by those aged 3.1-10 years (78.65%) and ≤ 3 years (58.19%). Higher prevalence was observed in female (78.13%) as compared to male (70.70%). Of the camel nasal swabs, 22 were found to be positive by RT-qPCR though with high Ct values. Moreover, 2,409 human serum samples were also collected from four provinces of Pakistan during 2016-2017. Among the sampled population, 840 humans were camel herders. Although we found a high rate of MERS-CoV antibody positive dromedaries (75.62%) in Pakistan, no neutralizing antibodies were detected in humans with and without contact to camels.
Collapse
|
33
|
Conzade R, Grant R, Malik MR, Elkholy A, Elhakim M, Samhouri D, Ben Embarek PK, Van Kerkhove MD. Reported Direct and Indirect Contact with Dromedary Camels among Laboratory-Confirmed MERS-CoV Cases. Viruses 2018; 10:v10080425. [PMID: 30104551 PMCID: PMC6115845 DOI: 10.3390/v10080425] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 07/30/2018] [Accepted: 08/09/2018] [Indexed: 12/16/2022] Open
Abstract
Dromedary camels (Camelus dromedarius) are now known to be the vertebrate animal reservoir that intermittently transmits the Middle East respiratory syndrome coronavirus (MERS-CoV) to humans. Yet, details as to the specific mechanism(s) of zoonotic transmission from dromedaries to humans remain unclear. The aim of this study was to describe direct and indirect contact with dromedaries among all cases, and then separately for primary, non-primary, and unclassified cases of laboratory-confirmed MERS-CoV reported to the World Health Organization (WHO) between 1 January 2015 and 13 April 2018. We present any reported dromedary contact: direct, indirect, and type of indirect contact. Of all 1125 laboratory-confirmed MERS-CoV cases reported to WHO during the time period, there were 348 (30.9%) primary cases, 455 (40.4%) non-primary cases, and 322 (28.6%) unclassified cases. Among primary cases, 191 (54.9%) reported contact with dromedaries: 164 (47.1%) reported direct contact, 155 (44.5%) reported indirect contact. Five (1.1%) non-primary cases also reported contact with dromedaries. Overall, unpasteurized milk was the most frequent type of dromedary product consumed. Among cases for whom exposure was systematically collected and reported to WHO, contact with dromedaries or dromedary products has played an important role in zoonotic transmission.
Collapse
Affiliation(s)
- Romy Conzade
- Department of Infectious Hazard Management, Health Emergencies Programme, World Health Organization, 1202 Geneva, Switzerland.
- Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Institute of Epidemiology, D-85764 Neuherberg, Germany.
| | - Rebecca Grant
- Department of Infectious Hazard Management, Health Emergencies Programme, World Health Organization, 1202 Geneva, Switzerland.
- Institut Pasteur, Centre for Global Health Research and Education, 75015 Paris, France.
| | - Mamunur Rahman Malik
- Department of Infectious Hazard Management, Health Emergencies Programme, World Health Organization Regional Office for the Eastern Mediterranean, 11371 Cairo, Egypt.
| | - Amgad Elkholy
- Department of Infectious Hazard Management, Health Emergencies Programme, World Health Organization Regional Office for the Eastern Mediterranean, 11371 Cairo, Egypt.
| | - Mohamed Elhakim
- Department of Infectious Hazard Management, Health Emergencies Programme, World Health Organization Regional Office for the Eastern Mediterranean, 11371 Cairo, Egypt.
| | - Dalia Samhouri
- Department of Country Preparedness and International Health Regulations, World Health Organization Regional Office for the Eastern Mediterranean, 11371 Cairo, Egypt.
| | - Peter K Ben Embarek
- Department of Food Safety and Zoonoses, World Health Organization, 1201 Geneva, Switzerland.
| | - Maria D Van Kerkhove
- Department of Infectious Hazard Management, Health Emergencies Programme, World Health Organization, 1202 Geneva, Switzerland.
| |
Collapse
|
34
|
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
|
35
|
Experimental infection of dromedaries with Middle East respiratory syndrome-Coronavirus is accompanied by massive ciliary loss and depletion of the cell surface receptor dipeptidyl peptidase 4. Sci Rep 2018; 8:9778. [PMID: 29950581 PMCID: PMC6021449 DOI: 10.1038/s41598-018-28109-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 06/15/2018] [Indexed: 12/13/2022] Open
Abstract
Middle East respiratory syndrome (MERS) represents an important respiratory disease accompanied by lethal outcome in one-third of human patients. Recent data indicate that dromedaries represent an important source of infection, although information regarding viral cell tropism and pathogenesis is sparse. In the current study, tissues of eight dromedaries receiving inoculation of MERS-Coronavirus (MERS-CoV) after recombinant Modified-Vaccinia-Virus-Ankara (MVA-S)-vaccination (n = 4), MVA-vaccination (mock vaccination, n = 2) and PBS application (mock vaccination, n = 2), respectively, were investigated. Tissues were analyzed by histology, immunohistochemistry, immunofluorescence, and scanning electron microscopy. MERS-CoV infection in mock-vaccinated dromedaries revealed high numbers of MERS-CoV-nucleocapsid positive cells, T cells, and macrophages within nasal turbinates and trachea at day four post infection. Double immunolabeling demonstrated cytokeratin (CK) 18 expressing epithelial cells to be the prevailing target cell of MERS-CoV, while CK5/6 and CK14 expressing cells did not co-localize with virus. In addition, virus was occasionally detected in macrophages. The acute disease was further accompanied by ciliary loss along with a lack of dipeptidyl peptidase 4 (DPP4), known to mediate virus entry. DPP4 was mainly expressed by human lymphocytes and dromedary monocytes, but overall the expression level was lower in dromedaries. The present study underlines significant species-specific manifestations of MERS and highlights ciliary loss as an important finding in dromedaries. The obtained results promote a better understanding of coronavirus infections, which pose major health challenges.
Collapse
|
36
|
Kasem S, Qasim I, Al-Hufofi A, Hashim O, Alkarar A, Abu-Obeida A, Gaafer A, Elfadil A, Zaki A, Al-Romaihi A, Babekr N, El-Harby N, Hussien R, Al-Sahaf A, Al-Doweriej A, Bayoumi F, Poon LLM, Chu DKW, Peiris M, Perera RAPM. Cross-sectional study of MERS-CoV-specific RNA and antibodies in animals that have had contact with MERS patients in Saudi Arabia. J Infect Public Health 2017; 11:331-338. [PMID: 28993171 PMCID: PMC7102853 DOI: 10.1016/j.jiph.2017.09.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 08/22/2017] [Accepted: 09/09/2017] [Indexed: 12/20/2022] Open
Abstract
Background Middle East respiratory syndrome coronavirus (MERS-CoV) is a newly emerged coronavirus that is associated with a severe respiratory disease in humans in the Middle East. The epidemiological profiles of the MERS-CoV infections suggest zoonotic transmission from an animal reservoir to humans. Methods This study was designed to investigate animal herds associated with Middle East respiratory syndrome (MERS)-infected patients in Saudi Arabia, during the last three years (2014–2016). Nasal swabs and serum samples from 584 dromedary camels, 39 sheep, 51 goats, and 2 cattle were collected. Nasal samples from camels, sheep, goats, and cattle were examined by real-time reverse-transcription PCR (RT-PCR) to detect MERS-CoV RNA, and the Anti-MERS ELISA assay was performed to detect camel humeral immune response (IgG) to MERS-CoV S1 antigen infection. The complete genome sequencing of ten MERS-CoV camel isolates and phylogenetic analysis was performed. Results The data indicated that seventy-five dromedary camels were positive for MERS-CoV RNA; the virus was not detected in sheep, goats, and cattle. MERS-CoV RNA from infected camels was not detected beyond 2 weeks after the first positive result was detected in nasal swabs obtained from infected camels. Anti-MERS ELISA assays showed that 70.9% of camels related to human cases had antibodies to MERS-CoV. The full genome sequences of the ten MERS-CoV camel isolates were identical to their corresponding patients and were grouped together within the larger MERS-CoV sequences cluster for human and camel isolates reported form the Arabian Peninsula. Conclusions These findings indicate that camels are a significant reservoir for the maintenance of MERS-CoVs, and they are an important source of human infection with MERS.
Collapse
Affiliation(s)
- Samy Kasem
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia; Department of Virology, Faculty of Veterinary Medicine, Kafrelsheikh University, El Geish Street, Kafrelsheikh 33516, Egypt.
| | - Ibraheem Qasim
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Ali Al-Hufofi
- Department of Veterinary Laboratory, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Osman Hashim
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Ali Alkarar
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Ali Abu-Obeida
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Albagir Gaafer
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Abdelhamid Elfadil
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Ahmed Zaki
- Department of Veterinary Laboratory, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Ahmed Al-Romaihi
- Department of Veterinary Laboratory, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Nasereldeen Babekr
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Nadr El-Harby
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Raed Hussien
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Ali Al-Sahaf
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Ali Al-Doweriej
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Faisal Bayoumi
- Department of Animal Resources, Ministry of Environment, Water and Agriculture, 65 King Abdulaziz Road, Riyadh 11195, Saudi Arabia
| | - Leo L M Poon
- Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Daniel K W Chu
- Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Malik Peiris
- Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong, China
| | - Ranawaka A P M Perera
- Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong, China
| |
Collapse
|