1
|
Mhlekude B, Postmus D, Stenzel S, Weiner J, Jansen J, Zapatero-Belinchón FJ, Olmer R, Richter A, Heinze J, Heinemann N, Mühlemann B, Schroeder S, Jones TC, Müller MA, Drosten C, Pich A, Thiel V, Martin U, Niemeyer D, Gerold G, Beule D, Goffinet C. Pharmacological inhibition of bromodomain and extra-terminal proteins induces an NRF-2-mediated antiviral state that is subverted by SARS-CoV-2 infection. PLoS Pathog 2023; 19:e1011657. [PMID: 37747932 PMCID: PMC10629670 DOI: 10.1371/journal.ppat.1011657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/07/2023] [Accepted: 09/04/2023] [Indexed: 09/27/2023] Open
Abstract
Inhibitors of bromodomain and extra-terminal proteins (iBETs), including JQ-1, have been suggested as potential prophylactics against SARS-CoV-2 infection. However, molecular mechanisms underlying JQ-1-mediated antiviral activity and its susceptibility to viral subversion remain incompletely understood. Pretreatment of cells with iBETs inhibited infection by SARS-CoV-2 variants and SARS-CoV, but not MERS-CoV. The antiviral activity manifested itself by reduced reporter expression of recombinant viruses, and reduced viral RNA quantities and infectious titers in the culture supernatant. While we confirmed JQ-1-mediated downregulation of expression of angiotensin-converting enzyme 2 (ACE2) and interferon-stimulated genes (ISGs), multi-omics analysis addressing the chromatin accessibility, transcriptome and proteome uncovered induction of an antiviral nuclear factor erythroid 2-related factor 2 (NRF-2)-mediated cytoprotective response as an additional mechanism through which JQ-1 inhibits SARS-CoV-2 replication. Pharmacological inhibition of NRF-2, and knockdown of NRF-2 and its target genes reduced JQ-1-mediated inhibition of SARS-CoV-2 replication. Serial passaging of SARS-CoV-2 in the presence of JQ-1 resulted in predominance of ORF6-deficient variant, which exhibited resistance to JQ-1 and increased sensitivity to exogenously administered type I interferon (IFN-I), suggesting a minimised need for SARS-CoV-2 ORF6-mediated repression of IFN signalling in the presence of JQ-1. Importantly, JQ-1 exhibited a transient antiviral activity when administered prophylactically in human airway bronchial epithelial cells (hBAECs), which was gradually subverted by SARS-CoV-2, and no antiviral activity when administered therapeutically following an established infection. We propose that JQ-1 exerts pleiotropic effects that collectively induce an antiviral state in the host, which is ultimately nullified by SARS-CoV-2 infection, raising questions about the clinical suitability of the iBETs in the context of COVID-19.
Collapse
Affiliation(s)
- Baxolele Mhlekude
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Virology and Innate Immunity Research Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Institute of Genetics, Technische Universität Braunschweig, Braunschweig, Germany
| | - Dylan Postmus
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Saskia Stenzel
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - January Weiner
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Jenny Jansen
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Francisco J. Zapatero-Belinchón
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Department of Clinical Microbiology, Virology & Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Ruth Olmer
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH—Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Anja Richter
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Julian Heinze
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nicolas Heinemann
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Barbara Mühlemann
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Simon Schroeder
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Terry C. Jones
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Centre for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge, United Kingdom
| | - Marcel A. Müller
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian Drosten
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Andreas Pich
- Institute of Toxicology, Hannover Medical School, Core Facility Proteomics, Hannover, Germany
| | - Volker Thiel
- Institute of Virology and Immunology (IVI), University of Bern, Bern, Switzerland
- Department of Infectious Diseases and Pathobiology, Vetsuisse Faculty, University of Bern, Bern, Switzerland
| | - Ulrich Martin
- Leibniz Research Laboratories for Biotechnology and Artificial Organs (LEBAO), Department of Cardiothoracic, Transplantation and Vascular Surgery, REBIRTH—Center for Translational Regenerative Medicine, Biomedical Research in Endstage and Obstructive Lung Disease Hannover (BREATH), German Center for Lung Research (DZL), Hannover Medical School, Hannover, Germany
| | - Daniela Niemeyer
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Gisa Gerold
- Department of Biochemistry, University of Veterinary Medicine Hannover, Hannover, Germany
- Institute of Experimental Virology, TWINCORE, Centre for Experimental and Clinical Infection Research; a joint venture between the Hannover Medical School and the Helmholtz Centre for Infection Research, Hannover, Germany
- Department of Clinical Microbiology, Virology & Wallenberg Centre for Molecular Medicine (WCMM), Umeå University, Umeå, Sweden
| | - Dieter Beule
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| | - Christine Goffinet
- Institute of Virology, Campus Charité Mitte, Charité—Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool United Kingdom
| |
Collapse
|
2
|
Kazmierski J, Friedmann K, Postmus D, Emanuel J, Fischer C, Jansen J, Richter A, Bosquillon de Jarcy L, Schüler C, Sohn M, Sauer S, Drosten C, Saliba A, Sander LE, Müller MA, Niemeyer D, Goffinet C. Nonproductive exposure of PBMCs to SARS-CoV-2 induces cell-intrinsic innate immune responses. Mol Syst Biol 2022; 18:e10961. [PMID: 35975552 PMCID: PMC9382356 DOI: 10.15252/msb.202210961] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 07/28/2022] [Accepted: 07/28/2022] [Indexed: 12/15/2022] Open
Abstract
Cell-intrinsic responses mounted in PBMCs during mild and severe COVID-19 differ quantitatively and qualitatively. Whether they are triggered by signals emitted by productively infected cells of the respiratory tract or result from physical interaction with virus particles remains unclear. Here, we analyzed susceptibility and expression profiles of PBMCs from healthy donors upon ex vivo exposure to SARS-CoV and SARS-CoV-2. In line with the absence of detectable ACE2 receptor expression, human PBMCs were refractory to productive infection. RT-PCR experiments and single-cell RNA sequencing revealed JAK/STAT-dependent induction of interferon-stimulated genes (ISGs) but not proinflammatory cytokines. This SARS-CoV-2-specific response was most pronounced in monocytes. SARS-CoV-2-RNA-positive monocytes displayed a lower ISG signature as compared to bystander cells of the identical culture. This suggests a preferential invasion of cells with a low ISG baseline profile or delivery of a SARS-CoV-2-specific sensing antagonist upon efficient particle internalization. Together, nonproductive physical interaction of PBMCs with SARS-CoV-2- and, to a much lesser extent, SARS-CoV particles stimulate JAK/STAT-dependent, monocyte-accentuated innate immune responses that resemble those detected in vivo in patients with mild COVID-19.
Collapse
Affiliation(s)
- Julia Kazmierski
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Kirstin Friedmann
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Dylan Postmus
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Jackson Emanuel
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Cornelius Fischer
- Scientific Genomics Platforms, Laboratory of Functional Genomics, Nutrigenomics and Systems BiologyMax Delbrück Center for Molecular MedicineBerlinGermany
| | - Jenny Jansen
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Anja Richter
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
| | - Laure Bosquillon de Jarcy
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Department of Infectious Diseases and Respiratory MedicineCharité ‐ Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health (BIH)BerlinGermany
| | - Christiane Schüler
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| | - Madlen Sohn
- Scientific Genomics Platforms, Laboratory of Functional Genomics, Nutrigenomics and Systems BiologyMax Delbrück Center for Molecular MedicineBerlinGermany
| | - Sascha Sauer
- Scientific Genomics Platforms, Laboratory of Functional Genomics, Nutrigenomics and Systems BiologyMax Delbrück Center for Molecular MedicineBerlinGermany
| | - Christian Drosten
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- German Center for Infection Research, Associated Partner CharitéBerlinGermany
| | - Antoine‐Emmanuel Saliba
- Helmholtz Institute for RNA‐based Infection Research (HIRI)Helmholtz‐Center for Infection Research (HZI)WürzburgGermany
| | - Leif Erik Sander
- Department of Infectious Diseases and Respiratory MedicineCharité ‐ Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health (BIH)BerlinGermany
| | - Marcel A Müller
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- German Center for Infection Research, Associated Partner CharitéBerlinGermany
| | - Daniela Niemeyer
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- German Center for Infection Research, Associated Partner CharitéBerlinGermany
| | - Christine Goffinet
- Institute of Virology, Campus Charité Mitte, Charité – Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of HealthBerlinGermany
| |
Collapse
|
3
|
Natarelli L, Virgili F, Weber C. SARS-CoV-2, Cardiovascular Diseases, and Noncoding RNAs: A Connected Triad. Int J Mol Sci 2021; 22:12243. [PMID: 34830125 PMCID: PMC8620514 DOI: 10.3390/ijms222212243] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 12/23/2022] Open
Abstract
Coronavirus Disease 2019 (COVID-19), caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), is characterized by important respiratory impairments frequently associated with severe cardiovascular damages. Moreover, patients with pre-existing comorbidity for cardiovascular diseases (CVD) often present a dramatic increase in inflammatory cytokines release, which increases the severity and adverse outcomes of the infection and, finally, mortality risk. Despite this evident association at the clinical level, the mechanisms linking CVD and COVID-19 are still blurry and unresolved. Noncoding RNAs (ncRNAs) are functional RNA molecules transcribed from DNA but usually not translated into proteins. They play an important role in the regulation of gene expression, either in relatively stable conditions or as a response to different stimuli, including viral infection, and are therefore considered a possible important target in the design of specific drugs. In this review, we introduce known associations and interactions between COVID-19 and CVD, discussing the role of ncRNAs within SARS-CoV-2 infection from the perspective of the development of efficient pharmacological tools to treat COVID-19 patients and taking into account the equally dramatic associated consequences, such as those affecting the cardiovascular system.
Collapse
Affiliation(s)
- Lucia Natarelli
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), 800336 Munich, Germany;
| | - Fabio Virgili
- Research Center for Food and Nutrition, Council for Agricultural Research and Economics, 00178 Rome, Italy;
| | - Christian Weber
- Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-Universität (LMU), 800336 Munich, Germany;
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, 80336 Munich, Germany
- Department of Biochemistry, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, 6229 HX Maastricht, The Netherlands
- Munich Cluster for Systems Neurology (SyNergy), Institute for Stroke and Dementia Research, 81377 Munich, Germany
| |
Collapse
|
4
|
Deif MA, Solyman AAA, Kamarposhti MA, Band SS, Hammam RE. A deep bidirectional recurrent neural network for identification of SARS-CoV-2 from viral genome sequences. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2021; 18:8933-8950. [PMID: 34814329 DOI: 10.3934/mbe.2021440] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
In this work, Deep Bidirectional Recurrent Neural Networks (BRNNs) models were implemented based on both Long Short-Term Memory (LSTM) and Gated Recurrent Unit (GRU) cells in order to distinguish between genome sequence of SARS-CoV-2 and other Corona Virus strains such as SARS-CoV and MERS-CoV, Common Cold and other Acute Respiratory Infection (ARI) viruses. An investigation of the hyper-parameters including the optimizer type and the number of unit cells, was also performed to attain the best performance of the BRNN models. Results showed that the GRU BRNNs model was able to discriminate between SARS-CoV-2 and other classes of viruses with a higher overall classification accuracy of 96.8% as compared to that of the LSTM BRNNs model having a 95.8% overall classification accuracy. The best hyper-parameters producing the highest performance for both models was obtained when applying the SGD optimizer and an optimum number of unit cells of 80 in both models. This study proved that the proposed GRU BRNN model has a better classification ability for SARS-CoV-2 thus providing an efficient tool to help in containing the disease and achieving better clinical decisions with high precision.
Collapse
Affiliation(s)
- Mohanad A Deif
- Department of Bioelectronics, Modern University of Technology and Information (MTI) University, Cairo 11571, Egypt
| | - Ahmed A A Solyman
- Department of Electrical and Electronics Engineering, Istanbul Gelisim University, Avcılar 34310, Turkey
| | | | - Shahab S Band
- Future Technology Research Center, College of Future, National Yunlin University of Science and Technology, 123 University Road, Yunlin 64002, Taiwan
| | - Rania E Hammam
- Department of Bioelectronics, Modern University of Technology and Information (MTI) University, Cairo 11571, Egypt
| |
Collapse
|
5
|
Rzymski P, Mamzer H, Nowicki M. The Main Sources and Potential Effects of COVID-19-Related Discrimination. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1318:705-725. [PMID: 33973207 DOI: 10.1007/978-3-030-63761-3_39] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The outbreak of a new coronavirus disease (COVID-19), which appeared in late 2019 and eventually resulted in the announcement of a pandemic by the World Health Organization, led to global fear and panic as well as the spread of false information and fake news from different sources. As a result, a sharp increase in prejudice, discrimination, and xenophobia against different groups of people was observed in different geographical locations. This chapter presents the psychological and social sources of stereotypes and prejudices that take forms in the COVID-19 pandemic. These sources can be located in psychosocial processes, such as (i) socially generated and reinforced fears; (ii) human responses to stress induced by certain types of stimuli; (iii) sense of helplessness based on the lack of control over reality; (iv) psychological responses reinforced by conformism (crowd psychology); and (v) the stigmatization process. The chapter also presents the main groups of increased risk of experiencing prejudice and discrimination during the COVID-19 pandemic (Asians, health-care workers, COVID-19 patients, and their relatives). Moreover, it provides a documented example of such behaviors. The groups at higher risk of more adverse effects of COVID-19 due to pre-pandemic discrimination are also discussed. Finally, initiatives taken to mitigate the discrimination associated with COVID-19 are presented, as well as the recommendations and good practices for preventing these behaviors during future outbreaks and for limiting discrimination against COVID-19 until the disease can be contained.
Collapse
Affiliation(s)
- Piotr Rzymski
- Department of Environmental Medicine, Poznan University of Medical Sciences, Poznań, Poland. .,Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Poznań, Poland.
| | - Hanna Mamzer
- Faculty of Sociology Adam Mickiewicz University, Szamarzewskiego 89 c, 60-568, Poznań, Poland
| | - Michał Nowicki
- Chair of Histology and Embryology, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
6
|
SARS-CoV-2 and Three Related Coronaviruses Utilize Multiple ACE2 Orthologs and Are Potently Blocked by an Improved ACE2-Ig. J Virol 2020; 94:JVI.01283-20. [PMID: 32847856 PMCID: PMC7592233 DOI: 10.1128/jvi.01283-20] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 08/20/2020] [Indexed: 12/17/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological agent of the currently uncontrolled coronavirus disease 2019 (COVID-19) pandemic. It is important to study the host range of SARS-CoV-2, because some domestic species might harbor the virus and transmit it back to humans. In addition, insight into the ability of SARS-CoV-2 and SARS-like viruses to utilize animal orthologs of the SARS-CoV-2 receptor ACE2 might provide structural insight into improving ACE2-based viral entry inhibitors. In this study, we found that ACE2 orthologs of a wide range of domestic and wild animals can support cell entry of SARS-CoV-2 and three related coronaviruses, providing insights into identifying animal hosts of these viruses. We also developed recombinant ACE2-Ig proteins that are able to potently block these viral infections, providing a promising approach to developing antiviral proteins broadly effective against these distinct coronaviruses. The ongoing coronavirus disease 2019 (COVID-19) pandemic has caused >20 million infections and >750,000 deaths. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiological agent of COVID-19, has been found closely related to the bat coronavirus strain RaTG13 (Bat-CoV RaTG13) and a recently identified pangolin coronavirus (Pangolin-CoV-2020). Here, we first investigated the ability of SARS-CoV-2 and three related coronaviruses to utilize animal orthologs of angiotensin-converting enzyme 2 (ACE2) for cell entry. We found that ACE2 orthologs of a wide range of domestic and wild mammals, including camels, cattle, horses, goats, sheep, cats, rabbits, and pangolins, were able to support cell entry of SARS-CoV-2, suggesting that these species might be able to harbor and spread this virus. In addition, the pangolin and bat coronaviruses, Pangolin-CoV-2020 and Bat-CoV RaTG13, were also found able to utilize human ACE2 and a number of animal-ACE2 orthologs for cell entry, indicating risks of spillover of these viruses into humans in the future. We then developed potently anticoronavirus ACE2-Ig proteins that are broadly effective against the four distinct coronaviruses. In particular, through truncating ACE2 at its residue 740 but not 615, introducing a D30E mutation, and adopting an antibody-like tetrameric-ACE2 configuration, we generated an ACE2-Ig variant that neutralizes SARS-CoV-2 at picomolar range. These data demonstrate that the improved ACE2-Ig variants developed in this study could potentially be developed to protect from SARS-CoV-2 and some other SARS-like viruses that might spillover into humans in the future. IMPORTANCE The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the etiological agent of the currently uncontrolled coronavirus disease 2019 (COVID-19) pandemic. It is important to study the host range of SARS-CoV-2, because some domestic species might harbor the virus and transmit it back to humans. In addition, insight into the ability of SARS-CoV-2 and SARS-like viruses to utilize animal orthologs of the SARS-CoV-2 receptor ACE2 might provide structural insight into improving ACE2-based viral entry inhibitors. In this study, we found that ACE2 orthologs of a wide range of domestic and wild animals can support cell entry of SARS-CoV-2 and three related coronaviruses, providing insights into identifying animal hosts of these viruses. We also developed recombinant ACE2-Ig proteins that are able to potently block these viral infections, providing a promising approach to developing antiviral proteins broadly effective against these distinct coronaviruses.
Collapse
|
7
|
Baldassarre A, Paolini A, Bruno SP, Felli C, Tozzi AE, Masotti A. Potential use of noncoding RNAs and innovative therapeutic strategies to target the 5'UTR of SARS-CoV-2. Epigenomics 2020; 12:1349-1361. [PMID: 32875809 PMCID: PMC7466951 DOI: 10.2217/epi-2020-0162] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
After the increasing number of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections all over the world, researchers and clinicians are struggling to find a vaccine or innovative therapeutic strategies to treat this viral infection. The severe acute respiratory syndrome coronavirus infection that occurred in 2002, Middle East respiratory syndrome (MERS) and other more common infectious diseases such as hepatitis C virus, led to the discovery of many RNA-based drugs. Among them, siRNAs and antisense locked nucleic acids have been demonstrated to have effective antiviral effects both in animal models and humans. Owing to the high genomic homology of SARS-CoV-2 and severe acute respiratory syndrome coronavirus (80–82%) the use of these molecules could be employed successfully also to target this emerging coronavirus. Trying to translate this approach to treat COVID-19, we analyzed the common structural features of viral 5’UTR regions that can be targeted by noncoding RNAs and we also identified miRNAs binding sites suitable for designing RNA-based drugs to be employed successfully against SARS-CoV-2.
Collapse
Affiliation(s)
- Antonella Baldassarre
- Children's Hospital Bambino Gesù-IRCCS, Research Laboratories; Multifactorial & Complex Phenotype Research Area, V.le di San Paolo 15, Rome 00146, Italy
| | - Alessandro Paolini
- Children's Hospital Bambino Gesù-IRCCS, Research Laboratories; Multifactorial & Complex Phenotype Research Area, V.le di San Paolo 15, Rome 00146, Italy
| | - Stefania Paola Bruno
- Children's Hospital Bambino Gesù-IRCCS, Research Laboratories; Multifactorial & Complex Phenotype Research Area, V.le di San Paolo 15, Rome 00146, Italy
| | - Cristina Felli
- Children's Hospital Bambino Gesù-IRCCS, Research Laboratories; Multifactorial & Complex Phenotype Research Area, V.le di San Paolo 15, Rome 00146, Italy
| | - Alberto Eugenio Tozzi
- Children's Hospital Bambino Gesù-IRCCS, Research Laboratories; Multifactorial & Complex Phenotype Research Area, V.le di San Paolo 15, Rome 00146, Italy
| | - Andrea Masotti
- Children's Hospital Bambino Gesù-IRCCS, Research Laboratories; Multifactorial & Complex Phenotype Research Area, V.le di San Paolo 15, Rome 00146, Italy
| |
Collapse
|
8
|
Pessi A. Cholesterol-conjugated peptide antivirals: a path to a rapid response to emerging viral diseases. J Pept Sci 2014; 21:379-86. [PMID: 25331523 PMCID: PMC7167725 DOI: 10.1002/psc.2706] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/01/2014] [Accepted: 09/15/2014] [Indexed: 12/18/2022]
Abstract
While it is now possible to identify and genetically fingerprint the causative agents of emerging viral diseases, often with extraordinary speed, suitable therapies cannot be developed with equivalent speed, because drug discovery requires information that goes beyond knowledge of the viral genome. Peptides, however, may represent a special opportunity. For all enveloped viruses, fusion between the viral and the target cell membrane is an obligatory step of the life cycle. Class I fusion proteins harbor regions with a repeating pattern of amino acids, the heptad repeats (HRs), that play a key role in fusion, and HR‐derived peptides such as enfuvirtide, in clinical use for HIV, can block the process. Because of their characteristic sequence pattern, HRs are easily identified in the genome by means of computer programs, providing the sequence of candidate peptide inhibitors directly from genomic information. Moreover, a simple chemical modification, the attachment of a cholesterol group, can dramatically increase the antiviral potency of HR‐derived inhibitors and simultaneously improve their pharmacokinetics. Further enhancement can be provided by dimerization of the cholesterol‐conjugated peptide. The examples reported so far include inhibitors of retroviruses, paramyxoviruses, orthomyxoviruses, henipaviruses, coronaviruses, and filoviruses. For some of these viruses, in vivo efficacy has been demonstrated in suitable animal models. The combination of bioinformatic lead identification and potency/pharmacokinetics improvement provided by cholesterol conjugation may form the basis for a rapid response strategy, where development of an emergency cholesterol‐conjugated therapeutic would immediately follow the availability of the genetic information of a new enveloped virus. Copyright © 2014 European Peptide Society and John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Antonello Pessi
- PeptiPharma, Viale Città D'Europa 679, 00141, Roma, Italy; JV Bio, Via Gaetano Salvatore 486, 80145, Napoli, Italy; CEINGE, Via Gaetano Salvatore 486, 80145, Napoli, Italy
| |
Collapse
|
9
|
de Wilde AH, Raj VS, Oudshoorn D, Bestebroer TM, van Nieuwkoop S, Limpens RWAL, Posthuma CC, van der Meer Y, Bárcena M, Haagmans BL, Snijder EJ, van den Hoogen BG. MERS-coronavirus replication induces severe in vitro cytopathology and is strongly inhibited by cyclosporin A or interferon-α treatment. J Gen Virol 2013; 94:1749-1760. [PMID: 23620378 PMCID: PMC3749523 DOI: 10.1099/vir.0.052910-0] [Citation(s) in RCA: 274] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Coronavirus (CoV) infections are commonly associated with respiratory and enteric disease in humans and animals. The 2003 outbreak of severe acute respiratory syndrome (SARS) highlighted the potentially lethal consequences of CoV-induced disease in humans. In 2012, a novel CoV (Middle East Respiratory Syndrome coronavirus; MERS-CoV) emerged, causing 49 human cases thus far, of which 23 had a fatal outcome. In this study, we characterized MERS-CoV replication and cytotoxicity in human and monkey cell lines. Electron microscopy of infected Vero cells revealed extensive membrane rearrangements, including the formation of double-membrane vesicles and convoluted membranes, which have been implicated previously in the RNA synthesis of SARS-CoV and other CoVs. Following infection, we observed rapidly increasing viral RNA synthesis and release of high titres of infectious progeny, followed by a pronounced cytopathology. These characteristics were used to develop an assay for antiviral compound screening in 96-well format, which was used to identify cyclosporin A as an inhibitor of MERS-CoV replication in cell culture. Furthermore, MERS-CoV was found to be 50–100 times more sensitive to alpha interferon (IFN-α) treatment than SARS-CoV, an observation that may have important implications for the treatment of MERS-CoV-infected patients. MERS-CoV infection did not prevent the IFN-induced nuclear translocation of phosphorylated STAT1, in contrast to infection with SARS-CoV where this block inhibits the expression of antiviral genes. These findings highlight relevant differences between these distantly related zoonotic CoVs in terms of their interaction with and evasion of the cellular innate immune response.
Collapse
Affiliation(s)
- Adriaan H de Wilde
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - V Stalin Raj
- Viroscience Lab, Erasmus MC, Rotterdam, The Netherlands
| | - Diede Oudshoorn
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Ronald W A L Limpens
- Section Electron Microscopy, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | - Clara C Posthuma
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Yvonne van der Meer
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Montserrat Bárcena
- Section Electron Microscopy, Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Eric J Snijder
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
10
|
Pessi A, Langella A, Capitò E, Ghezzi S, Vicenzi E, Poli G, Ketas T, Mathieu C, Cortese R, Horvat B, Moscona A, Porotto M. A general strategy to endow natural fusion-protein-derived peptides with potent antiviral activity. PLoS One 2012; 7:e36833. [PMID: 22666328 PMCID: PMC3353973 DOI: 10.1371/journal.pone.0036833] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 04/07/2012] [Indexed: 01/26/2023] Open
Abstract
Fusion between the viral and target cell membranes is an obligatory step for the infectivity of all enveloped virus, and blocking this process is a clinically validated therapeutic strategy. Viral fusion is driven by specialized proteins which, although specific to each virus, act through a common mechanism, the formation of a complex between two heptad repeat (HR) regions. The HR regions are initially separated in an intermediate termed “prehairpin”, which bridges the viral and cell membranes, and then fold onto each other to form a 6-helical bundle (6HB), driving the two membranes to fuse. HR-derived peptides can inhibit viral infectivity by binding to the prehairpin intermediate and preventing its transition to the 6HB. The antiviral activity of HR-derived peptides differs considerably among enveloped viruses. For weak inhibitors, potency can be increased by peptide engineering strategies, but sequence-specific optimization is time-consuming. In seeking ways to increase potency without changing the native sequence, we previously reported that attachment to the HR peptide of a cholesterol group (”cholesterol-tagging”) dramatically increases its antiviral potency, and simultaneously increases its half-life in vivo. We show here that antiviral potency may be increased by combining cholesterol-tagging with dimerization of the HR-derived sequence, using as examples human parainfluenza virus, Nipah virus, and HIV-1. Together, cholesterol-tagging and dimerization may represent strategies to boost HR peptide potency to levels that in some cases may be compatible with in vivo use, possibly contributing to emergency responses to outbreaks of existing or novel viruses.
Collapse
Affiliation(s)
| | | | | | - Silvia Ghezzi
- Viral Pathogens and Biosafety, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Vicenzi
- Viral Pathogens and Biosafety, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Guido Poli
- AIDS Immunopathogenesis Units, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, School of Medicine, Milan, Italy
| | - Thomas Ketas
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Cyrille Mathieu
- INSERM, Ecole Normale Supérieure de Lyon, Lyon, France
- Pedriatics, Weill Medical College of Cornell University, New York, New York, United States of America
| | | | - Branka Horvat
- INSERM, Ecole Normale Supérieure de Lyon, Lyon, France
- IFR128 BioSciences Lyon-Gerland Lyon-Sud, University of Lyon, Lyon, France
| | - Anne Moscona
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
- Pedriatics, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Matteo Porotto
- Pedriatics, Weill Medical College of Cornell University, New York, New York, United States of America
- * E-mail: (AP); (MP)
| |
Collapse
|
11
|
van den Worm SHE, Eriksson KK, Zevenhoven JC, Weber F, Züst R, Kuri T, Dijkman R, Chang G, Siddell SG, Snijder EJ, Thiel V, Davidson AD. Reverse genetics of SARS-related coronavirus using vaccinia virus-based recombination. PLoS One 2012; 7:e32857. [PMID: 22412934 PMCID: PMC3296753 DOI: 10.1371/journal.pone.0032857] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 01/31/2012] [Indexed: 02/06/2023] Open
Abstract
Severe acute respiratory syndrome (SARS) is a zoonotic disease caused by SARS-related coronavirus (SARS-CoV) that emerged in 2002 to become a global health concern. Although the original outbreak was controlled by classical public health measures, there is a real risk that another SARS-CoV could re-emerge from its natural reservoir, either in its original form or as a more virulent or pathogenic strain; in which case, the virus would be difficult to control in the absence of any effective antiviral drugs or vaccines. Using the well-studied SARS-CoV isolate HKU-39849, we developed a vaccinia virus-based SARS-CoV reverse genetic system that is both robust and biosafe. The SARS-CoV genome was cloned in separate vaccinia virus vectors, (vSARS-CoV-5prime and vSARS-CoV-3prime) as two cDNAs that were subsequently ligated to create a genome-length SARS-CoV cDNA template for in vitro transcription of SARS-CoV infectious RNA transcripts. Transfection of the RNA transcripts into permissive cells led to the recovery of infectious virus (recSARS-CoV). Characterization of the plaques produced by recSARS-CoV showed that they were similar in size to the parental SARS-CoV isolate HKU-39849 but smaller than the SARS-CoV isolate Frankfurt-1. Comparative analysis of replication kinetics showed that the kinetics of recSARS-CoV replication are similar to those of SARS-CoV Frankfurt-1, although the titers of virus released into the culture supernatant are approximately 10-fold less. The reverse genetic system was finally used to generate a recSARS-CoV reporter virus expressing Renilla luciferase in order to facilitate the analysis of SARS-CoV gene expression in human dendritic cells (hDCs). In parallel, a Renilla luciferase gene was also inserted into the genome of human coronavirus 229E (HCoV-229E). Using this approach, we demonstrate that, in contrast to HCoV-229E, SARS-CoV is not able to mediate efficient heterologous gene expression in hDCs.
Collapse
Affiliation(s)
- Sjoerd H. E. van den Worm
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Jessika C. Zevenhoven
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Friedemann Weber
- Department of Virology, University of Freiburg, Freiburg, Germany
| | - Roland Züst
- Institute of Immunobiology, Kantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Thomas Kuri
- Department of Virology, University of Freiburg, Freiburg, Germany
| | - Ronald Dijkman
- Institute of Immunobiology, Kantonal Hospital St. Gallen, St. Gallen, Switzerland
| | - Guohui Chang
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Stuart G. Siddell
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
| | - Eric J. Snijder
- Molecular Virology Laboratory, Department of Medical Microbiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Volker Thiel
- Institute of Immunobiology, Kantonal Hospital St. Gallen, St. Gallen, Switzerland
- Vetsuisse Faculty, University of Zürich, Zurich, Switzerland
| | - Andrew D. Davidson
- School of Cellular and Molecular Medicine, University of Bristol, Bristol, United Kingdom
- * E-mail:
| |
Collapse
|
12
|
Kuri T, Eriksson KK, Putics A, Züst R, Snijder EJ, Davidson AD, Siddell SG, Thiel V, Ziebuhr J, Weber F. The ADP-ribose-1''-monophosphatase domains of severe acute respiratory syndrome coronavirus and human coronavirus 229E mediate resistance to antiviral interferon responses. J Gen Virol 2011; 92:1899-1905. [PMID: 21525212 DOI: 10.1099/vir.0.031856-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Several plus-strand RNA viruses encode proteins containing macrodomains. These domains possess ADP-ribose-1″-phosphatase (ADRP) activity and/or bind poly(ADP-ribose), poly(A) or poly(G). The relevance of these activities in the viral life cycle has not yet been resolved. Here, we report that genetically engineered mutants of severe acute respiratory syndrome coronavirus (SARS-CoV) and human coronavirus 229E (HCoV-229E) expressing ADRP-deficient macrodomains displayed an increased sensitivity to the antiviral effect of alpha interferon compared with their wild-type counterparts. The data suggest that macrodomain-associated ADRP activities may have a role in viral escape from the innate immune responses of the host.
Collapse
Affiliation(s)
- Thomas Kuri
- Department of Virology, University of Freiburg, D-79008 Freiburg, Germany
| | - Klara K Eriksson
- Research Department, Kantonal Hospital St Gallen, St Gallen, Switzerland
| | - Akos Putics
- Institute of Virology and Immunology, University of Würzburg, D-97078 Würzburg, Germany
| | - Roland Züst
- Research Department, Kantonal Hospital St Gallen, St Gallen, Switzerland
| | - Eric J Snijder
- Molecular Virology Laboratory, Department of Medical Microbiology, Center of Infectious Diseases, Leiden University Medical Center, Leiden, The Netherlands
| | - Andrew D Davidson
- Division of Virology, Department of Cellular and Molecular Medicine, School of Medical and Veterinary Sciences, University of Bristol, Bristol, UK
| | - Stuart G Siddell
- Division of Virology, Department of Cellular and Molecular Medicine, School of Medical and Veterinary Sciences, University of Bristol, Bristol, UK
| | - Volker Thiel
- Research Department, Kantonal Hospital St Gallen, St Gallen, Switzerland
| | - John Ziebuhr
- Institute of Medical Virology, Justus Liebig University Giessen, D-35392 Giessen, Germany
| | - Friedemann Weber
- Institute for Virology, Philipps-University Marburg, D-35043 Marburg, Germany.,Department of Virology, University of Freiburg, D-79008 Freiburg, Germany
| |
Collapse
|
13
|
Rockx B, Feldmann F, Brining D, Gardner D, LaCasse R, Kercher L, Long D, Rosenke R, Virtaneva K, Sturdevant DE, Porcella SF, Mattoon J, Parnell M, Baric RS, Feldmann H. Comparative pathogenesis of three human and zoonotic SARS-CoV strains in cynomolgus macaques. PLoS One 2011; 6:e18558. [PMID: 21533129 PMCID: PMC3080360 DOI: 10.1371/journal.pone.0018558] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2011] [Accepted: 03/04/2011] [Indexed: 01/13/2023] Open
Abstract
The severe acute respiratory syndrome (SARS) epidemic was characterized by increased pathogenicity in the elderly due to an early exacerbated innate host response. SARS-CoV is a zoonotic pathogen that entered the human population through an intermediate host like the palm civet. To prevent future introductions of zoonotic SARS-CoV strains and subsequent transmission into the human population, heterologous disease models are needed to test the efficacy of vaccines and therapeutics against both late human and zoonotic isolates. Here we show that both human and zoonotic SARS-CoV strains can infect cynomolgus macaques and resulted in radiological as well as histopathological changes similar to those seen in mild human cases. Viral replication was higher in animals infected with a late human phase isolate compared to a zoonotic isolate. While there were significant differences in the number of host genes differentially regulated during the host responses between the three SARS-CoV strains, the top pathways and functions were similar and only apparent early during infection with the majority of genes associated with interferon signaling pathways. This study characterizes critical disease models in the evaluation and licensure of therapeutic strategies against SARS-CoV for human use.
Collapse
Affiliation(s)
- Barry Rockx
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, Montana, United States of America.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Cao YL, Wang Y, Guo R, Yang F, Zhang Y, Wang SH, Liu L. Identification and characterization of three novel small interference RNAs that effectively down-regulate the isolated nucleocapsid gene expression of SARS coronavirus. Molecules 2011; 16:1544-58. [PMID: 21317844 PMCID: PMC6259856 DOI: 10.3390/molecules16021544] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 02/09/2011] [Indexed: 01/20/2023] Open
Abstract
Nucleocapsid (N) protein of severe acute respiratory syndrome-associated coronavirus (SARS-CoV) is a major pathological determinant in the host that may cause host cell apoptosis, upregulate the proinflammatory cytokine production, and block innate immune responses. Therefore, N gene has long been thought an ideal target for the design of small interference RNA (siRNA). siRNA is a class of small non-coding RNAs with a size of 21-25nt that functions post-transcriptionally to block targeted gene expression. In this study, we analyzed the N gene coding sequences derived from 16 different isolates, and found that nucleotide deletions and substitutions are mainly located at the first 440nt sequence. Combining previous reports and the above sequence information, we create three novel siRNAs that specifically target the conserved and unexploited regions in the N gene. We show that these siRNAs could effectively and specifically block the isolated N gene expression in mammal cells. Furthermore, we provide evidence to show that N gene can effectively up-regulate M gene mediated interferon β (IFNβ) production, while blocking N gene expression by specific siRNA significantly reduces IFNβ gene expression. Our data indicate that the inhibitory effect of siRNA on the isolated N gene expression might be influenced by the sequence context around the targeted sites.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Liu
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86 10 65592203; Fax: +86 10 62737136
| |
Collapse
|
15
|
Lam TTY, Hon CC, Tang JW. Use of phylogenetics in the molecular epidemiology and evolutionary studies of viral infections. Crit Rev Clin Lab Sci 2010; 47:5-49. [PMID: 20367503 DOI: 10.3109/10408361003633318] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Since DNA sequencing techniques first became available almost 30 years ago, the amount of nucleic acid sequence data has increased enormously. Phylogenetics, which is widely applied to compare and analyze such data, is particularly useful for the analysis of genes from rapidly evolving viruses. It has been used extensively to describe the molecular epidemiology and transmission of the human immunodeficiency virus (HIV), the origins and subsequent evolution of the severe acute respiratory syndrome (SARS)-associated coronavirus (SCoV), and, more recently, the evolving epidemiology of avian influenza as well as seasonal and pandemic human influenza viruses. Recent advances in phylogenetic methods can infer more in-depth information about the patterns of virus emergence, adding to the conventional approaches in viral epidemiology. Examples of this information include estimations (with confidence limits) of the actual time of the origin of a new viral strain or its emergence in a new species, viral recombination and reassortment events, the rate of population size change in a viral epidemic, and how the virus spreads and evolves within a specific population and geographical region. Such sequence-derived information obtained from the phylogenetic tree can assist in the design and implementation of public health and therapeutic interventions. However, application of many of these advanced phylogenetic methods are currently limited to specialized phylogeneticists and statisticians, mainly because of their mathematical basis and their dependence on the use of a large number of computer programs. This review attempts to bridge this gap by presenting conceptual, technical, and practical aspects of applying phylogenetic methods in studies of influenza, HIV, and SCoV. It aims to provide, with minimal mathematics and statistics, a practical overview of how phylogenetic methods can be incorporated into virological studies by clinical and laboratory specialists.
Collapse
Affiliation(s)
- Tommy Tsan-Yuk Lam
- School of Biological Sciences, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | | | | |
Collapse
|
16
|
Yip CW, Hon CC, Shi M, Lam TTY, Chow KYC, Zeng F, Leung FCC. Phylogenetic perspectives on the epidemiology and origins of SARS and SARS-like coronaviruses. INFECTION GENETICS AND EVOLUTION 2009; 9:1185-96. [PMID: 19800030 PMCID: PMC7106296 DOI: 10.1016/j.meegid.2009.09.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2009] [Revised: 08/09/2009] [Accepted: 09/24/2009] [Indexed: 11/24/2022]
Abstract
Severe Acute Respiratory Syndrome (SARS) is a respiratory disease caused by a zoonotic coronavirus (CoV) named SARS-CoV (SCoV), which rapidly swept the globe after its emergence in rural China during late 2002. The origins of SCoV have been mysterious and controversial, until the recent discovery of SARS-like CoV (SLCoV) in bats and the proposal of bats as the natural reservior of the Coronaviridae family. In this article, we focused on discussing how phylogenetics contributed to our understanding towards the emergence and transmission of SCoV. We first reviewed the epidemiology of SCoV from a phylogenetic perspective and discussed the controversies over its phylogenetic origins. Then, we summarized the phylogenetic findings in relation to its zoonotic origins and the proposed inter-species viral transmission events. Finally, we also discussed how the discoveries of SCoV and SLCoV expanded our knowledge on the evolution of the Coronaviridae family as well as its implications on the possible future re-emergence of SCoV.
Collapse
Affiliation(s)
- Chi Wai Yip
- The School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | | | | | | | | | | | | |
Collapse
|
17
|
Canard B, Joseph JS, Kuhn P. International research networks in viral structural proteomics: again, lessons from SARS. Antiviral Res 2008; 78:47-50. [PMID: 18054092 PMCID: PMC2793675 DOI: 10.1016/j.antiviral.2007.09.007] [Citation(s) in RCA: 5] [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] [Received: 08/10/2007] [Revised: 09/12/2007] [Accepted: 09/24/2007] [Indexed: 11/18/2022]
Abstract
Emerging and re-emerging pathogens and bioterror threats require an organized and coherent response from the worldwide research community to maximize available resources and competencies with the primary goals to understand the pathogen and enable intervention. In 2001, the Structural Proteomics In Europe (SPINE) project prototyped the pan-viral structural genomic approach, and the Severe Acute Respiratory Syndrome (SARS) outbreak in 2003 accelerated the concept of structural characterization of all proteins from a viral proteome and the interaction with their host partners. Following that approach, in 2004 the center for Functional and Structural Proteomics for SARS-CoV related proteins was initiated as part of the US NIH NIAID proteomics resource centers. Across worldwide efforts in Asia, Europe and America, the international research teams working on SARS-CoV have now determined experimental structural information for 45% of the SARS-CoV proteins and 53% of all its soluble proteins. This data is fully available to the scientific community and is providing an unprecedented level of insight to this class of RNA viruses. The efforts and results by the international scientific community to the SARS outbreak are serving as an example and roadmap of a rapid response using modern research methods.
Collapse
Affiliation(s)
- Bruno Canard
- Architecture Et Fonction Des Macromolécules Biologiques UMR 6098, CNRS, Universités Aix-Marseille I & II, Case 932, 163 Avenue de Luminy, 13288 Marseille Cedex 9, France
| | - Jeremiah S. Joseph
- Department of Cell Biology, The Scripps Research Institute, 10550 N Torrey Pines Road, CB265 La Jolla, CA 92037, USA
| | - Peter Kuhn
- Department of Cell Biology, The Scripps Research Institute, 10550 N Torrey Pines Road, CB265 La Jolla, CA 92037, USA
| |
Collapse
|
18
|
Nagata N, Iwata N, Hasegawa H, Sato Y, Morikawa S, Saijo M, Itamura S, Saito T, Ami Y, Odagiri T, Tashiro M, Sata T. Pathology and virus dispersion in cynomolgus monkeys experimentally infected with severe acute respiratory syndrome coronavirus via different inoculation routes. Int J Exp Pathol 2007; 88:403-14. [PMID: 18039277 PMCID: PMC2517337 DOI: 10.1111/j.1365-2613.2007.00567.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Severe acute respiratory syndrome-associated coronavirus (SARS-CoV) causes SARS. The pathogenic mechanisms of SARS-CoV remain poorly understood. Six cynomolgus monkeys were inoculated with the HKU39849 isolate of SARS-CoV via four routes. After intranasal inoculation, the virus was isolated from respiratory swabs on days 2–7 postinoculation (p.i.) and virus genome was detected in intestinal tissues on day 7 p.i. Virus was not detected after intragastric inoculation. After intravenous inoculation, infectious virus was isolated from rectal swabs, and virus antigen was detected in intestinal cells on day 14 p.i. After intratracheal (i.t.) inoculation, virus antigen-positive alveolar cells and macrophages were found in lung and infectious virus was detected in lymphoid and intestinal tissues. The peribronchial lymph nodes showed evidence of an immune response. Lung tissue and/or fluid and/or the peribronchial lymph node of the intratracheally inoculated animals had high TNF-α, IL-8 and IL-12 levels. SARS lung lesions are only generated in monkeys by i.t. inoculation. The virus appears to spread into and perhaps via the intestinal and lymphatic systems. It has been suggested previously that viraemia may cause intestinal infections in SARS patients.
Collapse
Affiliation(s)
- Noriyo Nagata
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hu MC, Jones T, Kenney RT, Barnard DL, Burt DS, Lowell GH. Intranasal Protollin-formulated recombinant SARS S-protein elicits respiratory and serum neutralizing antibodies and protection in mice. Vaccine 2007; 25:6334-40. [PMID: 17640780 PMCID: PMC7115497 DOI: 10.1016/j.vaccine.2007.06.017] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2007] [Revised: 05/30/2007] [Accepted: 06/11/2007] [Indexed: 12/03/2022]
Abstract
The feasibility of developing a prophylactic vaccine against SARS was assessed by comparing the immune responses elicited by immunizing mice with a recombinant SARS spike glycoprotein (S-protein) formulated with different adjuvants, given by different routes. In both young and aged mice, an intranasal Protollin-formulated S-protein vaccine elicited high levels of antigen-specific IgG in serum, comparable to those elicited by an intramuscular Alum-adsorbed S-protein vaccine. Serum antibodies were shown to be virus neutralizing. Intranasal immunization of young mice with the Protollin-formulated vaccine elicited significant levels of antigen-specific lung IgA in contrast to mice immunized with the intramuscular vaccine in which no antigen-specific lung IgA was detected. Following live virus challenge of aged mice, no virus was detected in the lungs of intranasally immunized mice, in contrast to intramuscularly immunized mice whose lung virus titers were comparable to those observed in control mice.
Collapse
Affiliation(s)
- Mary C. Hu
- GlaxoSmithKline Biologicals North America of Washington, 19204 North Creek Parkway, Bothell, WA 98011, USA
| | - Taff Jones
- GlaxoSmithKline Biologicals North America of Quebec, 525 Cartier Blvd. West, Laval, Montreal, Quebec H7V 3S8, Canada
- Corresponding author. Present address: MedImmune, 297 N. Bernardo Ave., Mountain View, CA 94043, USA. Tel.: +1 650 603 2617.
| | - Richard T. Kenney
- GlaxoSmithKline Biologicals North America of Maryland, 6996 Columbia Gateway Drive, Columbia, MA 21046, USA
| | - Dale L. Barnard
- Institute for Antiviral Research, Department of ADVS, Utah State University, Logan, UT 84322, USA
| | - David S. Burt
- GlaxoSmithKline Biologicals North America of Quebec, 525 Cartier Blvd. West, Laval, Montreal, Quebec H7V 3S8, Canada
| | - George H. Lowell
- GlaxoSmithKline Biologicals North America of Quebec, 525 Cartier Blvd. West, Laval, Montreal, Quebec H7V 3S8, Canada
| |
Collapse
|
20
|
Ba L, Yi CE, Zhang L, Ho DD, Chen Z. Heterologous MVA-S prime Ad5-S boost regimen induces high and persistent levels of neutralizing antibody response against SARS coronavirus. Appl Microbiol Biotechnol 2007; 76:1131-6. [PMID: 17581748 PMCID: PMC7079952 DOI: 10.1007/s00253-007-1073-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 05/30/2007] [Accepted: 06/01/2007] [Indexed: 11/11/2022]
Abstract
Severe acute respiratory syndrome (SARS) is caused by a novel coronavirus (CoV), SARS-CoV. In previous studies, we showed that a SARS-CoV spike (S) glycoprotein-based modified vaccinia Ankara (MVA-S) vaccine could induce strong neutralizing antibody (Nab) response which might have played a critical role in protecting Chinese rhesus monkeys from the pathogenic viral challenge. To date, however, it remains unknown what the minimal level of Nab is required to achieve sterile immunity in humans. It is therefore important to explore techniques to maximize the level of Nab response in vivo. Here, we evaluate various vaccination regimens using combinations of DNA-S, MVA-S, and adenovirus type 5 (Ad5-S) vaccines. We show that in vaccinated mice and rabbits, a heterologous MVA-S prime with Ad5-S boost regimen induces the highest and most persistent level of Nab response when compared with other combinations. Interestingly, the initial level of Nab after prime does not necessarily predict the magnitude of the secondary response after the boost. Thus, our data provides a promising optimal regimen for vaccine development in humans against SARS-CoV infection.
Collapse
Affiliation(s)
- Lei Ba
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 1st Avenue, New York, NY 10016 USA
| | - Christopher E. Yi
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 1st Avenue, New York, NY 10016 USA
| | - Linqi Zhang
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 1st Avenue, New York, NY 10016 USA
| | - David D. Ho
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 1st Avenue, New York, NY 10016 USA
| | - Zhiwei Chen
- Aaron Diamond AIDS Research Center, The Rockefeller University, 455 1st Avenue, New York, NY 10016 USA
| |
Collapse
|
21
|
Vijaykrishna D, Smith GJD, Zhang JX, Peiris JSM, Chen H, Guan Y. Evolutionary insights into the ecology of coronaviruses. J Virol 2007; 81:4012-20. [PMID: 17267506 PMCID: PMC1866124 DOI: 10.1128/jvi.02605-06] [Citation(s) in RCA: 192] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Although many novel members of the Coronaviridae have recently been recognized in different species, the ecology of coronaviruses has not been established. Our study indicates that bats harbor a much wider diversity of coronaviruses than any other animal species. Dating of different coronavirus lineages suggests that bat coronaviruses are older than those recognized in other animals and that the human severe acute respiratory syndrome (SARS) coronavirus was directly derived from viruses from wild animals in wet markets of southern China. Furthermore, the most closely related bat and SARS coronaviruses diverged in 1986, an estimated divergence time of 17 years prior to the outbreak, suggesting that there may have been transmission via an unknown intermediate host. Analysis of lineage-specific selection pressure also indicated that only SARS coronaviruses in civets and humans were under significant positive selection, also demonstrating a recent interspecies transmission. Analysis of population dynamics revealed that coronavirus populations in bats have constant population growth, while viruses from all other hosts show epidemic-like increases in population. These results indicate that diverse coronaviruses are endemic in different bat species, with repeated introductions to other animals and occasional establishment in other species. Our findings suggest that bats are likely the natural hosts for all presently known coronavirus lineages and that all coronaviruses recognized in other species were derived from viruses residing in bats. Further surveillance of bat and other animal populations is needed to fully describe the ecology and evolution of this virus family.
Collapse
Affiliation(s)
- D Vijaykrishna
- State Key Laboratory of Emerging Infectious Diseases, Department of Microbiology, The University of Hong Kong, Li Ka Shing Faculty of Medicine Building, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | | | | | | | | | | |
Collapse
|
22
|
Shih YP, Chen CY, Liu SJ, Chen KH, Lee YM, Chao YC, Chen YMA. Identifying epitopes responsible for neutralizing antibody and DC-SIGN binding on the spike glycoprotein of the severe acute respiratory syndrome coronavirus. J Virol 2006; 80:10315-24. [PMID: 17041212 PMCID: PMC1641789 DOI: 10.1128/jvi.01138-06] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The severe acute respiratory syndrome-associated coronavirus (SARS-CoV) uses dendritic cell-specific ICAM-3 grabbing nonintegrin (DC-SIGN) to facilitate cell entry via cellular receptor-angiotensin-converting enzyme 2. For this project, we used recombinant baculoviruses expressing different lengths of SARS-CoV spike (S) protein in a capture assay to deduce the minimal DC-SIGN binding region. Our results identified the region location between amino acid (aa) residues 324 to 386 of the S protein. We then generated nine monoclonal antibodies (MAbs) against the S protein to map the DC-SIGN-binding domain using capture assays with pseudotyped viruses and observed that MAb SIa5 significantly blocked S protein-DC-SIGN interaction. An enhancement assay using the HKU39849 SARS-CoV strain and human immature dendritic cells confirmed our observation. Data from a pepscan analysis and M13 phage peptide display library system mapped the reactive MAb SIa5 epitope to aa residues 363 to 368 of the S protein. Results from a capture assay testing three pseudotyped viruses with mutated N-linked glycosylation sites of the S protein indicate that only two pseudotyped viruses (N330Q and N357Q, both of which lost glycosylation sites near the SIa5 epitope) had diminished DC-SIGN-binding capacity. We also noted that MAb SIb4 exerted a neutralizing effect against HKU39849; its reactive epitope was mapped to aa residues 435 to 439 of the S protein. We offer the data to facilitate the development of therapeutic agents and preventive vaccines against SARS-CoV infection.
Collapse
MESH Headings
- Antibodies, Monoclonal
- Antibodies, Viral
- Antigens, Viral/genetics
- Baculoviridae/genetics
- Base Sequence
- Binding Sites/genetics
- Cell Adhesion Molecules/metabolism
- DNA, Viral/genetics
- Dendritic Cells/immunology
- Dendritic Cells/virology
- Epitope Mapping
- Epitopes/chemistry
- Epitopes/genetics
- Glycosylation
- Humans
- In Vitro Techniques
- Lectins, C-Type/metabolism
- Membrane Glycoproteins/chemistry
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Models, Molecular
- Mutagenesis, Site-Directed
- Neutralization Tests
- Protein Binding
- Protein Structure, Tertiary
- Receptors, Cell Surface/metabolism
- Recombinant Proteins/chemistry
- Recombinant Proteins/genetics
- Recombinant Proteins/immunology
- Severe acute respiratory syndrome-related coronavirus/genetics
- Severe acute respiratory syndrome-related coronavirus/immunology
- Severe Acute Respiratory Syndrome/immunology
- Severe Acute Respiratory Syndrome/virology
- Spike Glycoprotein, Coronavirus
- Viral Envelope Proteins/chemistry
- Viral Envelope Proteins/genetics
- Viral Envelope Proteins/immunology
Collapse
Affiliation(s)
- Yi-Ping Shih
- AIDS Prevention and Research Center, National Yang-Ming University, Taipei 111, Taiwan, Republic of China
| | | | | | | | | | | | | |
Collapse
|
23
|
Yip CW, Hon CC, Zeng F, Chow KYC, Chan KH, Peiris JSM, Leung FCC. Naturally occurring anti-Escherichia coli protein antibodies in the sera of healthy humans cause analytical interference in a recombinant nucleocapsid protein-based enzyme-linked immunosorbent assay for serodiagnosis of severe acute respiratory syndrome. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2006; 14:99-101. [PMID: 17108287 PMCID: PMC1797702 DOI: 10.1128/cvi.00136-06] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
We reported the analytical interference of anti-Escherichia coli protein (EP) antibodies in human sera and residual EP in a recombinant nucleocapsid protein-based enzyme-linked immunosorbent assay as a possible source of false positives in severe acute respiratory syndrome serodiagnosis. The rate of false positives was significantly reduced by adding mouse anti-EP antiserum in the blocking step.
Collapse
Affiliation(s)
- Chi Wai Yip
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong, SAR, China
| | | | | | | | | | | | | |
Collapse
|
24
|
Peters GA, Tyler SD, Grose C, Severini A, Gray MJ, Upton C, Tipples GA. A full-genome phylogenetic analysis of varicella-zoster virus reveals a novel origin of replication-based genotyping scheme and evidence of recombination between major circulating clades. J Virol 2006; 80:9850-60. [PMID: 16973589 PMCID: PMC1617253 DOI: 10.1128/jvi.00715-06] [Citation(s) in RCA: 101] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Varicella-zoster virus (VZV) is a remarkably stable virus that until recently was thought to exhibit near-universal genetic homogeneity among circulating wild-type strains. In recent years, the expanding knowledge of VZV genetics has led to a number of groups proposing sequence-based typing schemes, but no study has yet examined the relationships between VZV genotypes at a full-genome level. A central hypothesis of this study is that VZV has coevolved with humankind. In this study, 11 additional full VZV genomic sequences are presented, bringing the current number of complete genomic sequences publicly available to 18. The full-genome alignment contained strains representing four distinct clades, but the possibility exists that a fifth clade comprised of African and Asian-like isolates was not represented. A consolidated VZV genotyping scheme employing the origin-associated region between reiteration region R4 and open reading frames (ORFs) 63 and 70 is described, one which accurately categorizes strains into one of four clades related to the geographic origin of the isolates. The full-genome alignment also provided evidence for recombination having occurred between the major circulating VZV clades. One Canadian clinical isolate was primarily Asian-like in origin, with most of the genome showing strong sequence identity to the Japanese-like clade B, with the exceptions being two putative recombination regions, located in ORFs 14 to 17 and ORFs 22 to 26, which showed clear similarity to the European/North American clade A. The very low rate of single-nucleotide polymorphisms scattered across the genome made full-genome sequencing the only definitive method for identifying specific VZV recombination events.
Collapse
Affiliation(s)
- Geoffrey A Peters
- National Microbiology Laboratory, 1015 Arlington Street, Winnipeg, Manitoba R3E 3R2, Canada
| | | | | | | | | | | | | |
Collapse
|
25
|
Zeng F, Hon CC, Yip CW, Law KM, Yeung YS, Chan KH, Malik Peiris JS, Leung FCC. Quantitative comparison of the efficiency of antibodies against S1 and S2 subunit of SARS coronavirus spike protein in virus neutralization and blocking of receptor binding: implications for the functional roles of S2 subunit. FEBS Lett 2006; 580:5612-20. [PMID: 16989815 PMCID: PMC7094555 DOI: 10.1016/j.febslet.2006.08.085] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2006] [Revised: 08/17/2006] [Accepted: 08/29/2006] [Indexed: 11/18/2022]
Abstract
Neutralizing effects of antibodies targeting the C‐terminal stalk (S2) subunit of the spike protein of severe acute respiratory syndrome coronavirus have previously been reported, although its mechanism remained elusive. In this study, high titered mouse antisera against the N‐terminal globular (S1) and S2 subunits of the S protein were generated and total immunoglobulin G (IgG) was purified from these antisera. The efficiency of these purified IgGs in virus neutralization and blocking of receptor binding were compared quantitatively using virus neutralization assay and a previously developed cell‐based receptor binding assay, respectively. We demonstrated that anti‐S1 IgG neutralizes the virus and binds to the membrane associated S protein more efficiently than anti‐S2 IgG does. Moreover, both anti‐S1 and anti‐S2 IgGs were able to abolish the binding between S protein and its cellular receptor(s), although anti‐S1 IgG showed a significantly higher blocking efficiency. The unexpected blocking ability of anti‐S2 IgG towards the receptor binding implied a possible role of the S2 subunit in virus docking process and argues against the current hypothesis of viral entry. On the other hand, the functional roles of the previously reported neutralizing epitopes within S2 subunit were investigated using an antigen specific antibody depletion assay. Depletion of antibodies against these regions significantly diminished, though not completely abolished, the neutralizing effects of anti‐S2 IgG. It suggests the absence of a major neutralizing domain on S2 protein. The possible ways of anti‐S2 IgGs to abolish the receptor binding and the factors restricting anti‐S2 IgGs to neutralize the virus are discussed.
Collapse
Affiliation(s)
- Fanya Zeng
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Chung Chau Hon
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Chi Wai Yip
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Ka Man Law
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Yin Shan Yeung
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Kwok Hung Chan
- Department of Microbiology, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Joseph S. Malik Peiris
- Department of Microbiology, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| | - Frederick Chi Ching Leung
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong, China
| |
Collapse
|
26
|
Zhang L, Zhang F, Yu W, He T, Yu J, Yi CE, Ba L, Li W, Farzan M, Chen Z, Yuen KY, Ho D. Antibody responses against SARS coronavirus are correlated with disease outcome of infected individuals. J Med Virol 2006; 78:1-8. [PMID: 16299724 PMCID: PMC7166884 DOI: 10.1002/jmv.20499] [Citation(s) in RCA: 148] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Most of the SARS‐CoV‐infected patients spontaneously recovered without clinical intervention while a small percentage succumbed to the disease. Here, we characterized temporal changes in N protein‐specific and S glycoprotein‐specific neutralizing antibody (Nab) responses in infected patients who have either recovered from or succumbed to SARS‐CoV infection. Recovered patients were found to have higher and sustainable levels of both N protein‐specific and S glycoprotein‐specific Nab responses, suggesting that antibody responses likely play an important role in determining the ultimate disease outcome of SARS‐CoV‐infected patients. J. Med. Virol. 78:1–8, 2006. © 2005 Wiley‐Liss, inc.
Collapse
Affiliation(s)
- Linqi Zhang
- The Aaron Diamond AIDS Research Center, The Rockefeller University, New York, New York 10016, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Wang M, Yan M, Xu H, Liang W, Kan B, Zheng B, Chen H, Zheng H, Xu Y, Zhang E, Wang H, Ye J, Li G, Li M, Cui Z, Liu YF, Guo RT, Liu XN, Zhan LH, Zhou DH, Zhao A, Hai R, Yu D, Guan Y, Xu J. SARS-CoV infection in a restaurant from palm civet. Emerg Infect Dis 2006; 11:1860-5. [PMID: 16485471 PMCID: PMC3367621 DOI: 10.3201/eid1112.041293] [Citation(s) in RCA: 205] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Epidemiologic investigations showed that 2 of 4 patients with severe acute respiratory syndrome (SARS) identified in the winter of 2003-2004 were a waitresss at a restaurant in Guangzhou, China, that served palm civets as food and a customer who ate in the restaurant ashort distance from animal cages. All 6 palm civets at the restaurant were positive for SARS-associated coronavirus (SARS-CoV). Partial spike (S) gene sequences of SARS-CoV from the 2 patients were identical to 4 of 5 Sgene viral sequences from palm civets. Phylogenetic analysis showed that SARS-CoV from palm civets in the restaurant was most closely related to animal isolates. SARS cases at the restaurant were the result of recent interspecies transfer from the putative palm civet reservoir, and not the result of continued circulation of SARS-CoV in the human population.
Collapse
Affiliation(s)
- Ming Wang
- Guangzhou Municipal Center for Disease Control and Prevention, Guangdong, People's Republic of China
| | - Meiying Yan
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Huifang Xu
- Guangzhou Municipal Center for Disease Control and Prevention, Guangdong, People's Republic of China
| | - Weili Liang
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Biao Kan
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Bojian Zheng
- University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Honglin Chen
- University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Han Zheng
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Yanmei Xu
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Enmin Zhang
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Hongxia Wang
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Jingrong Ye
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Guichang Li
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
| | - Machao Li
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
| | - Zhigang Cui
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Yu-Fei Liu
- Guangzhou Municipal Center for Disease Control and Prevention, Guangdong, People's Republic of China
| | - Rong-Tong Guo
- Guangzhou Municipal Center for Disease Control and Prevention, Guangdong, People's Republic of China
| | - Xiao-Ning Liu
- Guangzhou Municipal Center for Disease Control and Prevention, Guangdong, People's Republic of China
| | - Liu-Hua Zhan
- Guangzhou Municipal Center for Disease Control and Prevention, Guangdong, People's Republic of China
| | - Duan-Hua Zhou
- Guangzhou Municipal Center for Disease Control and Prevention, Guangdong, People's Republic of China
| | - Ailan Zhao
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Rong Hai
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Dongzhen Yu
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| | - Yi Guan
- University of Hong Kong, Hong Kong Special Administrative Region, People's Republic of China
| | - Jianguo Xu
- National Institute for Communicable Disease Control and Prevention, Beijing, People's Republic of China
- State Key Laboratory of Infectious Diseases Prevention and Control, Bejing, People's Republic of China
| |
Collapse
|
28
|
Tyler SD, Severini A. The complete genome sequence of herpesvirus papio 2 (Cercopithecine herpesvirus 16) shows evidence of recombination events among various progenitor herpesviruses. J Virol 2006; 80:1214-21. [PMID: 16414998 PMCID: PMC1346941 DOI: 10.1128/jvi.80.3.1214-1221.2006] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
We have sequenced the entire genome of herpesvirus papio 2 (HVP-2; Cercopithecine herpesvirus 16) strain X313, a baboon herpesvirus with close homology to other primate alphaherpesviruses, such as SA8, monkey B virus, and herpes simplex virus (HSV) type 1 and type 2. The genome of HVP-2 is 156,487 bp in length, with an overall GC content of 76.5%. The genome organization is identical to that of the other members of the genus Simplexvirus, with a long and a short unique region, each bordered by inverted repeats which end with an "a" sequence. All of the open reading frames detected in this genome were homologous and colinear with those of SA8 and B virus. The HSV gene RL1 (gamma(1)34.5; neurovirulence factor) is not present in HVP-2, as is the case for SA8 and B virus. The HVP-2 genome is 85% homologous to its closest relative, SA8. However, segment-by-segment bootstrap analysis of the genome revealed at least two regions that display closer homology to the corresponding sequences of B virus. The first region comprises the UL41 to UL44 genes, and the second region is located within the UL36 gene. We hypothesize that this localized and defined shift in homology is due to recombination events between an SA8-like progenitor of HVP-2 and a herpesvirus species more closely related to the B virus. Since some of the genes involved in these putative recombination events are determinants of virulence, a comparative analysis of their function may provide insight into the pathogenic mechanism of simplexviruses.
Collapse
Affiliation(s)
- Shaun D Tyler
- National Microbiology Laboratory, Canadian Science Centre for Human and Animal Health, 1015 Arlington Street, Winnipeg, Manitoba R3E 3R2, Canada
| | | |
Collapse
|
29
|
Nicholls JM, Butany J, Poon LLM, Chan KH, Beh SL, Poutanen S, Peiris JSM, Wong M. Time course and cellular localization of SARS-CoV nucleoprotein and RNA in lungs from fatal cases of SARS. PLoS Med 2006; 3:e27. [PMID: 16379499 PMCID: PMC1324951 DOI: 10.1371/journal.pmed.0030027] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2005] [Accepted: 10/24/2005] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Cellular localization of severe acute respiratory syndrome coronavirus (SARS-CoV) in the lungs of patients with SARS is important in confirming the etiological association of the virus with disease as well as in understanding the pathogenesis of the disease. To our knowledge, there have been no comprehensive studies investigating viral infection at the cellular level in humans. METHODS AND FINDINGS We collected the largest series of fatal cases of SARS with autopsy material to date by merging the pathological material from two regions involved in the 2003 worldwide SARS outbreak in Hong Kong, China, and Toronto, Canada. We developed a monoclonal antibody against the SARS-CoV nucleoprotein and used it together with in situ hybridization (ISH) to analyze the autopsy lung tissues of 32 patients with SARS from Hong Kong and Toronto. We compared the results of these assays with the pulmonary pathologies and the clinical course of illness for each patient. SARS-CoV nucleoprotein and RNA were detected by immunohistochemistry and ISH, respectively, primarily in alveolar pneumocytes and, less frequently, in macrophages. Such localization was detected in four of the seven patients who died within two weeks of illness onset, and in none of the 25 patients who died later than two weeks after symptom onset. CONCLUSIONS The pulmonary alveolar epithelium is the chief target of SARS-CoV, with macrophages infected subsequently. Viral replication appears to be limited to the first two weeks after symptom onset, with little evidence of continued widespread replication after this period. If antiviral therapy is considered for future treatment, it should be focused on this two-week period of acute clinical disease.
Collapse
Affiliation(s)
- John M Nicholls
- 1Department of Pathology, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Jagdish Butany
- 2Department of Pathology, University of Toronto, Toronto General Hospital, Toronto, Ontario, Canada
| | - Leo L. M Poon
- 3Department of Microbiology, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Kwok H Chan
- 3Department of Microbiology, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Swan Lip Beh
- 1Department of Pathology, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Susan Poutanen
- 4Department of Microbiology, Toronto Medical Laboratories and Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada
| | - J. S. Malik Peiris
- 3Department of Microbiology, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| | - Maria Wong
- 1Department of Pathology, The University of Hong Kong, Pok Fu Lam, Hong Kong SAR, China
| |
Collapse
|
30
|
Chow KY, Yeung YS, Hon CC, Zeng F, Law KM, Leung FC. Adenovirus-mediated expression of the C-terminal domain of SARS-CoV spike protein is sufficient to induce apoptosis in Vero E6 cells. FEBS Lett 2005; 579:6699-704. [PMID: 16310778 PMCID: PMC7094440 DOI: 10.1016/j.febslet.2005.10.065] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2005] [Revised: 10/18/2005] [Accepted: 10/25/2005] [Indexed: 02/08/2023]
Abstract
The pro-apoptotic properties of severe acute respiratory syndrome coronavirus (SARS-CoV) structural proteins were studied in vitro. By monitoring apoptosis indicators including chromatin condensation, cellular DNA fragmentation and cell membrane asymmetry, we demonstrated that the adenovirus-mediated over-expression of SARS-CoV spike (S) protein and its C-terminal domain (S2) induce apoptosis in Vero E6 cells in a time- and dosage-dependent manner, whereas the expression of its N-terminal domain (S1) and other structural proteins, including envelope (E), membrane (M) and nucleocapsid (N) protein do not. These findings suggest a possible role of S and S2 protein in SARS-CoV induced apoptosis and the molecular pathogenesis of SARS.
Collapse
Affiliation(s)
- Ken Y.C. Chow
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Yin Shan Yeung
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Chung Chau Hon
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Fanya Zeng
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Ka Man Law
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| | - Frederick C.C. Leung
- Department of Zoology, Kadoorie Biological Science Building, The University of Hong Kong, Pokfulam Road, Hong Kong SAR, China
| |
Collapse
|
31
|
Cheng M, Chan CWL, Cheung RCF, Bikkavilli RK, Zhao Q, Au SWN, Chan PKS, Lee SST, Cheng G, Ho WKK, Cheung WT. Cross-reactivity of antibody against SARS-coronavirus nucleocapsid protein with IL-11. Biochem Biophys Res Commun 2005; 338:1654-60. [PMID: 16263078 PMCID: PMC7092895 DOI: 10.1016/j.bbrc.2005.10.088] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Accepted: 10/17/2005] [Indexed: 01/08/2023]
Abstract
Infection of SARS-associated coronavirus (SARS-CoV) induced a strong anti-nucleocapsid (anti-N) antibody response. However, the pathophysiological significance of the anti-N antibodies in SARS pathogenesis is largely unknown. To profile the anti-N antibodies, a phage-displayed scFv library was prepared from mice immunized with heat-inactivated SARS-CoV-infected Vero E6 cell lysate. Specific anti-N scFvs were isolated by panning against a recombinant nucleocapsid protein and reactivity was confirmed with phage-ELISA. Sequence analysis indicated that two of the isolated anti-N scFv clones were identical and displayed a high homology with an scFv specific for interleukin 11 (IL-11), an anti-inflammatory cytokine derived from bone marrow stroma cells. In a neutralization assay, IL-11-induced STAT 3 phosphorylation in rat intestinal epithelial IEC-18 cells was completely suppressed by the anti-N scFv clone L9N01.
Collapse
Affiliation(s)
- Man Cheng
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, New Territories, Hong Kong, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kan B, Wang M, Jing H, Xu H, Jiang X, Yan M, Liang W, Zheng H, Wan K, Liu Q, Cui B, Xu Y, Zhang E, Wang H, Ye J, Li G, Li M, Cui Z, Qi X, Chen K, Du L, Gao K, Zhao YT, Zou XZ, Feng YJ, Gao YF, Hai R, Yu D, Guan Y, Xu J. Molecular evolution analysis and geographic investigation of severe acute respiratory syndrome coronavirus-like virus in palm civets at an animal market and on farms. J Virol 2005; 79:11892-900. [PMID: 16140765 PMCID: PMC1212604 DOI: 10.1128/jvi.79.18.11892-11900.2005] [Citation(s) in RCA: 252] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Massive numbers of palm civets were culled to remove sources for the reemergence of severe acute respiratory syndrome (SARS) in Guangdong Province, China, in January 2004, following SARS coronavirus detection in market animals. The virus was identified in all 91 palm civets and 15 raccoon dogs of animal market origin sampled prior to culling, but not in 1,107 palm civets later sampled at 25 farms, spread over 12 provinces, which were claimed to be the source of traded animals. Twenty-seven novel signature variation residues (SNVs) were identified on the spike gene and were analyzed for their phylogenetic relationships, based on 17 sequences obtained from animals in our study and from other published studies. Analysis indicated that the virus in palm civets at the live-animal market had evolved to infect humans. The evolutionary starting point was a prototype group consisting of three viral sequences of animal origin. Initially, seven SNV sites caused six amino acid changes, at positions 147, 228, 240, 479, 821, and 1080 of the spike protein, to generate low-pathogenicity viruses. One of these was linked to the first SARS patient in the 2003-2004 period. A further 14 SNVs caused 11 amino acid residue changes, at positions 360, 462, 472, 480, 487, 609, 613, 665, 743, 765, and 1163. The resulting high-pathogenicity groups were responsible for infections during the so-called early-phase epidemic of 2003. Finally, the remaining six SNVs caused four amino acid changes, at positions 227, 244, 344, and 778, which resulted in the group of viruses responsible for the global epidemic.
Collapse
Affiliation(s)
- Biao Kan
- State Key Laboratory for Infectious Disease Prevention and Control (China CDC), Chinese Center for Disease Control and Prevention, P.O. Box 5, Changping, Beijing 102206, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Li T, Zhang Y, Fu L, Yu C, Li X, Li Y, Zhang X, Rong Z, Wang Y, Ning H, Liang R, Chen W, Babiuk LA, Chang Z. siRNA targeting the leader sequence of SARS-CoV inhibits virus replication. Gene Ther 2005; 12:751-61. [PMID: 15772689 PMCID: PMC7091583 DOI: 10.1038/sj.gt.3302479] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
SARS-CoV (the SARS-Associated Coronavirus) was reported as a novel virus member in the coronavirus family, which was the cause of severe acute respiratory syndrome. Coronavirus replication occurs through a unique mechanism employing Leader sequence in the transcripts when initiating transcription from the genome. Therefore, we cloned the Leader sequence from SARS-CoV(BJ01), which is identical to that identified from SARS-CoV(HKU-39849), and constructed specific siRNA targeting the Leader sequence. Using EGFP and RFP reporter genes fused with the cloned SARS-CoV Leader sequence, we demonstrated that the siRNA targeting the Leader sequence decreased the mRNA abundance and protein expression levels of the reporter genes in 293T cells. By stably expressing the siRNA in Vero E6 cells, we provided data that the siRNA could effectively and specifically decrease the mRNA abundance of SARS-CoV genes as analyzed by RT-PCR and Northern blot. Our data indicated that the siRNA targeting the Leader sequence inhibited the replication of SARS-CoV in Vero E6 cells by silencing gene expression. We further demonstrated, via transient transfection experiments, that the siRNA targeting the Leader sequence had a much stronger inhibitory effect on SARS-CoV replication than the siRNAs targeting the Spike gene or the antisense oligodeoxynucleotides did. This report provides evidence that targeting Leader sequence using siRNA could be a powerful tool in inhibiting SARS-CoV replication.
Collapse
Affiliation(s)
- T Li
- Department of Biological Sciences and Biotechnology, Institute of Biomedicine, Tsinghua University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Wang X, Ni B, Du X, Zhao G, Gao W, Shi X, Zhang S, Zhang L, Wang D, Luo D, Xing L, Jiang H, Li W, Jiang M, Mao L, He Y, Xiao Y, Wu Y. Protection of Mammalian Cells from Severe Acute Respiratory Syndrome Coronavirus Infection by Equine Neutralizing Antibody. Antivir Ther 2005. [DOI: 10.1177/135965350501000504] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The aetiological agent for severe acute respiratory syndrome (SARS) has been determined to be a new type of coronavirus (SARS-CoV) that infects a wide range of mammalian hosts. Up to now, there have been no specific drugs to protect against SARS-CoV infection, thus developing effective strategies against this newly emerged viral infection warrants urgent efforts. Adoptive immune therapy with pathogen-specific heterologous immunoglobulin has been successfully used to control the dissemination of many viral infections. To investigate whether a neutralizing antibody against SARS-CoV raised in an artiodactylous host can have a protective role on primate cells, we prepared serum IgGs and their pepsin-digested F(ab’)2 fragments from horses inoculated with purified SARS-CoV (BJ-01 strain). The protective effect of the F(ab’)2 fragments against SARS-CoV infection was determined in cultured Vero E6 cells by cytopathic effect (CPE), MTT and plaque-forming assays and in a Balb/c mouse model by CPE and quantitative RT-PCR. The results showed the neutralization titres of F(ab’)2 from three horses all reached at least 1:1600, and 50 μg of the F(ab’)2 fragments could completely neutralize 1x104 TCID50 SARS-CoV in vivo. Additionally, we observed that F(ab’)2 against BJ-01 strain could also protect cells from infection by the variant GZ-01 strain in vitro and in vivo. Our work has provided experimental support for testing the protective equine immunoglobulin in future large primate or human trials.
Collapse
Affiliation(s)
- Xiliang Wang
- Department of Immunology, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Bing Ni
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Xinan Du
- Department of Immunology, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Guangyu Zhao
- Department of Immunology, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Wenda Gao
- Beth Israel Deaconess Medical Centre, Harvard Medical School, Boston, MA, USA
| | - Xinfu Shi
- Department of Immunology, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Songle Zhang
- Department of Immunology, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Liangyan Zhang
- Department of Immunology, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Dong Wang
- Department of Immunology, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Deyan Luo
- Department of Immunology, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Li Xing
- Department of Immunology, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Haiyan Jiang
- Department of Immunology, Institute of Microbiology and Epidemiology, Academy of Military Medical Sciences, Beijing, China
| | - Wanling Li
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Man Jiang
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Liwei Mao
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Yangdong He
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Yu Xiao
- Institute of Immunology, Third Military Medical University, Chongqing, China
| | - Yuzhang Wu
- Institute of Immunology, Third Military Medical University, Chongqing, China
| |
Collapse
|
35
|
Law PTW, Wong CH, Au TCC, Chuck CP, Kong SK, Chan PKS, To KF, Lo AWI, Chan JYW, Suen YK, Chan HYE, Fung KP, Waye MMY, Sung JJY, Lo YMD, Tsui SKW. The 3a protein of severe acute respiratory syndrome-associated coronavirus induces apoptosis in Vero E6 cells. J Gen Virol 2005; 86:1921-1930. [PMID: 15958670 DOI: 10.1099/vir.0.80813-0] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
An outbreak of severe acute respiratory syndrome (SARS) occurred in China and the first case emerged in mid-November 2002. The aetiological agent of this disease was found to be a previously unknown coronavirus, SARS-associated coronavirus (SARS-CoV). The detailed pathology of SARS-CoV infection and the host response to the viral infection are still not known. The 3a gene encodes a non-structural viral protein, which is predicted to be a transmembrane protein. In this study, it was shown that the 3a protein was expressed in the lungs and intestinal tissues of SARS patients and that the protein localized to the endoplasmic reticulum in 3a-transfected monkey kidney Vero E6 cells. In vitro experiments of chromatin condensation and DNA fragmentation suggested that the 3a protein may trigger apoptosis. These data showed that overexpression of a single SARS-CoV protein can induce apoptosis in vitro.
Collapse
Affiliation(s)
- Patrick T W Law
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
- Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Chi-Hang Wong
- Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Thomas C C Au
- Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Chi-Pang Chuck
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Siu-Kai Kong
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Paul K S Chan
- Department of Microbiology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Ka-Fai To
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Anthony W I Lo
- Department of Anatomical and Cellular Pathology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Judy Y W Chan
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Yick-Keung Suen
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - H Y Edwin Chan
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Kwok-Pui Fung
- The Croucher Laboratory for Human Genomics, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Mary M Y Waye
- The Croucher Laboratory for Human Genomics, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Joseph J Y Sung
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
- Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Y M Dennis Lo
- Department of Chemical Pathology, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
- Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| | - Stephen K W Tsui
- The Croucher Laboratory for Human Genomics, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
- Department of Biochemistry, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
- Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, China
| |
Collapse
|
36
|
Bacon LD, Hunter DB, Zhang HM, Brand K, Etches R. Retrospective evidence that the MHC (B haplotype) of chickens influences genetic resistance to attenuated infectious bronchitis vaccine strains in chickens. Avian Pathol 2005; 33:605-9. [PMID: 15763730 DOI: 10.1080/03079450400013147] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Infectious bronchitis is a respiratory disease of chickens that is caused by the coronavirus infectious bronchitis virus (IBV). Virtually all broiler and layer breeder flocks are routinely vaccinated against IBV. Two hatches of 1-day-old chicks from four lines were mistakenly vaccinated for infectious bronchitis using a moderately attenuated vaccine designed for chicks of an older age. The vaccination resulted in high mortality, and chicks from three of four lines died with signs typical of infectious bronchitis. The mortality that occurred using this less-attenuated vaccine was significantly influenced by the genetic line, and the MHC (B) haplotype in chickens of three B congenic lines. B congenic chickens possessing the B*15 haplotype were resistant in contrast to chickens possessing the B*13 or B*21 haplotypes. Chicks from two further hatches of the four lines were vaccinated appropriately with a more attenuated IBV vaccine, and only limited chick mortality was seen. These retrospective data from two repeated hatches confirm earlier data indicating chicken genes influence resistance to IBV, and indicate for the first time that genes tightly linked to the B haplotype are relevant in resistance to IBV. Due to extenuating circumstances it was not possible to verify results with chicks from F2 matings. Factors that may enhance definition of the role of the B haplotype in immune response to IBV, and the desirability for further analysis of a B haplotype-linked influence on immunity to IBV are discussed.
Collapse
Affiliation(s)
- L D Bacon
- Avian Disease and Oncology Laboratory, Agricultural Research Service, US Department of Agriculture, East Lansing, Michigan 48823, USA.
| | | | | | | | | |
Collapse
|
37
|
Chen Z, Zhang L, Qin C, Ba L, Yi CE, Zhang F, Wei Q, He T, Yu W, Yu J, Gao H, Tu X, Gettie A, Farzan M, Yuen KY, Ho DD. Recombinant modified vaccinia virus Ankara expressing the spike glycoprotein of severe acute respiratory syndrome coronavirus induces protective neutralizing antibodies primarily targeting the receptor binding region. J Virol 2005; 79:2678-88. [PMID: 15708987 PMCID: PMC548443 DOI: 10.1128/jvi.79.5.2678-2688.2005] [Citation(s) in RCA: 167] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Immunization with a killed or inactivated viral vaccine provides significant protection in animals against challenge with certain corresponding pathogenic coronaviruses (CoVs). However, the promise of this approach in humans is hampered by serious concerns over the risk of leaking live severe acute respiratory syndrome (SARS) viruses. In this study, we generated a SARS vaccine candidate by using the live-attenuated modified vaccinia virus Ankara (MVA) as a vector. The full-length SARS-CoV envelope Spike (S) glycoprotein gene was introduced into the deletion III region of the MVA genome. The newly generated recombinant MVA, ADS-MVA, is replication incompetent in mammalian cells and highly immunogenic in terms of inducing potent neutralizing antibodies in mice, rabbits, and monkeys. After two intramuscular vaccinations with ADS-MVA alone, the 50% inhibitory concentration in serum was achieved with reciprocal sera dilutions of more than 1,000- to 10,000-fold in these animals. Using fragmented S genes as immunogens, we also mapped a neutralizing epitope in the region of N-terminal 400 to 600 amino acids of the S glycoprotein (S400-600), which overlaps with the angiotensin-converting enzyme 2 (ACE2) receptor-binding region (RBR; S318-510). Moreover, using a recombinant soluble RBR-Fc protein, we were able to absorb and remove the majority of the neutralizing antibodies despite observing that the full S protein tends to induce a broader spectrum of neutralizing activities in comparison with fragmented S proteins. Our data suggest that a major mechanism for neutralizing SARS-CoV likely occurs through blocking the interaction between virus and the cellular receptor ACE2. In addition, ADS-MVA induced potent immune responses which very likely protected Chinese rhesus monkeys from pathogenic SARS-CoV challenge.
Collapse
Affiliation(s)
- Zhiwei Chen
- Aaron Diamond AIDS Research Center, The Rockefeller University, New York, NY 10016, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Lan YC, Liu TT, Yang JY, Lee CM, Chen YJ, Chan YJ, Lu JJ, Liu HF, Hsiung CA, Ho MS, Hsiao KJ, Chen HY, Chen YMA. Molecular epidemiology of severe acute respiratory syndrome-associated coronavirus infections in Taiwan. J Infect Dis 2005; 191:1478-89. [PMID: 15809907 PMCID: PMC7199491 DOI: 10.1086/428591] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2004] [Accepted: 11/01/2004] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND In 2003, Taiwan experienced a series of outbreaks of severe acute respiratory syndrome (SARS) and 1 laboratory-contamination accident. Here we describe a new phylogenetic analytical method to study the sources and dissemination paths of SARS-associated coronavirus (SARS-CoV) infections in Taiwan. METHODS A phylogenetic analytical tool for combining nucleotide sequences from 6 variable regions of a SARS-CoV genome was developed by use of 20 published SARS-CoV sequences; and this method was validated by use of 80 published SARS-CoV sequences. Subsequently, this new tool was applied to provide a better understanding of the entire complement of Taiwanese SARS-CoV isolates, including 20 previously published and 19 identified in this study. The epidemiological data were integrated with the results from the phylogenetic tree and from the nucleotide-signature pattern. RESULTS The topologies of phylogenetic trees generated by the new and the conventional strategies were similar, with the former having better robustness than the latter, especially in comparison with the maximum-likelihood trees: the new strategy revealed that during 2003 there were 5 waves of epidemic SARS-CoV infection, which belonged to 3 phylogenetic clusters in Taiwan. CONCLUSIONS The new strategy is more efficient than its conventional counterparts. The outbreaks of SARS in Taiwan originated from multiple sources.
Collapse
Affiliation(s)
- Yu-Ching Lan
- AIDS Prevention and Research Centre
- Institute of Public Health, and
- Reprints or correspondence: Prof. Yi-Ming A Chen, AIDS Prevention and Research Center, National Yang-Ming University, Li-Noun St., Section 2, Taipei, Taiwan 112 ()
| | - Tze-Tze Liu
- Genome Research Center, National Yang-Ming University, and
| | - Jyh-Yuan Yang
- Center for Disease Control, Department of Health, Executive Yuan, and
| | - Cheng-Ming Lee
- AIDS Prevention and Research Centre
- Institute of Public Health, and
| | - Yen-Ju Chen
- AIDS Prevention and Research Centre
- Institute of Public Health, and
| | - Yu-Jiun Chan
- AIDS Prevention and Research Centre
- Section of Virology, Department of Laboratory Medicine, Taipei Veterans General Hospital, and
| | - Jang-Jih Lu
- Department of Pathology, Tri-Service General Hospital, National Defense Medical Center, and
| | - Hsin-Fu Liu
- Department of Medical Research, Taipei Mackay Memorial Hospital, and
| | | | - Mei-Shang Ho
- Institute of Biomedical Sciences, Academia Sinica, Taiwan, Republic of China
| | | | - Hour-Young Chen
- Center for Disease Control, Department of Health, Executive Yuan, and
| | | |
Collapse
|
39
|
Kao RY, Tsui WHW, Lee TSW, Tanner JA, Watt RM, Huang JD, Hu L, Chen G, Chen Z, Zhang L, He T, Chan KH, Tse H, To APC, Ng LWY, Wong BCW, Tsoi HW, Yang D, Ho DD, Yuen KY. Identification of novel small-molecule inhibitors of severe acute respiratory syndrome-associated coronavirus by chemical genetics. ACTA ACUST UNITED AC 2005; 11:1293-9. [PMID: 15380189 PMCID: PMC7128553 DOI: 10.1016/j.chembiol.2004.07.013] [Citation(s) in RCA: 119] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2004] [Revised: 07/06/2004] [Accepted: 07/08/2004] [Indexed: 11/17/2022]
Abstract
The severe acute respiratory syndrome-associated coronavirus (SARS-CoV) infected more than 8,000 people across 29 countries and caused more than 900 fatalities. Based on the concept of chemical genetics, we screened 50,240 structurally diverse small molecules from which we identified 104 compounds with anti-SARS-CoV activity. Of these 104 compounds, 2 target the SARS-CoV main protease (Mpro), 7 target helicase (Hel), and 18 target spike (S) protein-angiotensin-converting enzyme 2 (ACE2)-mediated viral entry. The EC50 of the majority of the 104 compounds determined by SARS-CoV plaque reduction assay were found to be at low micromolar range. Three selected compounds, MP576, HE602, and VE607, validated to be inhibitors of SARS-CoV Mpro, Hel, and viral entry, respectively, exhibited potent antiviral activity (EC50 < 10 μM) and comparable inhibitory activities in target-specific in vitro assays.
Collapse
Affiliation(s)
- Richard Y Kao
- Department of Microbiology, The University of Hong Kong, Pokfulam, Hong Kong SAR, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Poon LLM, Guan Y, Nicholls JM, Yuen KY, Peiris JSM. The aetiology, origins, and diagnosis of severe acute respiratory syndrome. THE LANCET. INFECTIOUS DISEASES 2004; 4:663-71. [PMID: 15522678 PMCID: PMC7106534 DOI: 10.1016/s1473-3099(04)01172-7] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Severe acute respiratory syndrome (SARS) is a new infectious disease that first emerged in Guangdong province, China, in November, 2002. A novel coronavirus was later identified in patients with SARS. The detection of the virus in these patients, its absence in healthy controls or other patients with atypical pneumonia, and the reproduction of a similar disease in a relevant animal model fulfilled Koch's postulates for implicating this coronavirus as the causal agent of SARS. The full genome sequence was determined within weeks of the virus's identification. The rapid progress in the aetiology, the development of laboratory diagnostic tests, and the defining of routes of viral transmission were facilitated through a unique WHO-coordinated virtual network of laboratories, which shared information on a real-time basis through daily teleconferences. Subsequent studies have indicated that the SARS coronavirus is of animal origin, that its precursor is still present in animal populations within the region, and that live-animal markets in southern China may have provided the animal-human interphase that allowed this precursor virus to adapt to human-human transmission. These findings underscore the potential for the re-emergence of SARS and the need for laboratory tests for early diagnosis. However, the low viral load in the respiratory tract makes early diagnosis of SARS a diagnostic challenge, although improvements in the sensitivity of molecular diagnostic methods continue to be made.
Collapse
Affiliation(s)
- LLM Poon
- Department of Microbiology, University of Hong Kong, Hong Kong SAR, China
| | - Y Guan
- Department of Microbiology, University of Hong Kong, Hong Kong SAR, China
| | - JM Nicholls
- Department of Pathology, University of Hong Kong, Hong Kong SAR, China
| | - KY Yuen
- Department of Microbiology, University of Hong Kong, Hong Kong SAR, China
| | - JSM Peiris
- Department of Microbiology, University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
41
|
Visintin M, Meli GA, Cannistraci I, Cattaneo A. Intracellular antibodies for proteomics. J Immunol Methods 2004; 290:135-53. [PMID: 15261577 PMCID: PMC7126613 DOI: 10.1016/j.jim.2004.04.014] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/08/2004] [Indexed: 11/03/2022]
Abstract
The intracellular antibody technology has many applications for proteomics studies. The potential of intracellular antibodies for the systematic study of the proteome has been made possible by the development of new experimental strategies that allow the selection of antibodies under conditions of intracellular expression. The Intracellular Antibody Capture Technology (IACT) is an in vivo two-hybrid-based method originally developed for the selection of antibodies readily folded for ectopic expression. IACT has been used for the rapid and effective identification of novel antigen-antibody pairs in intracellular compartments and for the in vivo identification of epitopes recognized by selected intracellular antibodies. IACT opens the way to the use of intracellular antibody technology for large-scale applications in proteomics. In its present format, its use is however somewhat limited by the need of a preselection of the input phage antibody libraries on protein antigens or by the construction of an antibody library from mice immunized against the target protein(s), to provide an enriched input library to compensate for the suboptimal efficiency of transformation of the yeast cells. These enrichment steps require expressing the corresponding proteins, which represents a severe bottleneck for the scaling up of the technology. We describe here the construction of a single pot library of intracellular antibodies (SPLINT), a naïve library of scFv fragments expressed directly in the yeast cytoplasm in a format such that antigen-specific intrabodies can be isolated directly from gene sequences, with no manipulation whatsoever of the corresponding proteins. We describe also the isolation from SPLINT of a panel of intrabodies against a number of different proteins. The application of SPLINT on a genome-wide scale should help the systematic study of the functional organization of cell proteome.
Collapse
Affiliation(s)
- Michela Visintin
- Lay Line Genomics SpA, Via di Castel Romano 100, 00128 Rome, Italy
| | - Giovanni Antonio Meli
- International School for Advanced Studies (SISSA), Padriciano 99, 34012 Trieste, Italy
| | | | - Antonino Cattaneo
- Lay Line Genomics SpA, Via di Castel Romano 100, 00128 Rome, Italy
- International School for Advanced Studies (SISSA), Padriciano 99, 34012 Trieste, Italy
- Corresponding author. Lay Line Genomics SpA, Via di Castel Romano 100, 00128 Rome, Italy
| |
Collapse
|
42
|
Bukreyev A, Lamirande EW, Buchholz UJ, Vogel LN, Elkins WR, St Claire M, Murphy BR, Subbarao K, Collins PL. Mucosal immunisation of African green monkeys (Cercopithecus aethiops) with an attenuated parainfluenza virus expressing the SARS coronavirus spike protein for the prevention of SARS. Lancet 2004; 363:2122-7. [PMID: 15220033 PMCID: PMC7112367 DOI: 10.1016/s0140-6736(04)16501-x] [Citation(s) in RCA: 206] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND The outbreak of severe acute respiratory syndrome (SARS) in 2002 was caused by a previously unknown coronavirus-SARS coronavirus (SARS-CoV). We have developed an experimental SARS vaccine for direct immunisation of the respiratory tract, the major site of SARS- coronavirus transmission and disease. METHODS We expressed the complete SARS coronavirus envelope spike (S) protein from a recombinant attenuated parainfluenza virus (BHPIV3) that is being developed as a live attenuated, intranasal paediatric vaccine against human parainfluenza virus type 3 (HPIV3). We immunised eight African green monkeys, four with a single dose of BHPIV3/ SARS-S and four with a control, BHPIV3/Ctrl, administered via the respiratory tract. A SARS-coronavirus challenge was given to all monkeys 28 days after immunisation. FINDINGS Immunisation of animals with BHPIV3/SARS-S induced the production of SARS-coronavirus-neutralising serum antibodies, indicating that a systemic immune response resulted from mucosal immunisation. After challenge with SARS coronavirus, all monkeys in the control group shed SARS coronavirus, with shedding lasting 5-8 days. No viral shedding occurred in the group immunised with BHPIV3/SARS-S. INTERPRETATION A vectored mucosal vaccine expressing the SARS-coronavirus S protein alone may be highly effective in a single-dose format for the prevention of SARS.
Collapse
Affiliation(s)
- Alexander Bukreyev
- Laboratory of Infectious Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Pavlović-Lažetić GM, Mitić NS, Beljanski MV. Bioinformatics analysis of SARS coronavirus genome polymorphism. BMC Bioinformatics 2004; 5:65. [PMID: 15161495 PMCID: PMC434493 DOI: 10.1186/1471-2105-5-65] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2003] [Accepted: 05/25/2004] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND We have compared 38 isolates of the SARS-CoV complete genome. The main goal was twofold: first, to analyze and compare nucleotide sequences and to identify positions of single nucleotide polymorphism (SNP), insertions and deletions, and second, to group them according to sequence similarity, eventually pointing to phylogeny of SARS-CoV isolates. The comparison is based on genome polymorphism such as insertions or deletions and the number and positions of SNPs. RESULTS The nucleotide structure of all 38 isolates is presented. Based on insertions and deletions and dissimilarity due to SNPs, the dataset of all the isolates has been qualitatively classified into three groups each having their own subgroups. These are the A-group with "regular" isolates (no insertions / deletions except for 5' and 3' ends), the B-group of isolates with "long insertions", and the C-group of isolates with "many individual" insertions and deletions. The isolate with the smallest average number of SNPs, compared to other isolates, has been identified (TWH). The density distribution of SNPs, insertions and deletions for each group or subgroup, as well as cumulatively for all the isolates is also presented, along with the gene map for TWH. Since individual SNPs may have occurred at random, positions corresponding to multiple SNPs (occurring in two or more isolates) are identified and presented. This result revises some previous results of a similar type. Amino acid changes caused by multiple SNPs are also identified (for the annotated sequences, as well as presupposed amino acid changes for non-annotated ones). Exact SNP positions for the isolates in each group or subgroup are presented. Finally, a phylogenetic tree for the SARS-CoV isolates has been produced using the CLUSTALW program, showing high compatibility with former qualitative classification. CONCLUSIONS The comparative study of SARS-CoV isolates provides essential information for genome polymorphism, indication of strain differences and variants evolution. It may help with the development of effective treatment.
Collapse
Affiliation(s)
- Gordana M Pavlović-Lažetić
- Faculty of Mathematics, University of Belgrade, P.O.B. 550, Studentski trg 16, 11001 Belgrade, Serbia and Montenegro
| | - Nenad S Mitić
- Faculty of Mathematics, University of Belgrade, P.O.B. 550, Studentski trg 16, 11001 Belgrade, Serbia and Montenegro
| | - Miloš V Beljanski
- Institute of General and Physical Chemistry, P.O.B. 551, Studentski trg 16, 11001 Belgrade, Serbia and Montenegro
| |
Collapse
|
44
|
Hui RKH, Zeng F, Chan CMN, Yuen KY, Peiris JSM, Leung FCC. Reverse transcriptase PCR diagnostic assay for the coronavirus associated with severe acute respiratory syndrome. J Clin Microbiol 2004; 42:1994-9. [PMID: 15131160 PMCID: PMC404607 DOI: 10.1128/jcm.42.5.1994-1999.2004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2003] [Revised: 10/04/2003] [Accepted: 12/26/2003] [Indexed: 01/15/2023] Open
Abstract
Recent outbreaks of severe acute respiratory syndrome (SARS) have spurred intense research efforts around the world to deal with the serious threat to health posed by this novel coronavirus. A rapid, reliable diagnostic assay is needed for monitoring the spread of the disease. Here we report a method for eliminating false-negative results and increasing test sensitivity, based on the hypothesis that the message encoded by the nucleocapsid (N) gene is the most abundant during viral infection. Nasopharyngeal aspirates and stool samples were obtained from suspected SARS patients with major clinical symptoms and a significant history of close contact with infected patients. Total RNAs were extracted in a 96-well format, together with pig kidney epithelial (PK-15) cells as an internal control for extraction efficiency. PCR inhibitors were removed by ethanol precipitation, and a PCR for the pig beta-actin gene was used as a positive control for all clinical samples. Samples were analyzed by a reverse transcriptase PCR assay. Northern blot analysis was performed to demonstrate differences in subgenomic transcripts of the virus, and a real-time quantitative PCR was employed to compare the sensitivities of two loci (1b and N). The detection rate of the assay reached 44.4% on day 9 after the onset of the disease. The diagnostic PCR amplifying the N gene gave an average of a 26.0% (6.3 to 60.0%) stronger intensity signal than that for the 1b gene. In conclusion, the nucleocapsid gene represents an additional sensitive molecular marker for the diagnosis of the SARS coronavirus and can be further adapted for use in a high-throughput platform assay.
Collapse
Affiliation(s)
- Raymond K H Hui
- Department of Zoology, The University of Hong Kong, Hong Kong, Special Administrative Region, People's Republic of China
| | | | | | | | | | | |
Collapse
|
45
|
Stadler K, Masignani V, Eickmann M, Becker S, Abrignani S, Klenk HD, Rappuoli R. SARS--beginning to understand a new virus. Nat Rev Microbiol 2004; 1:209-18. [PMID: 15035025 PMCID: PMC7097337 DOI: 10.1038/nrmicro775] [Citation(s) in RCA: 378] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A new infectious disease, called severe acute respiratory syndrome (SARS), appeared in southern China in 2002. During the period from November 2002 to the summer of 2003, the World Health Organization recorded 8098 probable SARS cases and 774 deaths in 29 countries. A previously unknown coronavirus was isolated from FRhK-4 and Vero E6 cells inoculated with clinical specimens from patients. A virus with close homology to SARS-CoV was isolated from palm civets and racoon dogs, which are used as food in southern China In less than a month from the first indication that a coronavirus might be implicated in the disease, the nucleotide sequence of the virus was available, and diagnostic tests were set up. The phylogenetic analysis of the SARS-CoV genome revealed that the virus is distinct from the three known groups of coronaviruses and represents an early split-off from group 2. The development of antiviral drugs or vaccines is being investigated. Viral enzymes essential for virus replication, such as the RNA-dependent RNA polymerase (RdRp), the 3C-like cystein protease (3Clpro) and the helicases are the most attractive targets for antiviral molecules. Of the possible vaccine targets, the spike (S) protein represents the most promising one. So far, β-interferon is the only licensed drug available, which has been reported to interfere with virus replication in vitro. Should SARS return during the next winter, we will still need to rely mostly on quarantine measures to contain it.
The 114-day epidemic of the severe acute respiratory syndrome (SARS) swept 29 countries, affected a reported 8,098 people, left 774 patients dead and almost paralysed the Asian economy. Aggressive quarantine measures, possibly aided by rising summer temperatures, successfully terminated the first eruption of SARS and provided at least a temporal break, which allows us to consolidate what we have learned so far and plan for the future. Here, we review the genomics of the SARS coronavirus (SARS-CoV), its phylogeny, antigenic structure, immune response and potential therapeutic interventions should the SARS epidemic flare up again.
Collapse
Affiliation(s)
- Konrad Stadler
- IRIS, Chiron S.r.l., Via Fiorentina 1, Siena, 53100 Italy
| | - Vega Masignani
- IRIS, Chiron S.r.l., Via Fiorentina 1, Siena, 53100 Italy
| | - Markus Eickmann
- Institute of Virology, University of Marburg, Marburg, 35037 Germany
| | - Stephan Becker
- Institute of Virology, University of Marburg, Marburg, 35037 Germany
| | | | - Hans-Dieter Klenk
- Institute of Virology, University of Marburg, Marburg, 35037 Germany
| | - Rino Rappuoli
- IRIS, Chiron S.r.l., Via Fiorentina 1, Siena, 53100 Italy
| |
Collapse
|
46
|
Abstract
Respiratory viral infections, also known as the common cold, are the most common infections in humans. Despite their benign nature, they are a major cause of morbidity and mortality on a worldwide basis. Several viruses have been associated with such illness, of which rhinovirus is the most common. Symptom production is a combination of viral cytopathic effect and the activation of inflammatory pathways. Therefore, antiviral treatment alone may not be able to prevent these events. The optimal use of such agents also requires earlier initiation; therefore, it is important to develop accurate and rapid diagnostic techniques for respiratory viruses. Before any reliable and effective treatment is available, symptomatic therapies may remain the only possible choice of management.
Collapse
Affiliation(s)
- Dennis Wat
- Cystic Fibrosis Unit, Department of Child Health, University Hospital of Wales, Heath Park, CF14 4XW, UK
| |
Collapse
|
47
|
Zeng F, Chow KYC, Hon CC, Law KM, Yip CW, Chan KH, Peiris JS, Leung FCC. Characterization of humoral responses in mice immunized with plasmid DNAs encoding SARS-CoV spike gene fragments. Biochem Biophys Res Commun 2004; 315:1134-9. [PMID: 14985131 PMCID: PMC7111006 DOI: 10.1016/j.bbrc.2004.01.166] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2004] [Indexed: 01/01/2023]
Abstract
The immunological characteristics of SARS-CoV spike protein were investigated by administering mice with plasmids encoding various S gene fragments. We showed that the secreting forms of S1, S2 subunits and the N-terminus of S1 subunit (residues 18-495) were capable of eliciting SARS-CoV specific antibodies and the region immediate to N-terminus of matured S1 protein contained an important immunogenic determinant for elicitation of SARS-CoV specific antibodies. In addition, mice immunized with plasmids encoding S1 fragment developed a Th1-mediated antibody isotype switching. Another interesting finding was that mouse antibodies elicited separately by plasmids encoding S1 and S2 subunits cooperatively neutralized SARS-CoV but neither the S1 nor S2 specific antibodies did, suggesting the possible role of both S1 and S2 subunits in host cell docking and entry. These results provide insights into understanding the immunological characteristics of spike protein and the development of subunit vaccines against SARS-CoV.
Collapse
Affiliation(s)
- Fanya Zeng
- Department of Zoology, The University of Hong Kong, Pokfulam Rd, Hong Kong, SAR, China
| | - Ken Yan Ching Chow
- Department of Zoology, The University of Hong Kong, Pokfulam Rd, Hong Kong, SAR, China
| | - Chung Chau Hon
- Department of Zoology, The University of Hong Kong, Pokfulam Rd, Hong Kong, SAR, China
| | - Ka Man Law
- Department of Zoology, The University of Hong Kong, Pokfulam Rd, Hong Kong, SAR, China
| | - Chi Wai Yip
- Department of Zoology, The University of Hong Kong, Pokfulam Rd, Hong Kong, SAR, China
| | - Kwok Hung Chan
- Department of Microbiology, The University of Hong Kong, Pokfulam Rd, Hong Kong, SAR, China
| | - Joseph S.Malik Peiris
- Department of Microbiology, The University of Hong Kong, Pokfulam Rd, Hong Kong, SAR, China
| | | |
Collapse
|
48
|
Sutton G, Fry E, Carter L, Sainsbury S, Walter T, Nettleship J, Berrow N, Owens R, Gilbert R, Davidson A, Siddell S, Poon LL, Diprose J, Alderton D, Walsh M, Grimes JM, Stuart DI. The nsp9 replicase protein of SARS-coronavirus, structure and functional insights. Structure 2004; 12:341-53. [PMID: 14962394 PMCID: PMC7135010 DOI: 10.1016/j.str.2004.01.016] [Citation(s) in RCA: 183] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2003] [Revised: 12/30/2003] [Accepted: 01/06/2004] [Indexed: 02/04/2023]
Abstract
As part of a high-throughput structural analysis of SARS-coronavirus (SARS-CoV) proteins, we have solved the structure of the non-structural protein 9 (nsp9). This protein, encoded by ORF1a, has no designated function but is most likely involved with viral RNA synthesis. The protein comprises a single beta-barrel with a fold previously unseen in single domain proteins. The fold superficially resembles an OB-fold with a C-terminal extension and is related to both of the two subdomains of the SARS-CoV 3C-like protease (which belongs to the serine protease superfamily). nsp9 has, presumably, evolved from a protease. The crystal structure suggests that the protein is dimeric. This is confirmed by analytical ultracentrifugation and dynamic light scattering. We show that nsp9 binds RNA and interacts with nsp8, activities that may be essential for its function(s).
Collapse
Affiliation(s)
- Geoff Sutton
- Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Elizabeth Fry
- Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Lester Carter
- Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
- Oxford Protein Production Facility, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Sarah Sainsbury
- Oxford Protein Production Facility, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Tom Walter
- Oxford Protein Production Facility, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Joanne Nettleship
- Oxford Protein Production Facility, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Nick Berrow
- Oxford Protein Production Facility, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Ray Owens
- Oxford Protein Production Facility, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Robert Gilbert
- Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Andrew Davidson
- Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Stuart Siddell
- Department of Pathology and Microbiology, School of Medical Sciences, University of Bristol, University Walk, Bristol BS8 1TD, United Kingdom
| | - Leo L.M. Poon
- Department of Microbiology, The University of Hong Kong, Queen Mary Hospital, Pokfulam Road, SAR ROC, Hong Kong
| | - Jonathan Diprose
- Oxford Protein Production Facility, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - David Alderton
- Oxford Protein Production Facility, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - Martin Walsh
- CRG BM14, ESRF, B.P.220, F-38043 Grenoble CEDEX, France
| | - Jonathan M. Grimes
- Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
- Oxford Protein Production Facility, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| | - David I. Stuart
- Division of Structural Biology, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
- Oxford Protein Production Facility, The Henry Wellcome Building for Genomic Medicine, Oxford University, Roosevelt Drive, Oxford OX3 7BN, United Kingdom
| |
Collapse
|
49
|
Guan Y, Peiris JSM, Zheng B, Poon LLM, Chan KH, Zeng FY, Chan CWM, Chan MN, Chen JD, Chow KYC, Hon CC, Hui KH, Li J, Li VYY, Wang Y, Leung SW, Yuen KY, Leung FC. Molecular epidemiology of the novel coronavirus that causes severe acute respiratory syndrome. Lancet 2004; 363:99-104. [PMID: 14726162 PMCID: PMC7112497 DOI: 10.1016/s0140-6736(03)15259-2] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
BACKGROUND Severe acute respiratory syndrome (SARS) is a newly emerged disease caused by a novel coronavirus (SARS-CoV), which spread globally in early 2003, affecting over 30 countries. We have used molecular epidemiology to define the patterns of spread of the virus in Hong Kong and beyond. METHODS The case definition of SARS was based on that recommended by WHO. We genetically sequenced the gene for the S1 unit of the viral spike protein of viruses from patients with SARS in Hong Kong (138) and Guangdong (three) in February to April, 2003. We undertook phylogenetic comparisons with 27 other sequences available from public databases (Genbank). FINDINGS Most of the Hong Kong viruses (139/142), including those from a large outbreak in an apartment block, clustered closely together with the isolate from a single index case (HKU-33) who came from Guangdong to Hong Kong in late February. Three other isolates were genetically distinct from HKU-33 in Hong Kong during February, but none of these contributed substantially to the subsequent local outbreak. Viruses identified in Guangdong and Beijing were genetically more diverse. INTERPRETATION The molecular epidemiological evidence suggests that most SARS-CoV from the outbreak in Hong Kong, as well as the viruses from Canada, Vietnam, and Singapore, are genetically closely linked. Three viruses found in Hong Kong in February were phylogenetically distinct from the major cluster, which suggests that several introductions of the virus had occurred, but that only one was associated with the subsequent outbreak in Hong Kong, which in turn spread globally.
Collapse
Affiliation(s)
- Y Guan
- Department of Microbiology, University of Hong Kong, Hong Kong SAR, China
| | - JSM Peiris
- Department of Microbiology, University of Hong Kong, Hong Kong SAR, China
| | - B Zheng
- Department of Microbiology, University of Hong Kong, Hong Kong SAR, China
| | - LLM Poon
- Department of Microbiology, University of Hong Kong, Hong Kong SAR, China
| | - KH Chan
- Department of Microbiology, University of Hong Kong, Hong Kong SAR, China
| | - FY Zeng
- Department of Zoology, University of Hong Kong, Hong Kong SAR, China
| | - CWM Chan
- Department of Zoology, University of Hong Kong, Hong Kong SAR, China
| | - MN Chan
- Department of Zoology, University of Hong Kong, Hong Kong SAR, China
| | - JD Chen
- Department of Zoology, University of Hong Kong, Hong Kong SAR, China
| | - KYC Chow
- Department of Zoology, University of Hong Kong, Hong Kong SAR, China
| | - CC Hon
- Department of Zoology, University of Hong Kong, Hong Kong SAR, China
| | - KH Hui
- Department of Zoology, University of Hong Kong, Hong Kong SAR, China
| | - J Li
- Department of Zoology, University of Hong Kong, Hong Kong SAR, China
| | - VYY Li
- Department of Zoology, University of Hong Kong, Hong Kong SAR, China
| | - Y Wang
- Department of Zoology, University of Hong Kong, Hong Kong SAR, China
| | - SW Leung
- Department of Microbiology, University of Hong Kong, Hong Kong SAR, China
| | - KY Yuen
- Department of Microbiology, University of Hong Kong, Hong Kong SAR, China
| | - FC Leung
- Department of Zoology, University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
50
|
Chow KYC, Hon CC, Hui RKH, Wong RTY, Yip CW, Zeng F, Leung FCC. Molecular advances in severe acute respiratory syndrome-associated coronavirus (SARS-CoV). GENOMICS, PROTEOMICS & BIOINFORMATICS 2003; 1:247-62. [PMID: 15629054 PMCID: PMC5172416 DOI: 10.1016/s1672-0229(03)01031-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
The sudden outbreak of severe acute respiratory syndrome (SARS) in 2002 prompted the establishment of a global scientific network subsuming most of the traditional rivalries in the competitive field of virology. Within months of the SARS outbreak, collaborative work revealed the identity of the disastrous pathogen as SARS-associated coronavirus (SARS-CoV). However, although the rapid identification of the agent represented an important breakthrough, our understanding of the deadly virus remains limited. Detailed biological knowledge is crucial for the development of effective countermeasures, diagnostic tests, vaccines and antiviral drugs against the SARS-CoV. This article reviews the present state of molecular knowledge about SARS-CoV, from the aspects of comparative genomics, molecular biology of viral genes, evolution, and epidemiology, and describes the diagnostic tests and the anti-viral drugs derived so far based on the available molecular information.
Collapse
|