1
|
Ching WY, Adhikari P, Jawad B, Podgornik R. Towards Quantum-Chemical Level Calculations of SARS-CoV-2 Spike Protein Variants of Concern by First Principles Density Functional Theory. Biomedicines 2023; 11:517. [PMID: 36831053 PMCID: PMC9953097 DOI: 10.3390/biomedicines11020517] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/03/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
The spike protein (S-protein) is a crucial part of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), with its many domains responsible for binding, fusion, and host cell entry. In this review we use the density functional theory (DFT) calculations to analyze the atomic-scale interactions and investigate the consequences of mutations in S-protein domains. We specifically describe the key amino acids and functions of each domain, which are essential for structural stability as well as recognition and fusion processes with the host cell; in addition, we speculate on how mutations affect these properties. Such unprecedented large-scale ab initio calculations, with up to 5000 atoms in the system, are based on the novel concept of amino acid-amino acid-bond pair unit (AABPU) that allows for an alternative description of proteins, providing valuable information on partial charge, interatomic bonding and hydrogen bond (HB) formation. In general, our results show that the S-protein mutations for different variants foster an increased positive partial charge, alter the interatomic interactions, and disrupt the HB networks. We conclude by outlining a roadmap for future computational research of biomolecular virus-related systems.
Collapse
Affiliation(s)
- Wai-Yim Ching
- Department of Physics and Astronomy, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Puja Adhikari
- Department of Physics and Astronomy, University of Missouri-Kansas City, Kansas City, MO 64110, USA
| | - Bahaa Jawad
- Department of Physics and Astronomy, University of Missouri-Kansas City, Kansas City, MO 64110, USA
- Department of Applied Sciences, University of Technology, Baghdad 10066, Iraq
| | - Rudolf Podgornik
- School of Physical Sciences and Kavli Institute of Theoretical Science, University of Chinese Academy of Sciences, Beijing 100049, China
- CAS Key Laboratory of Soft Matter Physics, Institute of Physics, Chinese Academy of Sciences, Beijing 100090, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| |
Collapse
|
2
|
Zaman K, Shete AM, Mishra SK, Kumar A, Reddy MM, Sahay RR, Yadav S, Majumdar T, Pandey AK, Dwivedi GR, Deval H, Singh R, Behera SP, Kumar N, Patil S, Kumar A, Dudhmal M, Joshi Y, Shukla A, Gawande P, Kavathekar A, Kumar N, Kumar V, Kumar K, Singh RS, Kumar M, Tiwari S, Verma A, Yadav PD, Kant R. Omicron BA.2 lineage predominance in severe acute respiratory syndrome coronavirus 2 positive cases during the third wave in North India. Front Med (Lausanne) 2022; 9:955930. [PMID: 36405589 PMCID: PMC9666497 DOI: 10.3389/fmed.2022.955930] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Accepted: 10/03/2022] [Indexed: 01/25/2023] Open
Abstract
Background Recent studies on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) reveal that Omicron variant BA.1 and sub-lineages have revived the concern over resistance to antiviral drugs and vaccine-induced immunity. The present study aims to analyze the clinical profile and genome characterization of the SARS-CoV-2 variant in eastern Uttar Pradesh (UP), North India. Methods Whole-genome sequencing (WGS) was conducted for 146 SARS-CoV-2 samples obtained from individuals who tested coronavirus disease 2019 (COVID-19) positive between the period of 1 January 2022 and 24 February 2022, from three districts of eastern UP. The details regarding clinical and hospitalized status were captured through telephonic interviews after obtaining verbal informed consent. A maximum-likelihood phylogenetic tree was created for evolutionary analysis using MEGA7. Results The mean age of study participants was 33.9 ± 13.1 years, with 73.5% accounting for male patients. Of the 98 cases contacted by telephone, 30 (30.6%) had a travel history (domestic/international), 16 (16.3%) reported having been infected with COVID-19 in past, 79 (80.6%) had symptoms, and seven had at least one comorbidity. Most of the sequences belonged to the Omicron variant, with BA.1 (6.2%), BA.1.1 (2.7%), BA.1.1.1 (0.7%), BA.1.1.7 (5.5%), BA.1.17.2 (0.7%), BA.1.18 (0.7%), BA.2 (30.8%), BA.2.10 (50.7%), BA.2.12 (0.7%), and B.1.617.2 (1.3%) lineages. BA.1 and BA.1.1 strains possess signature spike mutations S:A67V, S:T95I, S:R346K, S:S371L, S:G446S, S:G496S, S:T547K, S:N856K, and S:L981F, and BA.2 contains S:V213G, S:T376A, and S:D405N. Notably, ins214EPE (S1- N-Terminal domain) mutation was found in a significant number of Omicron BA.1 and sub-lineages. The overall Omicron BA.2 lineage was observed in 79.5% of women and 83.2% of men. Conclusion The current study showed a predominance of the Omicron BA.2 variant outcompeting the BA.1 over a period in eastern UP. Most of the cases had a breakthrough infection following the recommended two doses of vaccine with four in five cases being symptomatic. There is a need to further explore the immune evasion properties of the Omicron variant.
Collapse
Affiliation(s)
- Kamran Zaman
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Anita M Shete
- Maximum Containment Facility, Indian Council of Medical Research-National Institute of Virology Pune (ICMR-NIV Pune), Pune, India
| | - Shailendra Kumar Mishra
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Abhinendra Kumar
- Maximum Containment Facility, Indian Council of Medical Research-National Institute of Virology Pune (ICMR-NIV Pune), Pune, India
| | - Mahendra M Reddy
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Rima R Sahay
- Maximum Containment Facility, Indian Council of Medical Research-National Institute of Virology Pune (ICMR-NIV Pune), Pune, India
| | - Shailendra Yadav
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Triparna Majumdar
- Maximum Containment Facility, Indian Council of Medical Research-National Institute of Virology Pune (ICMR-NIV Pune), Pune, India
| | - Ashok K Pandey
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Gaurav Raj Dwivedi
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Hirawati Deval
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Rajeev Singh
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Sthita Pragnya Behera
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Niraj Kumar
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Savita Patil
- Maximum Containment Facility, Indian Council of Medical Research-National Institute of Virology Pune (ICMR-NIV Pune), Pune, India
| | - Ashish Kumar
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Manisha Dudhmal
- Maximum Containment Facility, Indian Council of Medical Research-National Institute of Virology Pune (ICMR-NIV Pune), Pune, India
| | - Yash Joshi
- Maximum Containment Facility, Indian Council of Medical Research-National Institute of Virology Pune (ICMR-NIV Pune), Pune, India
| | - Aishwarya Shukla
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Pranita Gawande
- Maximum Containment Facility, Indian Council of Medical Research-National Institute of Virology Pune (ICMR-NIV Pune), Pune, India
| | - Asif Kavathekar
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Nalin Kumar
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Vijay Kumar
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Kamlesh Kumar
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Ravi Shankar Singh
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Manoj Kumar
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Shashikant Tiwari
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| | - Ajay Verma
- Maximum Containment Facility, Indian Council of Medical Research-National Institute of Virology Pune (ICMR-NIV Pune), Pune, India
| | - Pragya D Yadav
- Maximum Containment Facility, Indian Council of Medical Research-National Institute of Virology Pune (ICMR-NIV Pune), Pune, India
| | - Rajni Kant
- Indian Council of Medical Research-Regional Medical Research Centre Gorakhpur (ICMR-RMRC Gorakhpur), Gorakhpur, India
| |
Collapse
|
3
|
Xiang R, Wang Y, Wang L, Deng X, Huo S, Jiang S, Yu F. Neutralizing monoclonal antibodies against highly pathogenic coronaviruses. Curr Opin Virol 2022; 53:101199. [PMID: 35038651 PMCID: PMC8716168 DOI: 10.1016/j.coviro.2021.12.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 12/17/2021] [Accepted: 12/24/2021] [Indexed: 12/15/2022]
Abstract
The pandemic of Coronavirus Disease 2019 (COVID-19) caused by severe acute respiratory syndrome 2 coronavirus (SARS-CoV-2) is a continuing worldwide threat to human health and social economy. Historically, SARS-CoV-2 follows SARS and MERS as the third coronavirus spreading across borders and continents, but far more dangerous with long-lasting symptomatic consequences. The current situation is strong evidence that coronaviruses will continue to be pathogens of consequence in the future, thus calling for the development of neutralizing antibody-based prophylactics and therapeutics for prevention and treatment of COVID-19 and other human coronavirus diseases. This review summarized the progresses of developing neutralizing monoclonal antibodies against infection of SARS-CoV-2, SARS-CoV, and MERS-CoV, and discussed their potential applications in prevention and treatment of COVID-19 and other human coronavirus diseases.
Collapse
Affiliation(s)
- Rong Xiang
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Yang Wang
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Lili Wang
- Research Center of Chinese Jujube, Hebei Agricultural University, Baoding, China
| | - Xiaoqian Deng
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Shanshan Huo
- College of Life Sciences, Hebei Agricultural University, Baoding, China; Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Baoding, China.
| | - Shibo Jiang
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), School of Basic Medical Sciences, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China.
| | - Fei Yu
- College of Life Sciences, Hebei Agricultural University, Baoding, China; Hebei Key Laboratory of Analysis and Control of Zoonotic Pathogenic Microorganism, Baoding, China.
| |
Collapse
|
4
|
Abstract
Most of SARS-CoV-2 neutralizing antibodies (nAbs) targeted the receptor binding domain (RBD) of the SARS-CoV-2 spike (S) protein. However, mutations at RBD sequences found in the emerging SARS-CoV-2 variants greatly reduced the effectiveness of nAbs. Here we showed that four nAbs, S2-4D, S2-5D, S2-8D, and S2-4A, which recognized a conserved epitope in the S2 subunit of the S protein, can inhibit SARS-CoV-2 infection through blocking the S protein-mediated membrane fusion. Notably, these four nAbs exhibited broadly neutralizing activity against SARS-CoV-2 Alpha, Gamma, Delta, and Epsilon variants. Antisera collected from mice immunized with the identified epitope peptides of these four nAbs also exhibited potent virus neutralizing activity. Discovery of the S2-specific nAbs and their unique antigenic epitopes paves a new path for development of COVID-19 therapeutics and vaccines. IMPORTANCE The spike (S) protein on the surface of SARS-CoV-2 mediates receptor binding and virus-host cell membrane fusion during virus entry. Many neutralizing antibodies (nAbs), which targeted the receptor binding domain (RBD) of S protein, lost the neutralizing activity against the newly emerging SARS-CoV-2 variants with sequence mutations at the RBD. In contrast, the nAb against the highly conserved S2 subunit, which plays the key role in virus–host cell membrane fusion, was poorly discovered. We showed that four S2-specific nAbs, S2-4D, S2-5D, S2-8D, and S2-4A, inhibited SARS-CoV-2 infection through blocking the S protein-mediated membrane fusion. These nAbs exhibited broadly neutralizing activity against Alpha, Gamma, Delta, and Epsilon variants. Antisera induced by the identified epitope peptides also possessed potent neutralizing activity. This work not only unveiled the S2-specific nAbs but also discovered an immunodominant epitope in the S2 subunit that can be rationally designed as the broad-spectrum vaccine against the SARS-like coronaviruses.
Collapse
|
5
|
Essentials of COVID-19 and treatment approaches. DATA SCIENCE FOR COVID-19 2022. [PMCID: PMC8988944 DOI: 10.1016/b978-0-323-90769-9.00026-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The coronavirus family is as old as the 1930s when it first showed symptoms in chicken. The virus thereafter kept evolving and it has significantly taken over a large percentage of people worldwide in the form of this new pandemic. As of the present day, there is no treatment available for coronavirus disease 2019 (COVID-19) (caused by the severe acute respiratory syndrome coronavirus 2 [SARS-CoV-2]), although supportive therapy and preventive measures have shown a tremendous control rate among certain patients. Drugs like remdesivir, camostat, nafamostat, ritonavir/lopinavir, several monoclonal antibodies, and CPs are in their early phases of trials. There are approved by the WHO under an emergency use authorization program. Favipiravir has entered its phase 3 clinical trial and is supported by evidence to show no or less adverse effects in patients infected with SARS-CoV-2. Vaccine development is accelerating its pace, and vaccines will probably become available by the end of the year 2020.
Collapse
|
6
|
Koppisetti RK, Fulcher YG, Van Doren SR. Fusion Peptide of SARS-CoV-2 Spike Rearranges into a Wedge Inserted in Bilayered Micelles. J Am Chem Soc 2021; 143:13205-13211. [PMID: 34375093 PMCID: PMC8370118 DOI: 10.1021/jacs.1c05435] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Indexed: 11/28/2022]
Abstract
The receptor binding and proteolysis of Spike of SARS-CoV-2 release its S2 subunit to rearrange and catalyze viral-cell fusion. This deploys the fusion peptide for insertion into the cell membranes targeted. We show that this fusion peptide transforms from intrinsic disorder in solution into a wedge-shaped structure inserted in bilayered micelles, according to chemical shifts, 15N NMR relaxation, and NOEs. The globular fold of three helices contrasts the open, extended forms of this region observed in the electron density of compact prefusion states. In the hydrophobic, narrow end of the wedge, helices 1 and 2 contact the fatty acyl chains of phospholipids, according to NOEs and proximity to a nitroxide spin label deep in the membrane mimic. The polar end of the wedge may engage and displace lipid head groups and bind Ca2+ ions for membrane fusion. Polar helix 3 protrudes from the bilayer where it might be accessible to antibodies.
Collapse
Affiliation(s)
| | - Yan G. Fulcher
- Dept. of Biochemistry, University of Missouri, Columbia, MO 65211 USA
| | - Steven R. Van Doren
- Dept. of Biochemistry, University of Missouri, Columbia, MO 65211 USA
- Institute for Data Science and Informatics, University of Missouri, Columbia, MO 65211 USA
| |
Collapse
|
7
|
Asarnow D, Wang B, Lee WH, Hu Y, Huang CW, Faust B, Ng PML, Ngoh EZX, Bohn M, Bulkley D, Pizzorno A, Ary B, Tan HC, Lee CY, Minhat RA, Terrier O, Soh MK, Teo FJ, Yeap YYC, Seah SGK, Chan CEZ, Connelly E, Young NJ, Maurer-Stroh S, Renia L, Hanson BJ, Rosa-Calatrava M, Manglik A, Cheng Y, Craik CS, Wang CI. Structural insight into SARS-CoV-2 neutralizing antibodies and modulation of syncytia. Cell 2021; 184:3192-3204.e16. [PMID: 33974910 PMCID: PMC8064868 DOI: 10.1016/j.cell.2021.04.033] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 02/19/2021] [Accepted: 04/19/2021] [Indexed: 12/13/2022]
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is initiated by binding of the viral Spike protein to host receptor angiotensin-converting enzyme 2 (ACE2), followed by fusion of viral and host membranes. Although antibodies that block this interaction are in emergency use as early coronavirus disease 2019 (COVID-19) therapies, the precise determinants of neutralization potency remain unknown. We discovered a series of antibodies that potently block ACE2 binding but exhibit divergent neutralization efficacy against the live virus. Strikingly, these neutralizing antibodies can inhibit or enhance Spike-mediated membrane fusion and formation of syncytia, which are associated with chronic tissue damage in individuals with COVID-19. As revealed by cryoelectron microscopy, multiple structures of Spike-antibody complexes have distinct binding modes that not only block ACE2 binding but also alter the Spike protein conformational cycle triggered by ACE2 binding. We show that stabilization of different Spike conformations leads to modulation of Spike-mediated membrane fusion with profound implications for COVID-19 pathology and immunity.
Collapse
Affiliation(s)
- Daniel Asarnow
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF) School of Medicine, San Francisco, CA, USA; QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| | - Bei Wang
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Wen-Hsin Lee
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Yuanyu Hu
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Ching-Wen Huang
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Bryan Faust
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF) School of Medicine, San Francisco, CA, USA; QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| | - Patricia Miang Lon Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Eve Zi Xian Ngoh
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Markus Bohn
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - David Bulkley
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF) School of Medicine, San Francisco, CA, USA; QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| | - Andrés Pizzorno
- CIRI, Centre International de Recherche en Infectiologie (Team VirPath), Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Beatrice Ary
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Hwee Ching Tan
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Chia Yin Lee
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Rabiatul Adawiyah Minhat
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Olivier Terrier
- CIRI, Centre International de Recherche en Infectiologie (Team VirPath), Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France
| | - Mun Kuen Soh
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Frannie Jiuyi Teo
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Yvonne Yee Chin Yeap
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Shirley Gek Kheng Seah
- Biological Defence Program, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore
| | - Conrad En Zuo Chan
- Biological Defence Program, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore
| | - Emily Connelly
- Department of Pharmaceutical Chemistry, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Nicholas J Young
- Department of Pharmaceutical Chemistry, University of California, San Francisco (UCSF), San Francisco, CA, USA
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute, Agency for Science, Technology and Research (A(∗)STAR), 30 Biopolis Street, Matrix, Singapore 138671, Singapore; Infectious Diseases Laboratories (ID Labs), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; Department of Biological Sciences, Faculty of Science, National University of Singapore, Singapore 117558, Singapore
| | - Laurent Renia
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore; Infectious Diseases Laboratories (ID Labs), Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Brendon John Hanson
- Biological Defence Program, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore
| | - Manuel Rosa-Calatrava
- CIRI, Centre International de Recherche en Infectiologie (Team VirPath), Université de Lyon, INSERM U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 69007 Lyon, France; VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Aashish Manglik
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco (UCSF), San Francisco, CA, USA; Department of Anesthesia and Perioperative Care, UCSF, San Francisco, CA, USA.
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco (UCSF) School of Medicine, San Francisco, CA, USA; QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA; Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA.
| | - Charles S Craik
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA; Department of Pharmaceutical Chemistry, University of California, San Francisco (UCSF), San Francisco, CA, USA.
| | - Cheng-I Wang
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore.
| |
Collapse
|
8
|
Ratre YK, Kahar N, Bhaskar LVKS, Bhattacharya A, Verma HK. Molecular mechanism, diagnosis, and potential treatment for novel coronavirus (COVID-19): a current literature review and perspective. 3 Biotech 2021; 11:94. [PMID: 33520580 PMCID: PMC7832422 DOI: 10.1007/s13205-021-02657-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Novel coronavirus disease 2019 (COVID-19) is a positive-sense single-stranded RNA virus which belongs to the Coronaviridae family. COVID-19 outbreak became evident after the severe acute respiratory syndrome coronavirus and the Middle East respiratory syndrome coronavirus in the twenty-first century as the start of the third deadly coronavirus. Currently, research is at an early stage, and the exact etiological dimensions of COVID-19 are unknown. Several candidate drugs and plasma therapy have been considered and evaluated for the treatment of severe COVID-19 patients. These include clinically available drugs such as chloroquine, hydroxychloroquine, and lopinavir/ritonavir. However, understanding the pathogenic mechanisms of this virus is critical for predicting interaction with humans. Based on recent evidence, we have summarized the current virus biology in terms of the possible understanding of the various pathophysiologies, molecular mechanisms, recent efficient diagnostics, and therapeutic approaches to control the disease. In addition, we briefly reviewed the biochemistry of leading candidates for novel therapies and their current status in clinical trials. As information from COVID-19 is evolving rapidly, this review will help the researcher to consider new insights and potential therapeutic approaches based on up-to-date knowledge. Finally, this review illustrates a list of alternative therapeutic solutions for a viral infection.
Collapse
Affiliation(s)
| | - Namrata Kahar
- Department of Zoology, Guru Ghasidas Vishwavidyalaya, Bilaspur, India
| | | | - Antaripa Bhattacharya
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, Naples, Italy
| | - Henu Kumar Verma
- Developmental and Stem Cell Biology Lab, Institute of Experimental Endocrinology and Oncology CNR, Naples, Italy
| |
Collapse
|
9
|
Domains and Functions of Spike Protein in Sars-Cov-2 in the Context of Vaccine Design. Viruses 2021; 13:v13010109. [PMID: 33466921 PMCID: PMC7829931 DOI: 10.3390/v13010109] [Citation(s) in RCA: 201] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/11/2022] Open
Abstract
The spike protein in SARS-CoV-2 (SARS-2-S) interacts with the human ACE2 receptor to gain entry into a cell to initiate infection. Both Pfizer/BioNTech's BNT162b2 and Moderna's mRNA-1273 vaccine candidates are based on stabilized mRNA encoding prefusion SARS-2-S that can be produced after the mRNA is delivered into the human cell and translated. SARS-2-S is cleaved into S1 and S2 subunits, with S1 serving the function of receptor-binding and S2 serving the function of membrane fusion. Here, I dissect in detail the various domains of SARS-2-S and their functions discovered through a variety of different experimental and theoretical approaches to build a foundation for a comprehensive mechanistic understanding of how SARS-2-S works to achieve its function of mediating cell entry and subsequent cell-to-cell transmission. The integration of structure and function of SARS-2-S in this review should enhance our understanding of the dynamic processes involving receptor binding, multiple cleavage events, membrane fusion, viral entry, as well as the emergence of new viral variants. I highlighted the relevance of structural domains and dynamics to vaccine development, and discussed reasons for the spike protein to be frequently featured in the conspiracy theory claiming that SARS-CoV-2 is artificially created.
Collapse
|
10
|
Fouladirad S, Bach H. Development of Coronavirus Treatments Using Neutralizing Antibodies. Microorganisms 2021; 9:microorganisms9010165. [PMID: 33451069 PMCID: PMC7828509 DOI: 10.3390/microorganisms9010165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
The Coronavirus disease 2019 (COVID-19), caused by the novel coronavirus SARS-CoV-2, was first reported in December 2019 in Wuhan, Hubei province, China. This virus has led to 61.8 million cases worldwide being reported as of December 1st, 2020. Currently, there are no definite approved therapies endorsed by the World Health Organization for COVID-19, focusing only on supportive care. Treatment centers around symptom management, including oxygen therapy or invasive mechanical ventilation. Immunotherapy has the potential to play a role in the treatment of SARS-CoV-2. Monoclonal antibodies (mAbs), in particular, is a relatively new approach in the world of infectious diseases and has the benefit of overcoming challenges with serum therapy and intravenous immunoglobulins preparations. Here, we reviewed the articles published in PubMed with the purpose of summarizing the currently available evidence for the use of neutralizing antibodies as a potential treatment for coronaviruses. Studies reporting in vivo results were summarized and analyzed. Despite promising data from some studies, none of them progressed to clinical trials. It is expected that neutralizing antibodies might offer an alternative for COVID-19 treatment. Thus, there is a need for randomized trials to understand the potential use of this treatment.
Collapse
Affiliation(s)
- Saman Fouladirad
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z, Canada;
| | - Horacio Bach
- Department of Medicine, University of British Columbia, Vancouver, BC V6T 1Z, Canada;
- Division of Infectious Diseases, University of British Columbia, Vancouver, BC V6T 1Z, Canada
- Correspondence: ; Tel.: +1-604-727-9719; Fax: +1-604-875-4013
| |
Collapse
|
11
|
Xiu S, Dick A, Ju H, Mirzaie S, Abdi F, Cocklin S, Zhan P, Liu X. Inhibitors of SARS-CoV-2 Entry: Current and Future Opportunities. J Med Chem 2020; 63:12256-12274. [PMID: 32539378 PMCID: PMC7315836 DOI: 10.1021/acs.jmedchem.0c00502] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Indexed: 02/07/2023]
Abstract
Recently, a novel coronavirus initially designated 2019-nCoV but now termed SARS-CoV-2 has emerged and raised global concerns due to its virulence. SARS-CoV-2 is the etiological agent of "coronavirus disease 2019", abbreviated to COVID-19, which despite only being identified at the very end of 2019, has now been classified as a pandemic by the World Health Organization (WHO). At this time, no specific prophylactic or postexposure therapy for COVID-19 are currently available. Viral entry is the first step in the SARS-CoV-2 lifecycle and is mediated by the trimeric spike protein. Being the first stage in infection, entry of SARS-CoV-2 into host cells is an extremely attractive therapeutic intervention point. Within this review, we highlight therapeutic intervention strategies for anti-SARS-CoV, MERS-CoV, and other coronaviruses and speculate upon future directions for SARS-CoV-2 entry inhibitor designs.
Collapse
Affiliation(s)
- Siyu Xiu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Alexej Dick
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, Pennsylvania 19102, United States
| | - Han Ju
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Sako Mirzaie
- Department of Biochemistry, Sanandaj Branch, Islamic Azad University, Sanandaj 6616935391, Iran
| | - Fatemeh Abdi
- Department of Cellular and Molecular Biology, Islamic Azad University, Tehran North Branch, Tehran 1651153311, Iran
| | - Simon Cocklin
- Department of Biochemistry & Molecular Biology, Drexel University College of Medicine, Rooms 10307, 10309, and 10315, 245 North 15th Street, Philadelphia, Pennsylvania 19102, United States
| | - Peng Zhan
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| | - Xinyong Liu
- Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Shandong University, Jinan 250012, China
| |
Collapse
|
12
|
Kooshkaki O, Derakhshani A, Conradie AM, Hemmat N, Barreto SG, Baghbanzadeh A, Singh PK, Safarpour H, Asadzadeh Z, Najafi S, Brunetti O, Racanelli V, Silvestris N, Baradaran B. Coronavirus Disease 2019: A Brief Review of the Clinical Manifestations and Pathogenesis to the Novel Management Approaches and Treatments. Front Oncol 2020; 10:572329. [PMID: 33194671 PMCID: PMC7658542 DOI: 10.3389/fonc.2020.572329] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 09/02/2020] [Indexed: 12/16/2022] Open
Abstract
The recent outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or coronavirus disease 2019 (COVID-19) in China, which spread to the rest of the world, led the World Health Organization to classify it as a global pandemic. COVID-19 belongs to the Bettacoronavirus genus of the Coronaviridae family, and it mainly spreads through the respiratory tract. Studies have now confirmed a human-to-human transmission as the primary pathway of spread. COVID-19 patients with a history of diseases such as respiratory system diseases, immune deficiency, diabetes, cardiovascular disease, and cancer are prone to adverse events (admission to the intensive care unit requiring invasive ventilation or even death). The current focus has been on the development of novel therapeutics, including antivirals, monoclonal antibodies, and vaccines. However, although there is undoubtedly an urgent need to identify effective treatment options against infection with COVID-19, it is equally important to clarify management protocols for the other significant diseases from which these patients may suffer, including cancer. This review summarizes the current evidence regarding the epidemiology, pathogenesis, and management of patients with COVID-19. It also aims to provide the reader with insights into COVID-19 in pregnant patients and those with cancer, outlining necessary precautions relevant to cancer patients. Finally, we provide the available evidence on the latest potent antiviral drugs and vaccines of COVID-19 and the ongoing drug trials.
Collapse
Affiliation(s)
- Omid Kooshkaki
- Student Research Committee, Birjand University of Medical Sciences, Birjand, Iran
- Department of Immunology, Birjand University of Medical Sciences, Birjand, Iran
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, Bari, Italy
| | | | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Savio George Barreto
- Division of Surgery and Perioperative Medicine, Flinders Medical Centre, Adelaide, SA, Australia
- College of Medicine and Public Health, Flinders University, Adelaide, SA, Australia
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Pankaj Kumar Singh
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, United States
| | - Hossein Safarpour
- Cellularand Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Souzan Najafi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, Bari, Italy
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, University of Bari “AldoMoro”, Bari, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, Bari, Italy
- Department of Biomedical Sciences and Human Oncology, University of Bari “AldoMoro”, Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
13
|
Conzelmann C, Gilg A, Groß R, Schütz D, Preising N, Ständker L, Jahrsdörfer B, Schrezenmeier H, Sparrer KM, Stamminger T, Stenger S, Münch J, Müller JA. An enzyme-based immunodetection assay to quantify SARS-CoV-2 infection. Antiviral Res 2020; 181:104882. [PMID: 32738255 PMCID: PMC7388004 DOI: 10.1016/j.antiviral.2020.104882] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Revised: 07/12/2020] [Accepted: 07/14/2020] [Indexed: 01/07/2023]
Abstract
SARS-CoV-2 is a novel pandemic coronavirus that caused a global health and economic crisis. The development of efficient drugs and vaccines against COVID-19 requires detailed knowledge about SARS-CoV-2 biology. Several techniques to detect SARS-CoV-2 infection have been established, mainly based on counting infected cells by staining plaques or foci, or by quantifying the viral genome by PCR. These methods are laborious, time-consuming and expensive and therefore not suitable for a high sample throughput or rapid diagnostics. We here report a novel enzyme-based immunodetection assay that directly quantifies the amount of de novo synthesized viral spike protein within fixed and permeabilized cells. This in-cell ELISA enables a rapid and quantitative detection of SARS-CoV-2 infection in microtiter format, regardless of the virus isolate or target cell culture. It follows the established method of performing ELISA assays and does not require expensive instrumentation. Utilization of the in-cell ELISA allows to e.g. determine TCID50 of virus stocks, antiviral efficiencies (IC50 values) of drugs or neutralizing activity of sera. Thus, the in-cell spike ELISA represents a promising alternative to study SARS-CoV-2 infection and inhibition and may facilitate future research.
Collapse
Affiliation(s)
- Carina Conzelmann
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Andrea Gilg
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Desiree Schütz
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Nico Preising
- Core Facility Functional Peptidomics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Ludger Ständker
- Core Facility Functional Peptidomics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Bernd Jahrsdörfer
- Institute for Transfusion Medicine, Ulm University, 89081, Ulm, Germany,Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Services Baden-Württemberg-Hessen and University Hospital Ulm, 89081, Ulm, Germany
| | - Hubert Schrezenmeier
- Institute for Transfusion Medicine, Ulm University, 89081, Ulm, Germany,Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Services Baden-Württemberg-Hessen and University Hospital Ulm, 89081, Ulm, Germany
| | | | - Thomas Stamminger
- Institute of Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Steffen Stenger
- Institute of Medical Microbiology and Hygiene, Ulm University Medical Center, 89081, Ulm, Germany
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany,Core Facility Functional Peptidomics, Ulm University Medical Center, 89081, Ulm, Germany
| | - Janis A. Müller
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany,Corresponding author
| |
Collapse
|
14
|
Jahanshahlu L, Rezaei N. Monoclonal antibody as a potential anti-COVID-19. Biomed Pharmacother 2020; 129:110337. [PMID: 32534226 PMCID: PMC7269943 DOI: 10.1016/j.biopha.2020.110337] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 05/25/2020] [Accepted: 05/30/2020] [Indexed: 12/19/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is expanding rapidly, which made it as one of top priorities for scientists to develop novel treatment strategies. Researchers are racing to develop treatments based on antibodies to block and/or neutralize the coronavirus in affected patients. Initially, the genetic and structural similarity of the virus to severe acute respiratory syndrome coronavirus (SARS-CoV) created the potential for understanding disease pathogenesis. Researchers have published reports of specific monoclonal antibodies against to COVID-19 (B38, H4, 47D11) and hope that this method is effective. As well as studies on patients who are plasma therapy, the patient's condition shows improvement. The evidence for these studies is very promising and demonstrates the potential of monoclonal antibody therapy as a therapeutic approach and prevention of covid-19 infection.
Collapse
Affiliation(s)
- Leila Jahanshahlu
- School of Medical, Zanjan University of Medical Sciences, Zanjan, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Zanjan, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| |
Collapse
|
15
|
Zheng Z, Monteil VM, Maurer-Stroh S, Yew CW, Leong C, Mohd-Ismail NK, Cheyyatraivendran Arularasu S, Chow VTK, Lin RTP, Mirazimi A, Hong W, Tan YJ. Monoclonal antibodies for the S2 subunit of spike of SARS-CoV-1 cross-react with the newly-emerged SARS-CoV-2. ACTA ACUST UNITED AC 2020; 25. [PMID: 32700671 PMCID: PMC7376845 DOI: 10.2807/1560-7917.es.2020.25.28.2000291] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background A novel coronavirus, SARS-CoV-2, which emerged at the end of 2019 and causes COVID-19, has resulted in worldwide human infections. While genetically distinct, SARS-CoV-1, the aetiological agent responsible for an outbreak of severe acute respiratory syndrome (SARS) in 2002–2003, utilises the same host cell receptor as SARS-CoV-2 for entry: angiotensin-converting enzyme 2 (ACE2). Parts of the SARS-CoV-1 spike glycoprotein (S protein), which interacts with ACE2, appear conserved in SARS-CoV-2. Aim The cross-reactivity with SARS-CoV-2 of monoclonal antibodies (mAbs) previously generated against the S protein of SARS-CoV-1 was assessed. Methods The SARS-CoV-2 S protein sequence was aligned to those of SARS-CoV-1, Middle East respiratory syndrome (MERS) and common-cold coronaviruses. Abilities of mAbs generated against SARS-CoV-1 S protein to bind SARS-CoV-2 or its S protein were tested with SARS-CoV-2 infected cells as well as cells expressing either the full length protein or a fragment of its S2 subunit. Quantitative ELISA was also performed to compare binding of mAbs to recombinant S protein. Results An immunogenic domain in the S2 subunit of SARS-CoV-1 S protein is highly conserved in SARS-CoV-2 but not in MERS and human common-cold coronaviruses. Four murine mAbs raised against this immunogenic fragment could recognise SARS-CoV-2 S protein expressed in mammalian cell lines. In particular, mAb 1A9 was demonstrated to detect S protein in SARS-CoV-2-infected cells and is suitable for use in a sandwich ELISA format. Conclusion The cross-reactive mAbs may serve as useful tools for SARS-CoV-2 research and for the development of diagnostic assays for COVID-19.
Collapse
Affiliation(s)
- Zhiqiang Zheng
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.,Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Vanessa Marthe Monteil
- Public Health Agency of Sweden, Stockholm, Sweden.,Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| | - Sebastian Maurer-Stroh
- National Public Health Laboratory (NPHL), National Centre for Infectious Diseases (NCID), Singapore.,Department of Biological Sciences (DBS), National University of Singapore, Singapore.,Bioinformatics Institute (BII), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Chow Wenn Yew
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Carol Leong
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Nur Khairiah Mohd-Ismail
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.,Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Suganya Cheyyatraivendran Arularasu
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.,Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Vincent Tak Kwong Chow
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Raymond Tzer Pin Lin
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.,National Public Health Laboratory (NPHL), National Centre for Infectious Diseases (NCID), Singapore
| | - Ali Mirazimi
- National Veterinary Institute, Uppsala, Sweden.,Public Health Agency of Sweden, Stockholm, Sweden.,Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
| | - Wanjin Hong
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Yee-Joo Tan
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore.,Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore.,Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| |
Collapse
|
16
|
Benani A, Ben Mkaddem S. Mechanisms Underlying Potential Therapeutic Approaches for COVID-19. Front Immunol 2020; 11:1841. [PMID: 32793246 PMCID: PMC7385230 DOI: 10.3389/fimmu.2020.01841] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 07/08/2020] [Indexed: 12/16/2022] Open
Abstract
Coronavirus disease (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which is a betacoronavirus, and is associated with cytokine storm inflammation and lung injury, leading to respiratory distress. The transmission of the virus is mediated by human contact. To control and prevent the spread of this virus, the majority of people worldwide are facing quarantine; patients are being subjected to non-specific treatments under isolation. To prevent and stop the COVID-19 pandemic, several clinical trials are in the pipeline. The current clinical trials either target the intracellular replication and spread of the virus or the cytokine storm inflammation seen in COVID-19 cases during the later stages of the disease. Since both targeting strategies are different, the window drug administration plays a crucial role in the efficacy of the treatment. Here, we review the mechanism underlying SARS-CoV-2 cell infection and potential future therapeutic approaches.
Collapse
Affiliation(s)
- Abdelouaheb Benani
- Unité de Biologie Moléculaire, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Sanae Ben Mkaddem
- U978 Institut National de la Santé et de la Recherche Médicale, Bobigny, France.,UFR SMBH, Université Sorbonne Paris Nord, Bobigny, France
| |
Collapse
|
17
|
Corral-Lugo A, López-Siles M, López D, McConnell MJ, Martin-Galiano AJ. Identification and Analysis of Unstructured, Linear B-Cell Epitopes in SARS-CoV-2 Virion Proteins for Vaccine Development. Vaccines (Basel) 2020; 8:397. [PMID: 32698423 PMCID: PMC7564417 DOI: 10.3390/vaccines8030397] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 07/14/2020] [Accepted: 07/17/2020] [Indexed: 12/13/2022] Open
Abstract
The efficacy of SARS-CoV-2 nucleic acid-based vaccines may be limited by proteolysis of the translated product due to anomalous protein folding. This may be the case for vaccines employing linear SARS-CoV-2 B-cell epitopes identified in previous studies since most of them participate in secondary structure formation. In contrast, we have employed a consensus of predictors for epitopic zones plus a structural filter for identifying 20 unstructured B-cell epitope-containing loops (uBCELs) in S, M, and N proteins. Phylogenetic comparison suggests epitope switching with respect to SARS-CoV in some of the identified uBCELs. Such events may be associated with the reported lack of serum cross-protection between the 2003 and 2019 pandemic strains. Incipient variability within a sample of 1639 SARS-CoV-2 isolates was also detected for 10 uBCELs which could cause vaccine failure. Intermediate stages of the putative epitope switch events were observed in bat coronaviruses in which additive mutational processes possibly facilitating evasion of the bat immune system appear to have taken place prior to transfer to humans. While there was some overlap between uBCELs and previously validated SARS-CoV B-cell epitopes, multiple uBCELs had not been identified in prior studies. Overall, these uBCELs may facilitate the development of biomedical products for SARS-CoV-2.
Collapse
Affiliation(s)
- Andrés Corral-Lugo
- Intrahospital Infections Unit, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain; (A.C.-L.); (M.L.-S.)
| | - Mireia López-Siles
- Intrahospital Infections Unit, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain; (A.C.-L.); (M.L.-S.)
| | - Daniel López
- Immune Presentation and Regulation Unit, Instituto de Salud Carlos III, 28220 Madrid, Spain;
| | - Michael J. McConnell
- Intrahospital Infections Unit, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain; (A.C.-L.); (M.L.-S.)
| | - Antonio J. Martin-Galiano
- Intrahospital Infections Unit, National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), 28220 Madrid, Spain; (A.C.-L.); (M.L.-S.)
| |
Collapse
|
18
|
Fan X, Cao D, Kong L, Zhang X. Cryo-EM analysis of the post-fusion structure of the SARS-CoV spike glycoprotein. Nat Commun 2020; 11:3618. [PMID: 32681106 PMCID: PMC7367865 DOI: 10.1038/s41467-020-17371-6] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/25/2020] [Indexed: 12/28/2022] Open
Abstract
Global emergencies caused by the severe acute respiratory syndrome coronavirus (SARS-CoV), Middle-East respiratory syndrome coronavirus (MERS-CoV) and SARS-CoV-2 significantly endanger human health. The spike (S) glycoprotein is the key antigen and its conserved S2 subunit contributes to viral entry by mediating host-viral membrane fusion. However, structural information of the post-fusion S2 from these highly pathogenic human-infecting coronaviruses is still lacking. We used single-particle cryo-electron microscopy to show that the post-fusion SARS-CoV S2 forms a further rotated HR1-HR2 six-helix bundle and a tightly bound linker region upstream of the HR2 motif. The structures of pre- and post-fusion SARS-CoV S glycoprotein dramatically differ, resembling that of the Mouse hepatitis virus (MHV) and other class I viral fusion proteins. This structure suggests potential targets for the development of vaccines and therapies against a wide range of SARS-like coronaviruses. The spike (S) protein of coronaviruses is responsible for receptor recognition and the fusion between the viral membrane and the of cell host membrane. Here the authors report a cryo-EM structure of SARS-CoV post-fusion S2 trimer, providing insights into the fusion mechanism that could be useful for therapeutic development against coronaviruses.
Collapse
Affiliation(s)
- Xiaoyi Fan
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.,University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Duanfang Cao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.
| | - Lingfei Kong
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China
| | - Xinzheng Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China. .,University of Chinese Academy of Sciences, 100049, Beijing, China. .,Center for Biological Imaging, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 100101, Beijing, China.
| |
Collapse
|
19
|
Wang B, Asarnow D, Lee WH, Huang CW, Faust B, Ng PML, Ngoh EZX, Bohn M, Bulkley D, Pizzorno A, Tan HC, Lee CY, Minhat RA, Terrier O, Soh MK, Teo FJ, Yeap YYC, Hu Y, Seah SGK, Maurer-Stroh S, Renia L, Hanson BJ, Rosa-Calatrava M, Manglik A, Cheng Y, Craik CS, Wang CI. Bivalent binding of a fully human IgG to the SARS-CoV-2 spike proteins reveals mechanisms of potent neutralization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.07.14.203414. [PMID: 32699850 PMCID: PMC7373131 DOI: 10.1101/2020.07.14.203414] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In vitro antibody selection against pathogens from naïve combinatorial libraries can yield various classes of antigen-specific binders that are distinct from those evolved from natural infection1-4. Also, rapid neutralizing antibody discovery can be made possible by a strategy that selects for those interfering with pathogen and host interaction5. Here we report the discovery of antibodies that neutralize SARS-CoV-2, the virus responsible for the COVID-19 pandemic, from a highly diverse naïve human Fab library. Lead antibody 5A6 blocks the receptor binding domain (RBD) of the viral spike from binding to the host receptor angiotensin converting enzyme 2 (ACE2), neutralizes SARS-CoV-2 infection of Vero E6 cells, and reduces viral replication in reconstituted human nasal and bronchial epithelium models. 5A6 has a high occupancy on the viral surface and exerts its neutralization activity via a bivalent binding mode to the tip of two neighbouring RBDs at the ACE2 interaction interface, one in the "up" and the other in the "down" position, explaining its superior neutralization capacity. Furthermore, 5A6 is insensitive to several spike mutations identified in clinical isolates, including the D614G mutant that has become dominant worldwide. Our results suggest that 5A6 could be an effective prophylactic and therapeutic treatment of COVID-19.
Collapse
Affiliation(s)
- Bei Wang
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Daniel Asarnow
- Department of Biochemistry and Biophysics, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| | - Wen-Hsin Lee
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Ching-Wen Huang
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Bryan Faust
- Department of Biochemistry and Biophysics, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| | - Patricia Miang Lon Ng
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Eve Zi Xian Ngoh
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Markus Bohn
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - David Bulkley
- Department of Biochemistry and Biophysics, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
| | - Andrés Pizzorno
- Virologie et Pathologie Humaine - VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Hwee Ching Tan
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Chia Yin Lee
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Rabiatul Adawiyah Minhat
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Olivier Terrier
- Virologie et Pathologie Humaine - VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Mun Kuen Soh
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Frannie Jiuyi Teo
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Yvonne Yee Chin Yeap
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Yuanyu Hu
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| | - Shirley Gek Kheng Seah
- Biological Defence Program, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute, Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, Singapore 138671, Singapore
| | - Laurent Renia
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Brendon John Hanson
- Biological Defence Program, DSO National Laboratories, 27 Medical Drive, Singapore 117510, Singapore
| | - Manuel Rosa-Calatrava
- Virologie et Pathologie Humaine - VirPath team, Centre International de Recherche en Infectiologie (CIRI), INSERM U1111, CNRS UMR5308, ENS Lyon, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
- VirNext, Faculté de Médecine RTH Laennec, Université Claude Bernard Lyon 1, Université de Lyon, Lyon, France
| | - Aashish Manglik
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF), San Francisco, CA, USA
- Department of Anesthesia and Perioperative Care, UCSF, San Francisco, CA, USA
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
- Howard Hughes Medical Institute, UCSF, San Francisco, CA, USA
| | - Charles S. Craik
- QBI COVID-19 Research Group (QCRG), San Francisco, CA, USA
- Department of Pharmaceutical Chemistry, University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Cheng-I Wang
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), 8A Biomedical Grove, #03-06 Immunos, Singapore 138648, Singapore
| |
Collapse
|
20
|
Zheng Z, Monteil VM, Maurer-Stroh S, Yew CW, Leong C, Mohd-Ismail NK, Cheyyatraivendran Arularasu S, Chow VTK, Lin RTP, Mirazimi A, Hong W, Tan YJ. Monoclonal antibodies for the S2 subunit of spike of SARS-CoV-1 cross-react with the newly-emerged SARS-CoV-2. Euro Surveill 2020. [PMID: 32700671 DOI: 10.1101/2020.03.06.980037] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/12/2023] Open
Abstract
BackgroundA novel coronavirus, SARS-CoV-2, which emerged at the end of 2019 and causes COVID-19, has resulted in worldwide human infections. While genetically distinct, SARS-CoV-1, the aetiological agent responsible for an outbreak of severe acute respiratory syndrome (SARS) in 2002-2003, utilises the same host cell receptor as SARS-CoV-2 for entry: angiotensin-converting enzyme 2 (ACE2). Parts of the SARS-CoV-1 spike glycoprotein (S protein), which interacts with ACE2, appear conserved in SARS-CoV-2.AimThe cross-reactivity with SARS-CoV-2 of monoclonal antibodies (mAbs) previously generated against the S protein of SARS-CoV-1 was assessed.MethodsThe SARS-CoV-2 S protein sequence was aligned to those of SARS-CoV-1, Middle East respiratory syndrome (MERS) and common-cold coronaviruses. Abilities of mAbs generated against SARS-CoV-1 S protein to bind SARS-CoV-2 or its S protein were tested with SARS-CoV-2 infected cells as well as cells expressing either the full length protein or a fragment of its S2 subunit. Quantitative ELISA was also performed to compare binding of mAbs to recombinant S protein.ResultsAn immunogenic domain in the S2 subunit of SARS-CoV-1 S protein is highly conserved in SARS-CoV-2 but not in MERS and human common-cold coronaviruses. Four murine mAbs raised against this immunogenic fragment could recognise SARS-CoV-2 S protein expressed in mammalian cell lines. In particular, mAb 1A9 was demonstrated to detect S protein in SARS-CoV-2-infected cells and is suitable for use in a sandwich ELISA format.ConclusionThe cross-reactive mAbs may serve as useful tools for SARS-CoV-2 research and for the development of diagnostic assays for COVID-19.
Collapse
Affiliation(s)
- Zhiqiang Zheng
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Vanessa Marthe Monteil
- Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
- Public Health Agency of Sweden, Stockholm, Sweden
| | - Sebastian Maurer-Stroh
- Bioinformatics Institute (BII), A*STAR (Agency for Science, Technology and Research), Singapore
- Department of Biological Sciences (DBS), National University of Singapore, Singapore
- National Public Health Laboratory (NPHL), National Centre for Infectious Diseases (NCID), Singapore
| | - Chow Wenn Yew
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Carol Leong
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Nur Khairiah Mohd-Ismail
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Suganya Cheyyatraivendran Arularasu
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Vincent Tak Kwong Chow
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Raymond Tzer Pin Lin
- National Public Health Laboratory (NPHL), National Centre for Infectious Diseases (NCID), Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Ali Mirazimi
- National Veterinary Institute, Uppsala, Sweden
- Department of Laboratory Medicine, Karolinska Institute, Huddinge, Sweden
- Public Health Agency of Sweden, Stockholm, Sweden
| | - Wanjin Hong
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| | - Yee-Joo Tan
- Infectious Diseases programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
- Immunology programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore
| |
Collapse
|
21
|
Elhusseiny KM, Abd-Elhay FAE, Kamel MG. Possible therapeutic agents for COVID-19: a comprehensive review. Expert Rev Anti Infect Ther 2020; 18:1005-1020. [PMID: 32538209 DOI: 10.1080/14787210.2020.1782742] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
INTRODUCTION Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) has emerged in China. There are no available vaccines or antiviral drugs for COVID-19 patients. Herein, we represented possible therapeutic agents that may stand as a potential therapy against COVID-19. AREAS COVERED We searched PubMed, Google Scholar, and clinicaltrials.gov for relevant papers. We showed some agents with potentially favorable efficacy, acceptable safety as well as good pharmacokinetic profiles. Several therapies are under assessment to evaluate their efficacy and safety for COVID-19. However, some drugs were withdrawn due to their side effects after demonstrating some clinical efficacy. Indeed, the most effective therapies could be organ function support, convalescent plasma, anticoagulants, and immune as well as antiviral therapies, especially anti-influenza drugs due to the similarities between respiratory viruses regarding viral entry, uncoating, and replication. We encourage giving more attention to favipiravir, remdesivir, and measles vaccine. EXPERT OPINION A combination, at least dual or even triple therapy, of the aforementioned efficacious and safe therapies is greatly recommended for COVID-19. Further, patients should have a routine assessment for their coagulation and bleeding profiles as well as their inflammatory and cytokine concentrations.
Collapse
Affiliation(s)
- Khaled Mosaad Elhusseiny
- Faculty of Medicine, Al-Azhar University , Cairo, Egypt.,Sayed Galal University Hospital , Cairo, Egypt.,Egyptian Collaborative Research Team , Egypt
| | | | | |
Collapse
|
22
|
Poh CM, Carissimo G, Wang B, Amrun SN, Lee CYP, Chee RSL, Fong SW, Yeo NKW, Lee WH, Torres-Ruesta A, Leo YS, Chen MIC, Tan SY, Chai LYA, Kalimuddin S, Kheng SSG, Thien SY, Young BE, Lye DC, Hanson BJ, Wang CI, Renia L, Ng LFP. Two linear epitopes on the SARS-CoV-2 spike protein that elicit neutralising antibodies in COVID-19 patients. Nat Commun 2020; 11:2806. [PMID: 32483236 PMCID: PMC7264175 DOI: 10.1038/s41467-020-16638-2] [Citation(s) in RCA: 300] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022] Open
Abstract
Given the ongoing SARS-CoV-2 pandemic, identification of immunogenic targets against the coronavirus spike glycoprotein will provide crucial advances towards the development of sensitive diagnostic tools and potential vaccine candidate targets. In this study, using pools of overlapping linear B-cell peptides, we report two IgG immunodominant regions on SARS-CoV-2 spike glycoprotein that are recognised by sera from COVID-19 convalescent patients. Notably, one is specific to SARS-CoV-2, which is located in close proximity to the receptor binding domain. The other region, which is localised at the fusion peptide, could potentially function as a pan-SARS target. Functionally, antibody depletion assays demonstrate that antibodies targeting these immunodominant regions significantly alter virus neutralisation capacities. Taken together, identification and validation of these neutralising B-cell epitopes will provide insights towards the design of diagnostics and vaccine candidates against this high priority coronavirus.
Collapse
Affiliation(s)
- Chek Meng Poh
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
| | - Guillaume Carissimo
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
| | - Bei Wang
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
| | - Siti Naqiah Amrun
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
| | - Cheryl Yi-Pin Lee
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
| | - Rhonda Sin-Ling Chee
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
| | - Siew-Wai Fong
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
- Department of Biological Science, National University of Singapore, Singapore, Singapore
| | - Nicholas Kim-Wah Yeo
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
| | - Wen-Hsin Lee
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
| | - Anthony Torres-Ruesta
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore, 117596, Singapore
| | - Yee-Sin Leo
- National Centre for Infectious Diseases, 16 Jalan Tan Tock Seng, Singapore, 308442, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, 10 Medical Drive, Singapore, 117597, Singapore
| | - Mark I-Cheng Chen
- National Centre for Infectious Diseases, 16 Jalan Tan Tock Seng, Singapore, 308442, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, 12 Science Drive 2, #10-01, Singapore, 117549, Singapore
| | - Seow-Yen Tan
- Department of Infectious Diseases, Changi General Hospital, 2 Simei Street 3, Singapore, 529889, Singapore
| | - Louis Yi Ann Chai
- Department of Infectious Diseases, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
- Department of Medicine, National University Hospital, 5 Lower Kent Ridge Road, Singapore, 119074, Singapore
| | - Shirin Kalimuddin
- Department of Infectious Diseases, Singapore General Hospital, 31 Third Hospital Ave, #03-03 Bowyer Block C, Singapore, 168753, Singapore
- Emerging Infectious Disease Program, Duke-NUS Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Shirley Seah Gek Kheng
- Biological Defence Program, DSO National Laboratories, 27 Medical Drive, Singapore, 117510, Singapore
| | - Siew-Yee Thien
- Department of Infectious Diseases, Singapore General Hospital, 31 Third Hospital Ave, #03-03 Bowyer Block C, Singapore, 168753, Singapore
| | - Barnaby Edward Young
- National Centre for Infectious Diseases, 16 Jalan Tan Tock Seng, Singapore, 308442, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
| | - David C Lye
- National Centre for Infectious Diseases, 16 Jalan Tan Tock Seng, Singapore, 308442, Singapore
- Department of Infectious Diseases, Tan Tock Seng Hospital, 11 Jalan Tan Tock Seng, Singapore, 308433, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 11 Mandalay Road, Singapore, 308232, Singapore
- Yong Loo Lin School of Medicine, National University of Singapore and National University Health System, 10 Medical Drive, Singapore, 117597, Singapore
| | - Brendon John Hanson
- Biological Defence Program, DSO National Laboratories, 27 Medical Drive, Singapore, 117510, Singapore
| | - Cheng-I Wang
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
| | - Laurent Renia
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore
| | - Lisa F P Ng
- Singapore Immunology Network, Agency of Science, Technology and Research, Immunos, Biopolis, Singapore, 138648, Singapore.
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, 8 Medical Drive, Singapore, 117596, Singapore.
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, 8 West Derby Street, Liverpool, L7 3EA, United Kingdom.
| |
Collapse
|
23
|
Teo SHC, Wu JP, Mok CK, Tan YJ. A NS1-binding monoclonal antibody interacts with two residues that are highly conserved in seasonal as well as newly emerged influenza A virus. Pathog Dis 2019; 77:5370084. [PMID: 30839053 PMCID: PMC7273928 DOI: 10.1093/femspd/ftz012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 03/05/2019] [Indexed: 11/22/2022] Open
Abstract
The non-structural protein 1 (NS1) of influenza A virus (IAV) is a multifunctional protein that antagonizes host antiviral responses, modulating virus pathogenesis. As such, it serves as a good target for research and diagnostic assay development. In this study, we have generated a novel monoclonal antibody (mAb) 19H9 and epitope mapping revealed that two residues, P85 and Y89, of NS1 are essential for interacting with this mAb. Furthermore, residues P85 and Y89 are found to be highly conserved across different IAV subtypes, namely seasonal H1N1 and H3N2, as well as the highly pathogenic H5N1 and H5N6 avian strains. Indeed, mAb 19H9 exhibits broad cross-reactivity with IAV strains of different subtypes. The binding of mAb 19H9 to residue Y89 was further confirmed by the abrogation of interaction between NS1 and p85β. Additionally, mAb 19H9 also detected NS1 proteins expressed in IAV-infected cells, showing NS1 intracellular localization in the cytoplasm and nucleolus. To our knowledge, mAb 19H9 is the first murine mAb to bind at the juxtaposition between the N-terminal RNA-binding domain and C-terminal effector domain of NS1. It could serve as a useful research tool for studying the conformational plasticity and dynamic changes in NS1.
Collapse
Affiliation(s)
- Su Hui Catherine Teo
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, MD4, 5 Science Drive 2, Singapore 117545
| | - Jian-Ping Wu
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, MD4, 5 Science Drive 2, Singapore 117545
| | - Chee-Keng Mok
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, MD4, 5 Science Drive 2, Singapore 117545
| | - Yee-Joo Tan
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, MD4, 5 Science Drive 2, Singapore 117545
- Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research), 61 Biopolis Dr, Singapore 138673
- Correspondence author: Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, MD4, 5 Science Drive 2, Singapore 117545. Tel: +65 65163692; E-mail:
| |
Collapse
|
24
|
Ng OW, Tan YJ. Understanding bat SARS-like coronaviruses for the preparation of future coronavirus outbreaks - Implications for coronavirus vaccine development. Hum Vaccin Immunother 2016; 13:186-189. [PMID: 27644155 PMCID: PMC5287300 DOI: 10.1080/21645515.2016.1228500] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The severe acute respiratory syndrome coronavirus (SARS-CoV) first emerged in 2003, causing the SARS epidemic which resulted in a 10% fatality rate. The advancements in metagenomic techniques have allowed the identification of SARS-like coronaviruses (SL-CoVs) sequences that share high homology to the human SARS-CoV epidemic strains from wildlife bats, presenting concrete evidence that bats are the origin and natural reservoir of SARS-CoV. The application of reverse genetics further enabled that characterization of these bat CoVs and the prediction of their potential to cause disease in humans. The knowledge gained from such studies is valuable in the surveillance and preparation of a possible future outbreak caused by a spill-over of these bat SL-CoVs.
Collapse
Affiliation(s)
- Oi-Wing Ng
- a Department of Microbiology and Immunology , Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore , Singapore
| | - Yee-Joo Tan
- a Department of Microbiology and Immunology , Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore , Singapore.,b Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research) , Singapore
| |
Collapse
|
25
|
Wycoff K, Maclean J, Belle A, Yu L, Tran Y, Roy C, Hayden F. Anti-infective immunoadhesins from plants. PLANT BIOTECHNOLOGY JOURNAL 2015; 13:1078-93. [PMID: 26242703 PMCID: PMC4749143 DOI: 10.1111/pbi.12441] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2015] [Revised: 06/24/2015] [Accepted: 06/27/2015] [Indexed: 05/22/2023]
Abstract
Immunoadhesins are recombinant proteins that combine the ligand-binding region of a receptor or adhesion molecule with immunoglobulin constant domains. All FDA-approved immunoadhesins are designed to modulate the interaction of a human receptor with its normal ligand, such as Etanercept (Enbrel(®) ), which interferes with the binding of tumour necrosis factor (TNF) to the TNF-alpha receptor and is used to treat inflammatory diseases such as rheumatoid arthritis. Like antibodies, immunoadhesins have long circulating half-lives, are readily purified by affinity-based methods and have the avidity advantages conferred by bivalency. Immunoadhesins that incorporate normal cellular receptors for viruses or bacterial toxins hold great, but as yet unrealized, potential for treating infectious disease. As decoy receptors, immunoadhesins have potential advantages over pathogen-targeted monoclonal antibodies. Planet Biotechnology has specialized in developing anti-infective immunoadhesins using plant expression systems. An immunoadhesin incorporating the cellular receptor for anthrax toxin, CMG2, potently blocks toxin activity in vitro and protects animals against inhalational anthrax. An immunoadhesin based on the receptor for human rhinovirus, ICAM-1, potently blocks infection of human cells by one of the major causes of the common cold. An immunoadhesin targeting the MERS coronavirus is in an early stage of development. We describe here the unique challenges involved in designing and developing immunoadhesins targeting infectious diseases in the hope of inspiring further research into this promising class of drugs.
Collapse
Affiliation(s)
| | | | | | - Lloyd Yu
- Planet Biotechnology Inc., Hayward, CA, USA
| | - Y Tran
- Planet Biotechnology Inc., Hayward, CA, USA
| | - Chad Roy
- Tulane National Primate Research Center, Covington, LA, USA
| | - Frederick Hayden
- University of Virginia School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
26
|
Lam S, Nyo M, Phuektes P, Yew CW, Tan YJ, Chu JJH. A potent neutralizing IgM mAb targeting the N218 epitope on E2 protein protects against Chikungunya virus pathogenesis. MAbs 2015; 7:1178-94. [PMID: 26305993 DOI: 10.1080/19420862.2015.1083664] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Chikungunya virus (CHIKV) is a medically important human viral pathogen that causes Chikungunya fever accompanied with debilitating and persistent joint pain. Host-elicited or passively-transferred monoclonal antibodies (mAb) are essential mediators of CHIKV clearance. Therefore, this study aimed to generate and characterize a panel of mAbs for their neutralization efficacy against CHIKV infection in a cell-based and murine model. To evaluate their antigenicity and neutralization profile, indirect enzyme-linked immunosorbent assay (ELISA), an immunofluorescence assay (IFA) and a plaque reduction neutralization test were performed on mAbs of IgM isotype. CHIKV escape mutants against mAb 3E7b neutralization were generated, and reverse genetics techniques were then used to create an infectious CHIKV clone with a single mutation. 3E7b was also administered to neonate mice prior or after CHIKV infection. The survival rate, CHIKV burden in tissues and histopathology of the limb muscles were evaluated. Both IgM 3E7b and 8A2c bind strongly to native CHIKV surface and potently neutralize CHIKV replication. Further analyses of 3E7b binding and neutralization of CHIKV single-mutant clones revealed that N218 of CHIKV E2 protein is a potent neutralizing epitope. In a pre-binding neutralization assay, 3E7b blocks CHIKV attachment to permissive cells, possibly by binding to the surface-accessible E2-N218 residue. Prophylactic administration of 3E7b to neonate mice markedly reduced viremia and protected against CHIKV pathogenesis in various mice tissues. Given therapeutically at 4 h post-infection, 3E7b conferred 100% survival rate and similarly reduced CHIKV load in most mice tissues except the limb muscles. Collectively, these findings highlight the usefulness of 3E7b for future prophylactic or epitope-based vaccine design.
Collapse
Affiliation(s)
- Shirley Lam
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Min Nyo
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Patchara Phuektes
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Chow Wenn Yew
- b Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research) ; Singapore
| | - Yee Joo Tan
- b Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research) ; Singapore.,c Hepatitis Viruses and Newly Emerging Viruses; Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore
| | - Justin Jang Hann Chu
- a Laboratory of Molecular RNA Virology and Antiviral Strategies, Department of Microbiology; Yong Loo Lin School of Medicine, National University Health System, National University of Singapore ; Singapore.,b Institute of Molecular and Cell Biology, A*STAR (Agency for Science, Technology and Research) ; Singapore
| |
Collapse
|
27
|
Aweya JJ, Sze CW, Bayega A, Mohd-Ismail NK, Deng L, Hotta H, Tan YJ. NS5B induces up-regulation of the BH3-only protein, BIK, essential for the hepatitis C virus RNA replication and viral release. Virology 2014; 474:41-51. [PMID: 25463603 PMCID: PMC7127593 DOI: 10.1016/j.virol.2014.10.027] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 10/24/2014] [Indexed: 12/14/2022]
Abstract
Hepatitis C virus (HCV) induces cytopathic effects in the form of hepatocytes apoptosis thought to be resulted from the interaction between viral proteins and host factors. Using pathway specific PCR array, we identified 9 apoptosis-related genes that are dysregulated during HCV infection, of which the BH3-only pro-apoptotic Bcl-2 family protein, BIK, was consistently up-regulated at the mRNA and protein levels. Depletion of BIK protected host cells from HCV-induced caspase-3/7 activation but not the inhibitory effect of HCV on cell viability. Furthermore, viral RNA replication and release were significantly suppressed in BIK-depleted cells and over-expression of the RNA-dependent RNA polymerase, NS5B, was able to induce BIK expression. Immunofluorescence and co-immunoprecipitation assays showed co-localization and interaction of BIK and NS5B, suggesting that BIK may be interacting with the HCV replication complex through NS5B. These results imply that BIK is essential for HCV replication and that NS5B is able to induce BIK expression.
Collapse
Affiliation(s)
- Jude Juventus Aweya
- Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Ching Wooen Sze
- Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore
| | - Anthony Bayega
- Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A⁎STAR), Singapore 138673, Singapore
| | - Nur Khairiah Mohd-Ismail
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A⁎STAR), Singapore 138673, Singapore
| | - Lin Deng
- Division of Microbiology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Hak Hotta
- Division of Microbiology, Kobe University Graduate School of Medicine, 7-5-1 Kusunoki-cho, Chuo-ku, Kobe, Hyogo 650-0017, Japan
| | - Yee-Joo Tan
- Department of Microbiology, Yong Loo Lin School of Medicine, National University Health System (NUHS), National University of Singapore, Singapore; Institute of Molecular and Cell Biology, Agency for Science, Technology and Research (A⁎STAR), Singapore 138673, Singapore.
| |
Collapse
|