101
|
Assembly and Cellular Exit of Coronaviruses: Hijacking an Unconventional Secretory Pathway from the Pre-Golgi Intermediate Compartment via the Golgi Ribbon to the Extracellular Space. Cells 2021; 10:cells10030503. [PMID: 33652973 PMCID: PMC7996754 DOI: 10.3390/cells10030503] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/18/2021] [Accepted: 02/18/2021] [Indexed: 12/20/2022] Open
Abstract
Coronaviruses (CoVs) assemble by budding into the lumen of the intermediate compartment (IC) at the endoplasmic reticulum (ER)-Golgi interface. However, why CoVs have chosen the IC as their intracellular site of assembly and how progeny viruses are delivered from this compartment to the extracellular space has remained unclear. Here we address these enigmatic late events of the CoV life cycle in light of recently described properties of the IC. Of particular interest are the emerging spatial and functional connections between IC elements and recycling endosomes (REs), defined by the GTPases Rab1 and Rab11, respectively. The establishment of IC-RE links at the cell periphery, around the centrosome and evidently also at the noncompact zones of the Golgi ribbon indicates that—besides traditional ER-Golgi communication—the IC also promotes a secretory process that bypasses the Golgi stacks, but involves its direct connection with the endocytic recycling system. The initial confinement of CoVs to the lumen of IC-derived large transport carriers and their preferential absence from Golgi stacks is consistent with the idea that they exit cells following such an unconventional route. In fact, CoVs may share this pathway with other intracellularly budding viruses, lipoproteins, procollagen, and/or protein aggregates experimentally introduced into the IC lumen.
Collapse
|
102
|
Majid S, Khan MS, Rashid S, Niyaz A, Farooq R, Bhat SA, Wani HA, Qureshi W. COVID-19: Diagnostics, Therapeutic Advances, and Vaccine Development. CURRENT CLINICAL MICROBIOLOGY REPORTS 2021; 8:152-166. [PMID: 33614398 PMCID: PMC7883962 DOI: 10.1007/s40588-021-00157-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2021] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW Human race is currently facing the wrath of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a highly transmittable and pathogenic RNA virus, causing coronavirus disease 2019 (COVID-19), the worst ever global pandemic. Coronaviruses (CoVs) have emerged as a major public health concern. Urgent global response to COVID-19 outbreak has been to limit spread of SARS-CoV-2 via extensive monitoring and containment. Various treatment regimens have been adopted to manage COVID-19, with known drugs and drug combinations used to decrease the morbidity and mortality associated with COVID-19. Intensive research on various fronts including studying molecular and structural aspects of these viruses and unraveling the pathophysiology and mechanistic basis of COVID-19 aimed at developing effective prophylactic, therapeutic agents and vaccines has been carried out globally. RECENT FINDINGS No approved antiviral treatment except remdesivir exists for SARS-CoV-2 till date though novel drug targets have been identified. However, worldwide frantic and competitive vaccine development pharmaceutical race has borne fruit in the form of a number of promising candidate vaccines, out of which few have already received emergency use authorization by regulatory bodies in record time. SUMMARY This review highlights the painstaking efforts of healthcare workers and scientific community to successfully address the COVID-19 pandemic-though damage in the form of severe illness, loss of lives, and livelihood has left a serious mark. Focusing on extensive research on various therapeutic options and antiviral strategies including neutralizing antibodies, potential drugs, and drug targets, light has been shed on various diagnostic options and the amazing vaccine development process as well.
Collapse
Affiliation(s)
- Sabhiya Majid
- Department of Biochemistry, Government Medical College Srinagar and Associated SMHS and Super Speciality Hospital and Research Centre, University of Kashmir Srinagar, Srinagar, J&K 190010 India
| | - Mosin S. Khan
- Department of Biochemistry, Government Medical College Srinagar and Associated SMHS and Super Speciality Hospital and Research Centre, University of Kashmir Srinagar, Srinagar, J&K 190010 India
| | - Samia Rashid
- Department of Medicine, Government Medical College Srinagar and Associated SMHS and Super Speciality Hospital, Srinagar, J&K 190010 India
| | - Ayesha Niyaz
- SHKM Government Medical College, Mewat, Haryana India
| | - Rabia Farooq
- Department of Basic Medical Sciences, College of Medicine, University of Bisha, Bisha, 67714 Saudi Arabia
| | - Showkat A. Bhat
- Department of Biochemistry, Government Medical College Doda, Doda, J&K 182202 India
| | - Hilal A. Wani
- Department of Higher Education, Government of Jammu & Kashmir, Jammu, India
| | - Waseem Qureshi
- Registrar Academics, Government Medical College Srinagar, Srinagar, J&K 190010 India
| |
Collapse
|
103
|
Choudhary J, Dheeman S, Sharma V, Katiyar P, Karn SK, Sarangi MK, Chauhan AK, Verma G, Baliyan N. Insights of Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2) pandemic: a current review. Biol Proced Online 2021; 23:5. [PMID: 33526007 PMCID: PMC7849622 DOI: 10.1186/s12575-020-00141-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/25/2020] [Indexed: 01/08/2023] Open
Abstract
COVID-19, a pandemic of the 21st century caused by novel coronavirus SARS-CoV-2 was originated from China and shallowed world economy and human resource. The medical cures via herbal treatments, antiviral drugs, and vaccines still in progress, and studying rigorously. SARS-CoV-2 is more virulent than its ancestors due to evolution in the spike protein(s), mediates viral attachment to the host's membranes. The SARS-CoV-2 receptor-binding spike domain associates itself with human angiotensin-converting enzyme 2 (ACE-2) receptors. It causes respiratory ailments with irregularities in the hepatic, nervous, and gastrointestinal systems, as reported in humans suffering from COVID-19 and reviewed in the present article. There are several approaches, have been put forward by many countries under the world health organization (WHO) recommendations and some trial drugs were introduced for possible treatment of COVID-19, such as Lopinavir or Ritonavir, Arbidol, Chloroquine (CQ), Hydroxychloroquine (HCQ) and most important Remdesivir including other like Tocilizumab, Oritavancin, Chlorpromazine, Azithromycin, Baricitinib, etc. RT-PCR is the only and early detection test available besides the rapid test kit (serodiagnosis) used by a few countries due to unreasonable causes. Development of vaccine by several leader of pharmaceutical groups still under trial or waiting for approval for mass inoculation. Management strategies have been evolved by the recommendations of WHO, specifically important to control COVID-19 situations, in the pandemic era. This review will provide a comprehensive collection of studies to support future research and enhancement in our wisdom to combat COVID-19 pandemic and to serve humanity.
Collapse
Affiliation(s)
- Jyoti Choudhary
- Department of Microbiology, Chinmaya Degree College (Hemwati Nandan Bahuguna Garhwal University, Srinagar, Garhwal, Uttarakhand), Haridwar, Uttarakhand 249401 India
- Department of Botany and Microbiology, Gurukula Kangri Deemed to be University, Haridwar, Uttarakhand 249404 India
| | - Shrivardhan Dheeman
- Department of Microbiology, School of Life Sciences, Sardar Bhagwan Singh University, Dehradun, Uttarakhand 248161 India
| | - Vipin Sharma
- Department of Pharmaceuticals Sciences, Faculty of Ayurvedic and Medicinal Sciences, Gurukula Kangri Deemed to be University, Haridwar, Uttarakhand 249404 India
| | - Prashant Katiyar
- Department of Botany and Microbiology, Gurukula Kangri Deemed to be University, Haridwar, Uttarakhand 249404 India
| | - Santosh Kumar Karn
- Deaprtment of Biotechnology and Biochemistry, School of Life Sciences, Sardar Bhagwan Singh University, Dehradun, Uttarakhand 248161 India
| | - Manoj Kumar Sarangi
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences and Technology, Sardar Bhagwan Singh University, Dehradun, Uttarakhand 248161 India
| | - Ankit Kumar Chauhan
- Department of Botany and Microbiology, Gurukula Kangri Deemed to be University, Haridwar, Uttarakhand 249404 India
- Atal Bihari Vajpayee Institute of Medical Sciences and Dr. Ram Manohar Lohia Hospital, New Delhi, 110001 India
| | - Gaurav Verma
- Deaprtment of Microbiology, Shri Dev Suman Subharti Medical College, Ras Bihari Bose Subharti University, Dehradun, Uttarakhand 248001 India
| | - Nitin Baliyan
- Department of Botany and Microbiology, Gurukula Kangri Deemed to be University, Haridwar, Uttarakhand 249404 India
| |
Collapse
|
104
|
Thankam FG, Agrawal DK. Molecular chronicles of cytokine burst in patients with coronavirus disease 2019 (COVID-19) with cardiovascular diseases. J Thorac Cardiovasc Surg 2021; 161:e217-e226. [PMID: 32631657 PMCID: PMC7834736 DOI: 10.1016/j.jtcvs.2020.05.083] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 05/12/2020] [Accepted: 05/16/2020] [Indexed: 02/02/2023]
Affiliation(s)
| | - Devendra K. Agrawal
- Address for reprints: Devendra K. Agrawal, PhD (Biochem), PhD (Med Sci), MBA, Department of Translational Research, Western University of Health Sciences, 309 E Second St, Pomona, CA 91766
| |
Collapse
|
105
|
Wong NA, Saier MH. The SARS-Coronavirus Infection Cycle: A Survey of Viral Membrane Proteins, Their Functional Interactions and Pathogenesis. Int J Mol Sci 2021; 22:1308. [PMID: 33525632 PMCID: PMC7865831 DOI: 10.3390/ijms22031308] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 01/21/2021] [Accepted: 01/22/2021] [Indexed: 02/07/2023] Open
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is a novel epidemic strain of Betacoronavirus that is responsible for the current viral pandemic, coronavirus disease 2019 (COVID-19), a global health crisis. Other epidemic Betacoronaviruses include the 2003 SARS-CoV-1 and the 2009 Middle East Respiratory Syndrome Coronavirus (MERS-CoV), the genomes of which, particularly that of SARS-CoV-1, are similar to that of the 2019 SARS-CoV-2. In this extensive review, we document the most recent information on Coronavirus proteins, with emphasis on the membrane proteins in the Coronaviridae family. We include information on their structures, functions, and participation in pathogenesis. While the shared proteins among the different coronaviruses may vary in structure and function, they all seem to be multifunctional, a common theme interconnecting these viruses. Many transmembrane proteins encoded within the SARS-CoV-2 genome play important roles in the infection cycle while others have functions yet to be understood. We compare the various structural and nonstructural proteins within the Coronaviridae family to elucidate potential overlaps and parallels in function, focusing primarily on the transmembrane proteins and their influences on host membrane arrangements, secretory pathways, cellular growth inhibition, cell death and immune responses during the viral replication cycle. We also offer bioinformatic analyses of potential viroporin activities of the membrane proteins and their sequence similarities to the Envelope (E) protein. In the last major part of the review, we discuss complement, stimulation of inflammation, and immune evasion/suppression that leads to CoV-derived severe disease and mortality. The overall pathogenesis and disease progression of CoVs is put into perspective by indicating several stages in the resulting infection process in which both host and antiviral therapies could be targeted to block the viral cycle. Lastly, we discuss the development of adaptive immunity against various structural proteins, indicating specific vulnerable regions in the proteins. We discuss current CoV vaccine development approaches with purified proteins, attenuated viruses and DNA vaccines.
Collapse
Affiliation(s)
- Nicholas A. Wong
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0116, USA
| | - Milton H. Saier
- Department of Molecular Biology, Division of Biological Sciences, University of California at San Diego, La Jolla, CA 92093-0116, USA
| |
Collapse
|
106
|
Dimonte S, Babakir-Mina M, Hama-Soor T, Ali S. Genetic Variation and Evolution of the 2019 Novel Coronavirus. Public Health Genomics 2021; 24:54-66. [PMID: 33406522 PMCID: PMC7900485 DOI: 10.1159/000513530] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 11/27/2020] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION SARS-CoV-2 is a new type of coronavirus causing a pandemic severe acute respiratory syndrome (SARS-2). Coronaviruses are very diverting genetically and mutate so often periodically. The natural selection of viral mutations may cause host infection selectivity and infectivity. METHODS This study was aimed to indicate the diversity between human and animal coronaviruses through finding the rate of mutation in each of the spike, nucleocapsid, envelope, and membrane proteins. RESULTS The mutation rate is abundant in all 4 structural proteins. The most number of statistically significant amino acid mutations were found in spike receptor-binding domain (RBD) which may be because it is responsible for a corresponding receptor binding in a broad range of hosts and host selectivity to infect. Among 17 previously known amino acids which are important for binding of spike to angiotensin-converting enzyme 2 (ACE2) receptor, all of them are conservative among human coronaviruses, but only 3 of them significantly are mutated in animal coronaviruses. A single amino acid aspartate-454, that causes dissociation of the RBD of the spike and ACE2, and F486 which gives the strength of binding with ACE2 remain intact in all coronaviruses. DISCUSSION/CONCLUSION Observations of this study provided evidence of the genetic diversity and rapid evolution of SARS-CoV-2 as well as other human and animal coronaviruses.
Collapse
Affiliation(s)
| | | | - Taib Hama-Soor
- Technical College of Health, Sulaimani Polytechnic University, KGR, Sulaimani, Iraq
| | - Salar Ali
- Technical College of Health, Sulaimani Polytechnic University, KGR, Sulaimani, Iraq
| |
Collapse
|
107
|
Ghosh A, Bhattacharyya C, Biswas NK, Das A. Underpinning the Rudimentary/Underlying Mechanisms Involved in the Pathogenesis of SARS-CoV-2 (COVID-19) in Human Lung Cells. TARGETING CELLULAR SIGNALLING PATHWAYS IN LUNG DISEASES 2021:537-557. [DOI: 10.1007/978-981-33-6827-9_25] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
108
|
Zhang XY, Guo J, Wan X, Zhou JG, Jin WP, Lu J, Wang WH, Yang AN, Liu DX, Shi ZL, Yuan ZM, Li XG, Meng SL, Duan K, Wang ZJ, Yang XM, Shen S. Biochemical and antigenic characterization of the structural proteins and their post-translational modifications in purified SARS-CoV-2 virions of an inactivated vaccine candidate. Emerg Microbes Infect 2020; 9:2653-2662. [PMID: 33232205 PMCID: PMC7738289 DOI: 10.1080/22221751.2020.1855945] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In the face of COVID-19 pandemic caused by the newly emerged SARS-CoV-2, an inactivated, Vero cell-based, whole virion vaccine candidate has been developed and entered into phase III clinical trials within six months. Biochemical and immunogenic characterization of structural proteins and their post-translational modifications in virions, the end-products of the vaccine candidate, would be essential for the quality control and process development of vaccine products and for studying the immunogenicity and pathogenesis of SARS-CoV-2. By using a panel of rabbit antisera against virions and five structural proteins together with a convalescent serum, the spike (S) glycoprotein was shown to be N-linked glycosylated, PNGase F-sensitive, endoglycosidase H-resistant and cleaved by Furin-like proteases into S1 and S2 subunits. The full-length S and S1/S2 subunits could form homodimers/trimers. The membrane (M) protein was partially N-linked glycosylated; the accessory protein 3a existed in three different forms, indicative of cleavage and dimerization. Furthermore, analysis of the antigenicity of these proteins and their post-translationally modified forms demonstrated that S protein induced the strongest antibody response in both convalescent and immunized animal sera. Interestingly, immunization with the inactivated vaccine did not elicit antibody response against the S2 subunit, whereas strong antibody response against both S1 and S2 subunits was detected in the convalescent serum. Moreover, vaccination stimulated stronger antibody response against S multimers than did the natural infection. This study revealed that the native S glycoprotein stimulated neutralizing antibodies, while bacterially-expressed S fragments did not. The study on S modifications would facilitate design of S-based anti-SARS-CoV-2 vaccines.
Collapse
Affiliation(s)
- Xiao-Yu Zhang
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Jing Guo
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Xin Wan
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Jin-Ge Zhou
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Wei-Ping Jin
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Jia Lu
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Wen-Hui Wang
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - An-Na Yang
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Ding Xiang Liu
- South China Agricultural University, Guangdong Province Key Laboratory Microbial Signals & Disease Co, & Integrative Microbiology Research Center, Guangzhou, People's Republic of China
| | - Zheng-Li Shi
- Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Zhi-Ming Yuan
- Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, People's Republic of China
| | - Xin-Guo Li
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Sheng-Li Meng
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Kai Duan
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Ze-Jun Wang
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| | - Xiao-Ming Yang
- China National Biotech Group Company Ltd, Beijing, People's Republic of China
| | - Shuo Shen
- Wuhan Institute of Biological Products, Co. Ltd, Wuhan, People's Republic of China
| |
Collapse
|
109
|
Stavropoulou E, Bezirtzoglou E. Probiotics as a Weapon in the Fight Against COVID-19. Front Nutr 2020; 7:614986. [PMID: 33385008 PMCID: PMC7769760 DOI: 10.3389/fnut.2020.614986] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Affiliation(s)
- Elisavet Stavropoulou
- Department of Medicine, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland.,Service of Infectious Diseases, Central Institute of Valais Hospitals, Sion, Switzerland.,Department of Medicine, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Eugenia Bezirtzoglou
- Laboratory of Hygiene and Environmental Protection, Medical School, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
110
|
Mandala VS, McKay MJ, Shcherbakov AA, Dregni AJ, Kolocouris A, Hong M. Structure and drug binding of the SARS-CoV-2 envelope protein transmembrane domain in lipid bilayers. Nat Struct Mol Biol 2020; 27:1202-1208. [PMID: 33177698 PMCID: PMC7718435 DOI: 10.1038/s41594-020-00536-8] [Citation(s) in RCA: 276] [Impact Index Per Article: 55.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Accepted: 10/28/2020] [Indexed: 12/19/2022]
Abstract
An essential protein of the SARS-CoV-2 virus, the envelope protein E, forms a homopentameric cation channel that is important for virus pathogenicity. Here we report a 2.1-Å structure and the drug-binding site of E's transmembrane domain (ETM), determined using solid-state NMR spectroscopy. In lipid bilayers that mimic the endoplasmic reticulum-Golgi intermediate compartment (ERGIC) membrane, ETM forms a five-helix bundle surrounding a narrow pore. The protein deviates from the ideal α-helical geometry due to three phenylalanine residues, which stack within each helix and between helices. Together with valine and leucine interdigitation, these cause a dehydrated pore compared with the viroporins of influenza viruses and HIV. Hexamethylene amiloride binds the polar amino-terminal lumen, whereas acidic pH affects the carboxy-terminal conformation. Thus, the N- and C-terminal halves of this bipartite channel may interact with other viral and host proteins semi-independently. The structure sets the stage for designing E inhibitors as antiviral drugs.
Collapse
Affiliation(s)
- Venkata S Mandala
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Matthew J McKay
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Aurelio J Dregni
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Antonios Kolocouris
- Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, Athens, Greece
| | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
111
|
Yadav M, Dhagat S, Eswari JS. Emerging strategies on in silico drug development against COVID-19: challenges and opportunities. Eur J Pharm Sci 2020; 155:105522. [PMID: 32827661 PMCID: PMC7438372 DOI: 10.1016/j.ejps.2020.105522] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 08/14/2020] [Accepted: 08/18/2020] [Indexed: 12/22/2022]
Abstract
The importance of coronaviruses as human pathogen has been highlighted by the recent outbreak of SARS-CoV-2 leading to the search of suitable drugs to overcome respiratory infections caused by the virus. Due to the lack of specific drugs against coronavirus, the existing antiviral and antimalarial drugs are currently being administered to the patients infected with SARS-CoV-2. The scientists are also considering repurposing of some of the existing drugs as a suitable option in search of effective drugs against coronavirus till the establishment of a potent drug and/or vaccine. Computer-aided drug discovery provides a promising attempt to enable scientists to develop new and target specific drugs to combat any disease. The discovery of novel targets for COVID-19 using computer-aided drug discovery tools requires knowledge of the structure of coronavirus and various target proteins present in the virus. Targeting viral proteins will make the drug specific against the virus, thereby, increasing the chances of viral mortality. Hence, this review provides the structure of SARS-CoV-2 virus along with the important viral components involved in causing infection. It also focuses on the role of various target proteins in disease, the mechanism by which currently administered drugs act against the virus and the repurposing of few drugs. The gap arising from the absence of specific drugs is addressed by proposing potential antiviral drug targets which might provide insights into structure-based drug development against SARS-CoV-2.
Collapse
Affiliation(s)
- Manisha Yadav
- Department of Biotechnology, National Institute of Technology Raipur, C.G., 492010, India
| | - Swasti Dhagat
- Department of Biotechnology, National Institute of Technology Raipur, C.G., 492010, India
| | - J Satya Eswari
- Department of Biotechnology, National Institute of Technology Raipur, C.G., 492010, India.
| |
Collapse
|
112
|
Yu KK, Fischinger S, Smith MT, Atyeo C, Cizmeci D, Wolf CR, Layton ED, Logue JK, Aguilar MS, Shuey K, Loos C, Yu J, Franko N, Choi RY, Wald A, Barouch DH, Koelle DM, Lauffenburger D, Chu HY, Alter G, Seshadri C. T cell and antibody functional correlates of severe COVID-19. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.11.25.20235150. [PMID: 33269369 PMCID: PMC7709190 DOI: 10.1101/2020.11.25.20235150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Comorbid medical illnesses, such as obesity and diabetes, are associated with more severe COVID-19, hospitalization, and death. However, the role of the immune system in mediating these clinical outcomes has not been determined. We used multi-parameter flow cytometry and systems serology to comprehensively profile the functions of T cells and antibodies targeting spike, nucleocapsid, and envelope proteins in a convalescent cohort of COVID-19 subjects who were either hospitalized (n=20) or not hospitalized (n=40). To avoid confounding, subjects were matched by age, sex, ethnicity, and date of symptom onset. Surprisingly, we found that the magnitude and functional breadth of virus-specific CD4 T cell and antibody responses were consistently higher among hospitalized subjects, particularly those with medical comorbidities. However, an integrated analysis identified more coordination between polyfunctional CD4 T-cells and antibodies targeting the S1 domain of spike among subjects that were not hospitalized. These data reveal a functionally diverse and coordinated response between T cells and antibodies targeting SARS-CoV-2 which is reduced in the presence of comorbid illnesses that are known risk factors for severe COVID-19. Our data suggest that isolated measurements of the magnitudes of spike-specific immune responses are likely insufficient to anticipate vaccine efficacy in high-risk populations.
Collapse
Affiliation(s)
- Krystle K.Q. Yu
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Stephanie Fischinger
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
- PhD program in Immunology and Virology, University of Duisburg-Essen, Essen, Germany
| | - Malisa T. Smith
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Caroline Atyeo
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
- PhD program in Virology, Division of Medical Sciences, Harvard University, Boston, MA, USA
| | - Deniz Cizmeci
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Caitlin R. Wolf
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Erik D. Layton
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Jennifer K. Logue
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Melissa S. Aguilar
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Kiel Shuey
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Carolin Loos
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jingyou Yu
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Nicholas Franko
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | | | - Anna Wald
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Department of Epidemiology, University of Washington School of Public Health, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Dan H. Barouch
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - David M. Koelle
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington School of Medicine, Seattle, WA, USA
- Vaccine and Infectious Diseases Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Benaroya Research Institute, Seattle, WA, USA
| | - Douglas Lauffenburger
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Helen Y. Chu
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Boston, MA, USA
| | - Chetan Seshadri
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
113
|
Rajarshi K, Khan R, Singh MK, Ranjan T, Ray S, Ray S. Essential functional molecules associated with SARS-CoV-2 infection: Potential therapeutic targets for COVID-19. Gene 2020; 768:145313. [PMID: 33220345 PMCID: PMC7673215 DOI: 10.1016/j.gene.2020.145313] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023]
Abstract
The whole world is still suffering substantially from the coronavirus disease 2019 (COVID-19) outbreak. Several protein-based molecules that are associated with the Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which are essential for its functionality, survival, and pathogenesis have been identified and are considered as potential therapeutic targets. These protein-based molecules are either structural/non-structural components of SARS-CoV-2 or host factors, which play a crucial role in this infection. Developing drug molecules against these essential functional molecules to hinder their regular functioning and associated physiological pathways could be promising for successful clinical management of this novel coronavirus infection. The review aims to highlight the functional molecules that play crucial roles in SARS-CoV-2 pathogenesis. We have emphasized how these potential druggable targets could be beneficial in tackling the COVID-19 crisis.
Collapse
Affiliation(s)
- Keshav Rajarshi
- School of Community Science and Technology (SOCSAT), Indian Institute of Engineering Science and Technology (IIEST), Shibpur, Howrah, West Bengal 711103, India
| | - Rajni Khan
- Motihari College of Engineering, Motihari 845401, India
| | | | - Tushar Ranjan
- Department of Molecular Biology and Genetic Engineering, Bihar Agriculture University, Sabour, Bhagalpur, India.
| | - Sandipan Ray
- Department of Systems Pharmacology & Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Shashikant Ray
- Department of Biotechnology, Mahatma Gandhi Central University Motihari, 845401, India.
| |
Collapse
|
114
|
Silva LR, da Silva Santos-Júnior PF, de Andrade Brandão J, Anderson L, Bassi ÊJ, Xavier de Araújo-Júnior J, Cardoso SH, da Silva-Júnior EF. Druggable targets from coronaviruses for designing new antiviral drugs. Bioorg Med Chem 2020; 28:115745. [PMID: 33007557 PMCID: PMC7836322 DOI: 10.1016/j.bmc.2020.115745] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/26/2020] [Accepted: 08/29/2020] [Indexed: 01/18/2023]
Abstract
Severe respiratory infections were highlighted in the SARS-CoV outbreak in 2002, as well as MERS-CoV, in 2012. Recently, the novel CoV (COVID-19) has led to severe respiratory damage to humans and deaths in Asia, Europe, and Americas, which allowed the WHO to declare the pandemic state. Notwithstanding all impacts caused by Coronaviruses, it is evident that the development of new antiviral agents is an unmet need. In this review, we provide a complete compilation of all potential antiviral agents targeting macromolecular structures from these Coronaviruses (Coronaviridae), providing a medicinal chemistry viewpoint that could be useful for designing new therapeutic agents.
Collapse
Affiliation(s)
- Leandro Rocha Silva
- Chemistry and Biotechnology Institute, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, Brazil; Laboratory of Organic and Medicinal Synthesis, Federal University of Alagoas, Campus Arapiraca, Manoel Severino Barbosa Avenue, Arapiraca 57309-005, Brazil
| | | | - Júlia de Andrade Brandão
- IMUNOREG - Immunoregulation Research Group, Laboratory of Research in Virology and Immunology, Institute of Biological Sciences and Health, Federal University of Alagoas, Campus AC. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, Brazil
| | - Letícia Anderson
- IMUNOREG - Immunoregulation Research Group, Laboratory of Research in Virology and Immunology, Institute of Biological Sciences and Health, Federal University of Alagoas, Campus AC. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, Brazil; CESMAC University Center, Cônego Machado Street, Maceió 57051-160, Brazil
| | - Ênio José Bassi
- IMUNOREG - Immunoregulation Research Group, Laboratory of Research in Virology and Immunology, Institute of Biological Sciences and Health, Federal University of Alagoas, Campus AC. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, Brazil
| | - João Xavier de Araújo-Júnior
- Chemistry and Biotechnology Institute, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, Brazil; Laboratory of Medicinal Chemistry, Pharmaceutical Sciences Institute, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, Brazil
| | - Sílvia Helena Cardoso
- Laboratory of Organic and Medicinal Synthesis, Federal University of Alagoas, Campus Arapiraca, Manoel Severino Barbosa Avenue, Arapiraca 57309-005, Brazil
| | - Edeildo Ferreira da Silva-Júnior
- Chemistry and Biotechnology Institute, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, Brazil; Laboratory of Medicinal Chemistry, Pharmaceutical Sciences Institute, Federal University of Alagoas, Campus A.C. Simões, Lourival Melo Mota Avenue, Maceió 57072-970, Brazil.
| |
Collapse
|
115
|
Marak BN, Dowarah J, Khiangte L, Singh VP. Step toward repurposing drug discovery for COVID-19 therapeutics through in silico approach. Drug Dev Res 2020; 82:374-392. [PMID: 33170521 DOI: 10.1002/ddr.21757] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/06/2020] [Accepted: 09/08/2020] [Indexed: 12/15/2022]
Abstract
The outbreak of SARS-CoV-2 has become a threat to global health and has led to a global economic crisis. Although the researchers worldwide are putting tremendous effort toward gaining more insights into this zoonotic virus and developing vaccines and therapeutic drugs, no vaccine or drug is yet available to combat COVID-19 effectively. Drug discovery is often a laborious, time-consuming, and expensive task. In this time of crisis, employing computational methods could provide a feasible alternative approach that can potentially be used for drug discovery. Therefore, a library of several antiparasitic and anti-inflammatory drugs was virtually screened against SARS-CoV-2 proteases to identify potential inhibitors. The identified inhibitory drugs were further analyzed to confirm their activities against SARS-CoV-2. Our results could prove to be helpful in repurposing the drug discovery approach, which could substantially reduce the expenses, time, and resources required.
Collapse
Affiliation(s)
- Brilliant N Marak
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl, Mizoram, India
| | - Jayanta Dowarah
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl, Mizoram, India
| | - Laldingluaia Khiangte
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl, Mizoram, India
| | - Ved Prakash Singh
- Department of Chemistry, School of Physical Sciences, Mizoram University, Aizawl, Mizoram, India
| |
Collapse
|
116
|
Hassan SS, Choudhury PP, Roy B. Molecular phylogeny and missense mutations at envelope proteins across coronaviruses. Genomics 2020; 112:4993-5004. [PMID: 32927009 PMCID: PMC7486180 DOI: 10.1016/j.ygeno.2020.09.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/03/2020] [Indexed: 12/18/2022]
Abstract
Envelope (E) protein is one of the structural viroporins (76-109 amino acids long) present in the coronavirus. Sixteen sequentially different E-proteins were observed from a total of 4917 available complete SARS-CoV-2 genomes as on 18th June 2020 in the NCBI database. The missense mutations over the envelope protein across various coronaviruses of the β-genus were analyzed to know the immediate parental origin of the envelope protein of SARS-CoV-2. The evolutionary origin is also endorsed by the phylogenetic analysis of the envelope proteins comparing sequence homology as well as amino acid conservations.
Collapse
Affiliation(s)
- Sk Sarif Hassan
- Department of Mathematics, Pingla Thana Mahavidyalaya, Maligram, Paschim Medinipur, 721140, West Bengal, India.
| | - Pabitra Pal Choudhury
- Applied Statistics Unit, Indian Statistical Institute, Kolkata 700108, West Bengal, India
| | - Bidyut Roy
- Human Genetics Unit, Indian Statistical Institute, Kolkata 700108, West Bengal, India.
| |
Collapse
|
117
|
Akhand MRN, Azim KF, Hoque SF, Moli MA, Joy BD, Akter H, Afif IK, Ahmed N, Hasan M. Genome based evolutionary lineage of SARS-CoV-2 towards the development of novel chimeric vaccine. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2020; 85:104517. [PMID: 32882432 PMCID: PMC7462568 DOI: 10.1016/j.meegid.2020.104517] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 07/28/2020] [Accepted: 08/25/2020] [Indexed: 02/07/2023]
Abstract
The present study aimed to predict a novel chimeric vaccine by simultaneously targeting four major structural proteins via the establishment of ancestral relationship among different strains of coronaviruses. Conserved regions from the homologous protein sets of spike glycoprotein, membrane protein, envelope protein and nucleocapsid protein were identified through multiple sequence alignment. The phylogeny analyses of whole genome stated that four proteins reflected the close ancestral relation of SARS-CoV-2 to SARS-COV-1 and bat coronavirus. Numerous immunogenic epitopes (both T cell and B cell) were generated from the common fragments which were further ranked on the basis of antigenicity, transmembrane topology, conservancy level, toxicity and allergenicity pattern and population coverage analysis. Top putative epitopes were combined with appropriate adjuvants and linkers to construct a novel multiepitope subunit vaccine against COVID-19. The designed constructs were characterized based on physicochemical properties, allergenicity, antigenicity and solubility which revealed the superiority of construct V3 in terms safety and efficacy. Essential molecular dynamics and normal mode analysis confirmed minimal deformability of the refined model at molecular level. In addition, disulfide engineering was investigated to accelerate the stability of the protein. Molecular docking study ensured high binding affinity between construct V3 and HLA cells, as well as with different host receptors. Microbial expression and translational efficacy of the constructs were checked using pET28a(+) vector of E. coli strain K12. However, the in vivo and in vitro validation of suggested vaccine molecule might be ensured with wet lab trials using model animals for the implementation of the presented data.
Collapse
Affiliation(s)
- Mst Rubaiat Nazneen Akhand
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh,Department of Biochemistry and Chemistry, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Kazi Faizul Azim
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh,Department of Microbial Biotechnology, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Syeda Farjana Hoque
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh,Department of Pharmaceuticals and Industrial Biotechnology, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Mahmuda Akther Moli
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh,Department of Pharmaceuticals and Industrial Biotechnology, Sylhet Agricultural University, Sylhet 3100, Bangladesh
| | - Bijit Das Joy
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh,Department of Biochemistry and Chemistry, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Hafsa Akter
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Ibrahim Khalil Afif
- Department of Biotechnology and Genetic Engineering, Noakhali Science and Technology University, Noakhali, Bangladesh
| | - Nadim Ahmed
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh
| | - Mahmudul Hasan
- Faculty of Biotechnology and Genetic Engineering, Sylhet Agricultural University, Sylhet, 3100, Bangladesh; Department of Pharmaceuticals and Industrial Biotechnology, Sylhet Agricultural University, Sylhet 3100, Bangladesh.
| |
Collapse
|
118
|
Saurabh K, Solovchuk M, Sheu TWH. Investigating ion transport inside the pentameric ion channel encoded in COVID-19 E protein. Phys Rev E 2020; 102:052408. [PMID: 33327170 DOI: 10.1103/physreve.102.052408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 10/16/2020] [Indexed: 11/07/2022]
Abstract
Ion flow inside an ion channel can be described through continuum based Born-Poisson-Nernst-Planck (BPNP) equations in conjunction with the Lennard-Jones potential. Keeping in mind the ongoing pandemic, in this study, an attempt has been made to understand the selectivity and the current voltage relation of the COVID-19 E protein pentameric ion channel. Two ionic species, namely Na^{+} and Cl^{-}, have been considered here. E protein is one of the smallest structural protein which is embedded in the outer membrane of the virus. Once the virus is inside the host cell, this protein is expressed abundantly and is responsible for activities such as replication and budding of the virus. In the literature, we can find a few experimental studies focusing on understanding the activity of the channel formed by E proteins of different viruses. Here, we attempt the same study for the COVID-19 E protein ion channel through mathematical modeling. The channel geometry is calculated from the protein data bank file which was provided by NARLabs, Taiwan, using the hole program. Further, it was used to obtain the charge distribution using the pdbtopqr online program. The immersed boundary-lattice Boltzmann method (IB-LBM) has been implemented to numerically solve the system of equations in the channel generated by the protein data bank file. Further, an in-house code which operates on multiple GPUs and uses the cuda platform has been developed to achieve the goal of performing the current investigation.
Collapse
Affiliation(s)
- Kumar Saurabh
- Department of Engineering Science and Ocean Engineering, National Taiwan University, Taipei 10617, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 35053, Taiwan
- NTU High Performance and Scientific Computing Center, National Taiwan University, Taipei 10617, Taiwan
| | - Maxim Solovchuk
- Department of Engineering Science and Ocean Engineering, National Taiwan University, Taipei 10617, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Tony Wen-Hann Sheu
- Department of Engineering Science and Ocean Engineering, National Taiwan University, Taipei 10617, Taiwan
- Center for Advanced Study in Theoretical Sciences (CASTS), National Taiwan University, Taipei 10617, Taiwan
- NTU High Performance and Scientific Computing Center, National Taiwan University, Taipei 10617, Taiwan
| |
Collapse
|
119
|
Dey D, Borkotoky S, Banerjee M. In silico identification of Tretinoin as a SARS-CoV-2 envelope (E) protein ion channel inhibitor. Comput Biol Med 2020; 127:104063. [PMID: 33126128 PMCID: PMC7574788 DOI: 10.1016/j.compbiomed.2020.104063] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/25/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022]
Abstract
Viroporins are oligomeric, pore forming, viral proteins that play critical roles in the life cycle of pathogenic viruses. Viroporins like HIV-1 Vpu, Alphavirus 6 K, Influenza M2, HCV p7, and Picornavirus 2B, form discrete aqueous passageways which mediate ion and small molecule transport in infected cells. The alterations in host membrane structures induced by viroporins is essential for key steps in the virus life cycle like entry, replication and egress. Any disruption in viroporin functionality severely compromises viral pathogenesis. The envelope (E) protein encoded by coronaviruses is a viroporin with ion channel activity and has been shown to be crucial for the assembly and pathophysiology of coronaviruses. We used a combination of virtual database screening, molecular docking, all-atom molecular dynamics simulation and MM-PBSA analysis to test four FDA approved drugs - Tretinoin, Mefenamic Acid, Ondansetron and Artemether - as potential inhibitors of ion channels formed by SARS-CoV-2 E protein. Interaction and binding energy analysis showed that electrostatic interactions and polar solvation energy were the major driving forces for binding of the drugs, with Tretinoin being the most promising inhibitor. Tretinoin bound within the lumen of the channel formed by E protein, which is lined by hydrophobic residues like Phe, Val and Ala, indicating its potential for blocking the channel and inhibiting the viroporin functionality of E. In control simulations, tretinoin demonstrated a lower binding energy with a known target as compared to SARS-CoV-2 E protein. This work thus highlights the possibility of exploring Tretinoin as a potential SARS-CoV-2 E protein ion channel blocker and virus assembly inhibitor, which could be an important therapeutic strategy in the treatment for coronaviruses. Screened a database of 1806 small molecule compounds from DrugBank for potential SARS CoV-2 E protein inhibitors. Tested four FDA-approved drugs Tetinoin, Mefenamic acid, Ondansetron and Artemether as SARS CoV-2 E protein inhibitors. Used a combination of molecular docking, MD simulations and MM-PBSA analysis to propose Tretinoin as a promising inhibitor.
Collapse
Affiliation(s)
- Debajit Dey
- School of Medicine, University of Maryland Baltimore, United States
| | - Subhomoi Borkotoky
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, India
| | - Manidipa Banerjee
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, India.
| |
Collapse
|
120
|
McClenaghan C, Hanson A, Lee SJ, Nichols CG. Coronavirus Proteins as Ion Channels: Current and Potential Research. Front Immunol 2020; 11:573339. [PMID: 33154751 PMCID: PMC7586316 DOI: 10.3389/fimmu.2020.573339] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 09/22/2020] [Indexed: 01/22/2023] Open
Abstract
Coronavirus (CoV) outbreaks have recently emerged as a global public health threat due to their exceptional zoonotic potential — a feature arising from their ability to infect a diverse range of potential hosts combined with their high capacity for mutation and recombination. After Severe Acute Respiratory Syndrome (SARS) CoV-1 in 2003 and Middle East Respiratory Syndrome (MERS) CoV in 2012, with the current SARS-CoV-2 pandemic we are now in the midst of the third deadly international CoV outbreak in less than 20 years. Coronavirus outbreaks present a critical threat to global public health and an urgent necessity for therapeutic options. Here, we critically examine the current evidence for ion channel activity in CoV proteins and the potential for modulation as a therapeutic approach.
Collapse
Affiliation(s)
- Conor McClenaghan
- Center for Investigation of Membrane Excitability Diseases, and Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Alex Hanson
- Center for Investigation of Membrane Excitability Diseases, and Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Sun-Joo Lee
- Center for Investigation of Membrane Excitability Diseases, and Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
| | - Colin G Nichols
- Center for Investigation of Membrane Excitability Diseases, and Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, United States
| |
Collapse
|
121
|
Chakhalian D, Shultz RB, Miles CE, Kohn J. Opportunities for biomaterials to address the challenges of COVID-19. J Biomed Mater Res A 2020; 108:1974-1990. [PMID: 32662571 PMCID: PMC7405498 DOI: 10.1002/jbm.a.37059] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 07/10/2020] [Indexed: 12/19/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has revealed major shortcomings in our ability to mitigate transmission of infectious viral disease and provide treatment to patients, resulting in a public health crisis. Within months of the first reported case in China, the virus has spread worldwide at an unprecedented rate. COVID-19 illustrates that the biomaterials community was engaged in significant research efforts against bacteria and fungi with relatively little effort devoted to viruses. Accordingly, biomaterials scientists and engineers will have to participate in multidisciplinary antiviral research over the coming years. Although tissue engineering and regenerative medicine have historically dominated the field of biomaterials, current research holds promise for providing transformative solutions to viral outbreaks. To facilitate collaboration, it is imperative to establish a mutual language and adequate understanding between clinicians, industry partners, and research scientists. In this article, clinical perspectives are shared to clearly define emerging healthcare needs that can be met by biomaterials solutions. Strategies and opportunities for novel biomaterials intervention spanning diagnostics, treatment strategies, vaccines, and virus-deactivating surface coatings are discussed. Ultimately this review serves as a call for the biomaterials community to become a leading contributor to the prevention and management of the current and future viral outbreaks.
Collapse
Affiliation(s)
- Daniel Chakhalian
- Department of Chemistry and Chemical BiologyRutgers – The State University of New JerseyPiscatawayNew JerseyUSA
| | - Robert B. Shultz
- Department of Chemistry and Chemical BiologyRutgers – The State University of New JerseyPiscatawayNew JerseyUSA
- Department of NeurosurgeryUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
- Department of BioengineeringUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Catherine E. Miles
- Department of Chemistry and Chemical BiologyRutgers – The State University of New JerseyPiscatawayNew JerseyUSA
| | - Joachim Kohn
- Department of Chemistry and Chemical BiologyRutgers – The State University of New JerseyPiscatawayNew JerseyUSA
| |
Collapse
|
122
|
Ojha S, Chadha H, Mahor S. COVID-19: Pathogenesis and Pharmacological Basis for Use of Passive Antibody Therapy. CURRENT DRUG THERAPY 2020. [DOI: 10.2174/1574885515999200813193747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background:
Coronaviruses (CoVs), having enveloped RNA of positive strand, are
mainly responsible for enzootic infections in mammals. The mortality of CoVs has been proved as
they can cross the species barrier very easily and infect humans. Most recently, the outbreak of
coronavirus induced COVID-19 emerged in the city of Wuhan, Hubei province of China and became
the third highly pathogenic coronavirus infecting nearly 230 countries.
Objective:
To review the literature available about pathogenic Coronavirures with emphasis on
pathogenesis of COVID-19, and passive antibody therapy prospective.
Methods:
This study reviewed relevant published literature to provide (1) structural similarities
between coronaviruses and therapeutic methodologies used on SARS-CoV, MERS treatment which
might help scientists in understanding novel COVID-19 infection, (2) understanding COVID-19
pathogenesis that may help in identification of appropriate therapeutic targets to develop specific
and effective anti-viral drugs as well as immunizing agents against this novel emerging pathogen
and (3) to discuss existing knowledge on the passive immune therapy against similar coronaviruses
SARS-CoV and MERS-CoV with emphasis on COVID-19 pandemic treatment.
Conclusion:
COVID 19 coronavirus has shown resemblance to viral infections like SARS-CoV,
MERS infection. Historically, it has been proved that the prevention of disease, when exposed to a
biological system, is mainly a function of the immune response of that infected individual. To fight
against these infections, passive antibody therapy is the only available countermeasure that could
provide immediate immunity against infection. Passive antibody results in protection irrespective of
the immune status of the host. This therapy can be advantageous in countering the biological attack,
post exposure preventions, low toxicity and peculiar activity.
Collapse
Affiliation(s)
- Smriti Ojha
- Faculty of Pharmacy, Sardar Patel College of Pharmacy, Gorakhpur, Uttar Pradesh, India
| | - Hina Chadha
- Department of Pharmaceutical Sciences, Vishveshwarya Group of Institutions, Greater Noida, Uttar Pradesh, India
| | - Seema Mahor
- Department of Pharmaceutical Sciences, Vishveshwarya Group of Institutions, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
123
|
Lin P, Wang M, Wei Y, Kim T, Wei X. Coronavirus in human diseases: Mechanisms and advances in clinical treatment. MedComm (Beijing) 2020; 1:270-301. [PMID: 33173860 PMCID: PMC7646666 DOI: 10.1002/mco2.26] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 07/20/2020] [Accepted: 07/21/2020] [Indexed: 02/05/2023] Open
Abstract
Coronaviruses (CoVs), a subfamily of coronavirinae, are a panel of single-stranded RNA virus. Human coronavirus (HCoV) strains (HCoV-229E, HCoV-OC43, HCoV-HKU1, HCoV-NL63) usually cause mild upper respiratory diseases and are believed to be harmless. However, other HCoVs, associated with severe acute respiratory syndrome, Middle East respiratory syndrome, and COVID-19, have been identified as important pathogens due to their potent infectivity and lethality worldwide. Moreover, currently, no effective antiviral drugs treatments are available so far. In this review, we summarize the biological characters of HCoVs, their association with human diseases, and current therapeutic options for the three severe HCoVs. We also highlight the discussion about novel treatment strategies for HCoVs infections.
Collapse
Affiliation(s)
- Panpan Lin
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy and Cancer Center National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu China
| | - Manni Wang
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy and Cancer Center National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy and Cancer Center National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu China
| | - Taewan Kim
- Wexner Medical Center The Ohio State University Columbus Ohio 43210 USA
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target State Key Laboratory of Biotherapy and Cancer Center National Clinical Research Center for Geriatrics West China Hospital Sichuan University Chengdu China
| |
Collapse
|
124
|
Asghari A, Naseri M, Safari H, Saboory E, Parsamanesh N. The Novel Insight of SARS-CoV-2 Molecular Biology and Pathogenesis and Therapeutic Options. DNA Cell Biol 2020; 39:1741-1753. [PMID: 32716648 DOI: 10.1089/dna.2020.5703] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
On December 31, 2019, a novel coronavirus, being the third highly infective CoV and named as coronavirus disease 2019 (COVID-19) in the city of Wuhan, was announced by the World Health Organization. COVID-19 has a 2% mortality rate, is known as the third extremely infective CoV infection, and has a mortality rate less than MERS-CoV and SARS-CoV. The CoV family comprises a chief number of positive single-stranded ss (+) RNA viruses that are recognized in mammals. The 2019-nCoV patients showed that the angiotensin-converting enzyme II (ACE2) was the same for SARS-CoV. Structural proteins have an essential role in virus released and budding to various host cells. Notably, evidence indicated human-to-human transmission, along with several exported patients of virus infection worldwide. Nowadays, no licensed antivirals drugs or vaccines for being utilized against these coronavirus infections are recognized. There is an urgent requirement for an extensive research of CoV infections to disclose the route of extension, pathogenesis, and diagnosis and then to recognize the therapeutic targets to facilitate disease control and surveillance. In this article, we present an overview of the common biological criteria of CoVs and explain pathogenesis with a focus on the therapeutic approach to suggest potential goals for treating and monitoring this emerging zoonotic disease.
Collapse
Affiliation(s)
- Arghavan Asghari
- Student Research Committee and Birjand University of Medical Sciences, Birjand, Iran
| | - Mohsen Naseri
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Hamidreza Safari
- Department of Immunology, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran
| | - Ehsan Saboory
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Negin Parsamanesh
- Zanjan Metabolic Diseases Research Center, Zanjan University of Medical Sciences, Zanjan, Iran
| |
Collapse
|
125
|
Patel CN, Kumar SP, Pandya HA, Rawal RM. Identification of potential inhibitors of coronavirus hemagglutinin-esterase using molecular docking, molecular dynamics simulation and binding free energy calculation. Mol Divers 2020; 25:421-433. [PMID: 32996011 PMCID: PMC7524381 DOI: 10.1007/s11030-020-10135-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 08/18/2020] [Indexed: 02/07/2023]
Abstract
Abstract The pandemic outbreak of the Corona viral infection has become a critical global health issue. Biophysical and structural evidence shows that spike protein possesses a high binding affinity towards host angiotensin-converting enzyme 2 and viral hemagglutinin-acetylesterase (HE) glycoprotein receptor. We selected HE as a target in this study to identify potential inhibitors using a combination of various computational approaches such as molecular docking, ADMET analysis, dynamics simulations and binding free energy calculations. Virtual screening of NPACT compounds identified 3,4,5-Trihydroxy-1,8-bis[(2R,3R)-3,5,7-trihydroxy-3,4-dihydro-2H-chromen-2-yl]benzo[7]annulen-6-one, Silymarin, Withanolide D, Spirosolane and Oridonin as potential HE inhibitors with better binding energy. Furthermore, molecular dynamics simulations for 100 ns time scale revealed that most of the key HE contacts were retained throughout the simulations trajectories. Binding free energy calculations using MM/PBSA approach ranked the top-five potential NPACT compounds which can act as effective HE inhibitors. Graphic abstract ![]()
Electronic supplementary material The online version of this article (10.1007/s11030-020-10135-w) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chirag N Patel
- Department of Botany, Bioinformatics, and Climate Change Impacts Management, University School of Sciences, Gujarat University, Ahmedabad, 380009, India
| | - Sivakumar Prasanth Kumar
- Department of Life Sciences, University School of Sciences, Gujarat University, Ahmedabad, 380009, India
| | - Himanshu A Pandya
- Department of Botany, Bioinformatics, and Climate Change Impacts Management, University School of Sciences, Gujarat University, Ahmedabad, 380009, India
| | - Rakesh M Rawal
- Department of Life Sciences, University School of Sciences, Gujarat University, Ahmedabad, 380009, India.
| |
Collapse
|
126
|
Mandala VS, McKay MJ, Shcherbakov AA, Dregni AJ, Kolocouris A, Hong M. Structure and Drug Binding of the SARS-CoV-2 Envelope Protein in Phospholipid Bilayers. RESEARCH SQUARE 2020:rs.3.rs-77124. [PMID: 32995764 PMCID: PMC7523133 DOI: 10.21203/rs.3.rs-77124/v1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the ongoing COVID-19 pandemic. Successful development of vaccines and antivirals against SARS-CoV-2 requires a comprehensive understanding of the essential proteins of the virus. The envelope (E) protein of SARS-CoV-2 assembles into a cation-selective channel that mediates virus budding, release, and host inflammation response. E blockage reduces virus pathogenicity while E deletion attenuates the virus. Here we report the 2.4 Å structure and drug-binding site of E's transmembrane (TM) domain, determined using solid-state nuclear magnetic resonance (NMR) spectroscopy. In lipid bilayers that mimic the endoplasmic reticulum Golgi intermediate compartment (ERGIC) membrane, ETM forms a five-helix bundle surrounding a narrow central pore. The middle of the TM segment is distorted from the ideal a-helical geometry due to three regularly spaced phenylalanine residues, which stack within each helix and between neighboring helices. These aromatic interactions, together with interhelical Val and Leu interdigitation, cause a dehydrated pore compared to the viroporins of influenza and HIV viruses. Hexamethylene amiloride and amantadine bind shallowly to polar residues at the N-terminal lumen, while acidic pH affects the C-terminal conformation. These results indicate that SARS-CoV-2 E forms a structurally robust but bipartite channel whose N- and C-terminal halves can interact with drugs, ions and other viral and host proteins semi-independently. This structure establishes the atomic basis for designing E inhibitors as antiviral drugs against SARS-CoV-2.
Collapse
Affiliation(s)
- Venkata S. Mandala
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Matthew J. McKay
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Alexander A. Shcherbakov
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Aurelio J. Dregni
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| | - Antonios Kolocouris
- Department of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis Zografou, Athens 15771, Greece
| | - Mei Hong
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
127
|
Hartenian E, Nandakumar D, Lari A, Ly M, Tucker JM, Glaunsinger BA. The molecular virology of coronaviruses. J Biol Chem 2020; 295:12910-12934. [PMID: 32661197 PMCID: PMC7489918 DOI: 10.1074/jbc.rev120.013930] [Citation(s) in RCA: 333] [Impact Index Per Article: 66.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/13/2020] [Indexed: 12/14/2022] Open
Abstract
Few human pathogens have been the focus of as much concentrated worldwide attention as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of COVID-19. Its emergence into the human population and ensuing pandemic came on the heels of severe acute respiratory syndrome coronavirus (SARS-CoV) and Middle East respiratory syndrome coronavirus (MERS-CoV), two other highly pathogenic coronavirus spillovers, which collectively have reshaped our view of a virus family previously associated primarily with the common cold. It has placed intense pressure on the collective scientific community to develop therapeutics and vaccines, whose engineering relies on a detailed understanding of coronavirus biology. Here, we present the molecular virology of coronavirus infection, including its entry into cells, its remarkably sophisticated gene expression and replication mechanisms, its extensive remodeling of the intracellular environment, and its multifaceted immune evasion strategies. We highlight aspects of the viral life cycle that may be amenable to antiviral targeting as well as key features of its biology that await discovery.
Collapse
Affiliation(s)
- Ella Hartenian
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - Divya Nandakumar
- Department of Plant and Microbial Biology, University of California, Berkeley, California, USA
| | - Azra Lari
- Department of Plant and Microbial Biology, University of California, Berkeley, California, USA
| | - Michael Ly
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA
| | - Jessica M Tucker
- Department of Plant and Microbial Biology, University of California, Berkeley, California, USA
| | - Britt A Glaunsinger
- Department of Molecular and Cell Biology, University of California, Berkeley, California, USA; Department of Plant and Microbial Biology, University of California, Berkeley, California, USA; Howard Hughes Medical Institute, University of California, Berkeley, California, USA.
| |
Collapse
|
128
|
Hadi J, Dunowska M, Wu S, Brightwell G. Control Measures for SARS-CoV-2: A Review on Light-Based Inactivation of Single-Stranded RNA Viruses. Pathogens 2020; 9:E737. [PMID: 32911671 PMCID: PMC7558314 DOI: 10.3390/pathogens9090737] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 09/04/2020] [Accepted: 09/05/2020] [Indexed: 12/20/2022] Open
Abstract
SARS-CoV-2 is a single-stranded RNA virus classified in the family Coronaviridae. In this review, we summarize the literature on light-based (UV, blue, and red lights) sanitization methods for the inactivation of ssRNA viruses in different matrixes (air, liquid, and solid). The rate of inactivation of ssRNA viruses in liquid was higher than in air, whereas inactivation on solid surfaces varied with the type of surface. The efficacy of light-based inactivation was reduced by the presence of absorptive materials. Several technologies can be used to deliver light, including mercury lamp (conventional UV), excimer lamp (UV), pulsed-light, and light-emitting diode (LED). Pulsed-light technologies could inactivate viruses more quickly than conventional UV-C lamps. Large-scale use of germicidal LED is dependent on future improvements in their energy efficiency. Blue light possesses virucidal potential in the presence of exogenous photosensitizers, although femtosecond laser (ultrashort pulses) can be used to circumvent the need for photosensitizers. Red light can be combined with methylene blue for application in medical settings, especially for sanitization of blood products. Future modelling studies are required to establish clearer parameters for assessing susceptibility of viruses to light-based inactivation. There is considerable scope for improvement in the current germicidal light-based technologies and practices.
Collapse
Affiliation(s)
- Joshua Hadi
- AgResearch Ltd., Hopkirk Research Institute, Cnr University Ave and Library Road, Massey University, Palmerston North 4442, New Zealand; (J.H.); (S.W.)
| | - Magdalena Dunowska
- School of Veterinary Science, Massey University Manawatu (Turitea) Tennent Drive, Palmerston North 4474, New Zealand;
| | - Shuyan Wu
- AgResearch Ltd., Hopkirk Research Institute, Cnr University Ave and Library Road, Massey University, Palmerston North 4442, New Zealand; (J.H.); (S.W.)
| | - Gale Brightwell
- AgResearch Ltd., Hopkirk Research Institute, Cnr University Ave and Library Road, Massey University, Palmerston North 4442, New Zealand; (J.H.); (S.W.)
- New Zealand Food Safety Science and Research Centre, Massey University Manawatu (Turitea) Tennent Drive, Palmerston North 4474, New Zealand
| |
Collapse
|
129
|
De Maio F, Lo Cascio E, Babini G, Sali M, Della Longa S, Tilocca B, Roncada P, Arcovito A, Sanguinetti M, Scambia G, Urbani A. Improved binding of SARS-CoV-2 Envelope protein to tight junction-associated PALS1 could play a key role in COVID-19 pathogenesis. Microbes Infect 2020; 22:592-597. [PMID: 32891874 PMCID: PMC7473260 DOI: 10.1016/j.micinf.2020.08.006] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 12/14/2022]
Abstract
The Envelope (E) protein of SARS-CoV-2 is the most enigmatic protein among the four structural ones. Most of its current knowledge is based on the direct comparison to the SARS E protein, initially mistakenly undervalued and subsequently proved to be a key factor in the ER-Golgi localization and in tight junction disruption. We compared the genomic sequences of E protein of SARS-CoV-2, SARS-CoV and the closely related genomes of bats and pangolins obtained from the GISAID and GenBank databases. When compared to the known SARS E protein, we observed a significant difference in amino acid sequence in the C-terminal end of SARS-CoV-2 E protein. Subsequently, in silico modelling analyses of E proteins conformation and docking provide evidences of a strengthened binding of SARS-CoV-2 E protein with the tight junction-associated PALS1 protein. Based on our computational evidences and on data related to SARS-CoV, we believe that SARS-CoV-2 E protein interferes more stably with PALS1 leading to an enhanced epithelial barrier disruption, amplifying the inflammatory processes, and promoting tissue remodelling. These findings raise a warning on the underestimated role of the E protein in the pathogenic mechanism and open the route to detailed experimental investigations.
Collapse
Affiliation(s)
- Flavio De Maio
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy; Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Ettore Lo Cascio
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Gabriele Babini
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy.
| | - Michela Sali
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy; Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Stefano Della Longa
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Bruno Tilocca
- Department of Health Science, University "Magna Græcia" of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Paola Roncada
- Department of Health Science, University "Magna Græcia" of Catanzaro, Viale Europa, 88100 Catanzaro, Italy
| | - Alessandro Arcovito
- Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy; Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy
| | - Maurizio Sanguinetti
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy; Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Microbiologia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Giovanni Scambia
- Dipartimento di Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy; Dipartimento di Scienze della Vita e Sanità Pubblica - Sezione di Ginecologia ed Ostetricia, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| | - Andrea Urbani
- Dipartimento di Scienze di Laboratorio e Infettivologiche, Fondazione Policlinico Universitario "A. Gemelli", IRCCS, Largo A. Gemelli 8, 00168 Roma, Italy; Dipartimento di Scienze Biotecnologiche di Base, Cliniche Intensivologiche e Perioperatorie - Sezione di Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168 Roma, Italy
| |
Collapse
|
130
|
Duart G, García-Murria MJ, Grau B, Acosta-Cáceres JM, Martínez-Gil L, Mingarro I. SARS-CoV-2 envelope protein topology in eukaryotic membranes. Open Biol 2020; 10:200209. [PMID: 32898469 PMCID: PMC7536074 DOI: 10.1098/rsob.200209] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 08/18/2020] [Indexed: 02/06/2023] Open
Abstract
Coronavirus E protein is a small membrane protein found in the virus envelope. Different coronavirus E proteins share striking biochemical and functional similarities, but sequence conservation is limited. In this report, we studied the E protein topology from the new SARS-CoV-2 virus both in microsomal membranes and in mammalian cells. Experimental data reveal that E protein is a single-spanning membrane protein with the N-terminus being translocated across the membrane, while the C-terminus is exposed to the cytoplasmic side (Ntlum/Ctcyt). The defined membrane protein topology of SARS-CoV-2 E protein may provide a useful framework to understand its interaction with other viral and host components and contribute to establish the basis to tackle the pathogenesis of SARS-CoV-2.
Collapse
Affiliation(s)
| | | | | | | | - Luis Martínez-Gil
- Departament de Bioquímica i Biologia Molecular, Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BioTecMed), Universitat de ValènciaE-46100 Burjassot, Spain
| | - Ismael Mingarro
- Departament de Bioquímica i Biologia Molecular, Estructura de Recerca Interdisciplinar en Biotecnologia i Biomedicina (ERI BioTecMed), Universitat de ValènciaE-46100 Burjassot, Spain
| |
Collapse
|
131
|
Hu WT, Howell JC, Ozturk T, Benameur K, Bassit LC, Ramonell R, Cashman KS, Pirmohammed S, Roback JD, Marconi VC, Yang I, Mac VV, Smith D, Sanz I, Wharton W, Lee FEH, Schinazi RF. Antibody Profiles According to Mild or Severe SARS-CoV-2 Infection, Atlanta, Georgia, USA, 2020. Emerg Infect Dis 2020; 26:2974-2978. [PMID: 32857691 PMCID: PMC7706962 DOI: 10.3201/eid2612.203334] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Among patients with coronavirus disease (COVID-19), IgM levels increased early after symptom onset for those with mild and severe disease, but IgG levels increased early only in those with severe disease. A similar pattern was observed in a separate serosurveillance cohort. Mild COVID-19 should be investigated separately from severe COVID-19.
Collapse
|
132
|
Abstract
Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is a newly emerging, highly transmissible, and pathogenic coronavirus in humans that has caused global public health emergencies and economic crises. To date, millions of infections and thousands of deaths have been reported worldwide, and the numbers continue to rise. Currently, there is no specific drug or vaccine against this deadly virus; therefore, there is a pressing need to understand the mechanism(s) through which this virus enters the host cell. Viral entry into the host cell is a multistep process in which SARS-CoV-2 utilizes the receptor-binding domain (RBD) of the spike (S) glycoprotein to recognize angiotensin-converting enzyme 2 (ACE2) receptors on the human cells; this initiates host-cell entry by promoting viral-host cell membrane fusion through large-scale conformational changes in the S protein. Receptor recognition and fusion are critical and essential steps of viral infections and are key determinants of the viral host range and cross-species transmission. In this review, we summarize the current knowledge on the origin and evolution of SARS-CoV-2 and the roles of key viral factors. We discuss the structure of RNA-dependent RNA polymerase (RdRp) of SARS-CoV-2 and its significance in drug discovery and explain the receptor recognition mechanisms of coronaviruses. Further, we provide a comparative analysis of the SARS-CoV and SARS-CoV-2 S proteins and their receptor-binding specificity and discuss the differences in their antigenicity based on biophysical and structural characteristics.
Collapse
|
133
|
Sarkar M, Saha S. Structural insight into the role of novel SARS-CoV-2 E protein: A potential target for vaccine development and other therapeutic strategies. PLoS One 2020; 15:e0237300. [PMID: 32785274 PMCID: PMC7423102 DOI: 10.1371/journal.pone.0237300] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/25/2020] [Indexed: 01/15/2023] Open
Abstract
The outbreak of COVID-19 across the world has posed unprecedented and global challenges on multiple fronts. Most of the vaccine and drug development has focused on the spike proteins and viral RNA-polymerases and main protease for viral replication. Using the bioinformatics and structural modelling approach, we modelled the structure of the envelope (E)-protein of novel SARS-CoV-2. The E-protein of this virus shares sequence similarity with that of SARS- CoV-1, and is highly conserved in the N-terminus regions. Incidentally, compared to spike proteins, E proteins demonstrate lower disparity and mutability among the isolated sequences. Using homology modelling, we found that the most favorable structure could function as a gated ion channel conducting H+ ions. Combining pocket estimation and docking with water, we determined that GLU 8 and ASN 15 in the N-terminal region were in close proximity to form H-bonds which was further validated by insertion of the E protein in an ERGIC-mimic membrane. Additionally, two distinct "core" structures were visible, the hydrophobic core and the central core, which may regulate the opening/closing of the channel. We propose this as a mechanism of viral ion channeling activity which plays a critical role in viral infection and pathogenesis. In addition, it provides a structural basis and additional avenues for vaccine development and generating therapeutic interventions against the virus.
Collapse
Affiliation(s)
- Manish Sarkar
- Department of Biochemistry, Bose Institute, Kolkata, India
| | - Soham Saha
- Laboratory for Perception and Memory, Institut Pasteur, Paris, France
- Centre National de la Recherche Scientifique (CNRS), Unité Mixte de Recherche (UMR-3571), Paris, France
| |
Collapse
|
134
|
Mukherjee S, Bhattacharyya D, Bhunia A. Host-membrane interacting interface of the SARS coronavirus envelope protein: Immense functional potential of C-terminal domain. Biophys Chem 2020; 266:106452. [PMID: 32818817 PMCID: PMC7418743 DOI: 10.1016/j.bpc.2020.106452] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/01/2020] [Accepted: 08/01/2020] [Indexed: 12/12/2022]
Abstract
The Envelope (E) protein in SARS Coronavirus (CoV) is a small structural protein, incorporated as part of the envelope. A major fraction of the protein has been known to be associated with the host membranes, particularly organelles related to intracellular trafficking, prompting CoV packaging and propagation. Studies have elucidated the central hydrophobic transmembrane domain of the E protein being responsible for much of the viroporin activity in favor of the virus. However, newer insights into the organizational principles at the membranous compartments within the host cells suggest further complexity of the system. The lesser hydrophobic Carboxylic-terminal of the protein harbors interesting amino acid sequences- suggesting at the prevalence of membrane-directed amyloidogenic properties that remains mostly elusive. These highly conserved segments indicate at several potential membrane-associated functional roles that can redefine our comprehensive understanding of the protein. This should prompt further studies in designing and characterizing of effective targeted therapeutic measures. The SARS CoV Envelope protein is a small structural protein of the virus, responsible for viroporin like activity. Membrane- E protein interaction provides an useful insight into gaining mechanistic insight into its viroporin functions. The central hydrophobic transmembrane domain of E protein, known to affect ion-channel formation. The C-terminal region of the protein show further potential host-membrane directed functional roles. The highly conserved amyloidogenic amino acid stretches of the C-terminal suggest for its contribution to CoV propagation.
Collapse
Affiliation(s)
- Shruti Mukherjee
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII(M), Kolkata 700054, India
| | - Dipita Bhattacharyya
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII(M), Kolkata 700054, India
| | - Anirban Bhunia
- Department of Biophysics, Bose Institute, P-1/12 CIT Scheme VII(M), Kolkata 700054, India.
| |
Collapse
|
135
|
Artika IM, Dewantari AK, Wiyatno A. Molecular biology of coronaviruses: current knowledge. Heliyon 2020; 6:e04743. [PMID: 32835122 PMCID: PMC7430346 DOI: 10.1016/j.heliyon.2020.e04743] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 08/13/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023] Open
Abstract
The emergence of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) late December 2019 in Wuhan, China, marked the third introduction of a highly pathogenic coronavirus into the human population in the twenty-first century. The constant spillover of coronaviruses from natural hosts to humans has been linked to human activities and other factors. The seriousness of this infection and the lack of effective, licensed countermeasures clearly underscore the need of more detailed and comprehensive understanding of coronavirus molecular biology. Coronaviruses are large, enveloped viruses with a positive sense single-stranded RNA genome. Currently, coronaviruses are recognized as one of the most rapidly evolving viruses due to their high genomic nucleotide substitution rates and recombination. At the molecular level, the coronaviruses employ complex strategies to successfully accomplish genome expression, virus particle assembly and virion progeny release. As the health threats from coronaviruses are constant and long-term, understanding the molecular biology of coronaviruses and controlling their spread has significant implications for global health and economic stability. This review is intended to provide an overview of our current basic knowledge of the molecular biology of coronaviruses, which is important as basic knowledge for the development of coronavirus countermeasures.
Collapse
Affiliation(s)
- I. Made Artika
- Biosafety Level 3 Unit, Eijkman Institute for Molecular Biology, Jalan Diponegoro 69, Jakarta, 10430, Indonesia
- Department of Biochemistry, Faculty of Mathematics and Natural Sciences, Bogor Agricultural University, Darmaga Campus, Bogor, 16680, Indonesia
| | - Aghnianditya Kresno Dewantari
- Emerging Virus Research Unit, Eijkman Institute for Molecular Biology, Jalan Diponegoro 69, Jakarta, 10430, Indonesia
| | - Ageng Wiyatno
- Emerging Virus Research Unit, Eijkman Institute for Molecular Biology, Jalan Diponegoro 69, Jakarta, 10430, Indonesia
| |
Collapse
|
136
|
Abstract
The current global pandemic COVID-19 caused by the SARS-CoV-2 virus has already inflicted insurmountable damage both to the human lives and global economy. There is an immediate need for identification of effective drugs to contain the disastrous virus outbreak. Global efforts are already underway at a war footing to identify the best drug combination to address the disease. In this review, an attempt has been made to understand the SARS-CoV-2 life cycle, and based on this information potential druggable targets against SARS-CoV-2 are summarized. Also, the strategies for ongoing and future drug discovery against the SARS-CoV-2 virus are outlined. Given the urgency to find a definitive cure, ongoing drug repurposing efforts being carried out by various organizations are also described. The unprecedented crisis requires extraordinary efforts from the scientific community to effectively address the issue and prevent further loss of human lives and health.
Collapse
Affiliation(s)
- Ambrish Saxena
- Indian Institute of Technology Tirupati, Tirupati, India
| |
Collapse
|
137
|
Bhowmik D, Nandi R, Jagadeesan R, Kumar N, Prakash A, Kumar D. Identification of potential inhibitors against SARS-CoV-2 by targeting proteins responsible for envelope formation and virion assembly using docking based virtual screening, and pharmacokinetics approaches. INFECTION GENETICS AND EVOLUTION 2020; 84:104451. [PMID: 32640381 PMCID: PMC7335633 DOI: 10.1016/j.meegid.2020.104451] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/25/2020] [Accepted: 06/29/2020] [Indexed: 02/06/2023]
Abstract
WHO has declared the outbreak of COVID-19 as a public health emergency of international concern. The ever-growing new cases have called for an urgent emergency for specific anti-COVID-19 drugs. Three structural proteins (Membrane, Envelope and Nucleocapsid protein) play an essential role in the assembly and formation of the infectious virion particles. Thus, the present study was designed to identify potential drug candidates from the unique collection of 548 anti-viral compounds (natural and synthetic anti-viral), which target SARS-CoV-2 structural proteins. High-end molecular docking analysis was performed to characterize the binding affinity of the selected drugs-the ligand, with the SARS-CoV-2 structural proteins, while high-level Simulation studies analyzed the stability of drug-protein interactions. The present study identified rutin, a bioflavonoid and the antibiotic, doxycycline, as the most potent inhibitor of SARS-CoV-2 envelope protein. Caffeic acid and ferulic acid were found to inhibit SARS-CoV-2 membrane protein while the anti-viral agent's simeprevir and grazoprevir showed a high binding affinity for nucleocapsid protein. All these compounds not only showed excellent pharmacokinetic properties, absorption, metabolism, minimal toxicity and bioavailability but were also remain stabilized at the active site of proteins during the MD simulation. Thus, the identified lead compounds may act as potential molecules for the development of effective drugs against SARS-CoV-2 by inhibiting the envelope formation, virion assembly and viral pathogenesis.
Collapse
Affiliation(s)
- Deep Bhowmik
- Department of Microbiology, Assam University, Silchar 788011, Assam, India
| | - Rajat Nandi
- Department of Microbiology, Assam University, Silchar 788011, Assam, India
| | - Rahul Jagadeesan
- CAS in Crystallography and Biophysics, Guindy Campus, University of Madras, Chennai 600025, India
| | - Niranjan Kumar
- School of Computational and Integrative Sciences, Jawaharlal Nehru University, New Delhi 110067, India
| | - Amresh Prakash
- Amity Institute of Integrative Sciences and Health, Amity University Haryana, Gurgaon 122413, India
| | - Diwakar Kumar
- Department of Microbiology, Assam University, Silchar 788011, Assam, India.
| |
Collapse
|
138
|
Luo Z, Ang MJY, Chan SY, Yi Z, Goh YY, Yan S, Tao J, Liu K, Li X, Zhang H, Huang W, Liu X. Combating the Coronavirus Pandemic: Early Detection, Medical Treatment, and a Concerted Effort by the Global Community. RESEARCH (WASHINGTON, D.C.) 2020; 2020:6925296. [PMID: 32607499 PMCID: PMC7315394 DOI: 10.34133/2020/6925296] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 04/20/2020] [Indexed: 01/08/2023]
Abstract
The World Health Organization (WHO) has declared the outbreak of 2019 novel coronavirus, known as 2019-nCoV, a pandemic, as the coronavirus has now infected over 2.6 million people globally and caused more than 185,000 fatalities as of April 23, 2020. Coronavirus disease 2019 (COVID-19) causes a respiratory illness with symptoms such as dry cough, fever, sudden loss of smell, and, in more severe cases, difficulty breathing. To date, there is no specific vaccine or treatment proven effective against this viral disease. Early and accurate diagnosis of COVID-19 is thus critical to curbing its spread and improving health outcomes. Reverse transcription-polymerase chain reaction (RT-PCR) is commonly used to detect the presence of COVID-19. Other techniques, such as recombinase polymerase amplification (RPA), loop-mediated isothermal amplification (LAMP), clustered regularly interspaced short palindromic repeats (CRISPR), and microfluidics, have allowed better disease diagnosis. Here, as part of the effort to expand screening capacity, we review advances and challenges in the rapid detection of COVID-19 by targeting nucleic acids, antigens, or antibodies. We also summarize potential treatments and vaccines against COVID-19 and discuss ongoing clinical trials of interventions to reduce viral progression.
Collapse
Affiliation(s)
- Zichao Luo
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Melgious Jin Yan Ang
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, Singapore 117456, Singapore
| | - Siew Yin Chan
- Frontiers Science Center for Flexible Electronics & Shaanxi Institute of Flexible Electronics, Northwestern Polytechnical University, Xi'an 710072, China
| | - Zhigao Yi
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Yi Yiing Goh
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- NUS Graduate School for Integrative Sciences and Engineering, Singapore 117456, Singapore
| | - Shuangqian Yan
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
| | - Jun Tao
- Sports Medical Centre, The Second Affiliated Hospital of Nanchang University, Nanchang 330000, China
| | - Kai Liu
- State Key Laboratory of Rare Earth Resource Utilization, Chang Chun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Xiaosong Li
- Department of Oncology, The Fourth Medical Center of Chinese People's Liberation Army General Hospital, Beijing 100048, China
| | - Hongjie Zhang
- State Key Laboratory of Rare Earth Resource Utilization, Chang Chun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
- Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics & Shaanxi Institute of Flexible Electronics, Northwestern Polytechnical University, Xi'an 710072, China
- Key Laboratory of Flexible Electronics & Institute of Advanced Materials, Nanjing Tech University, Nanjing 211816, China
| | - Xiaogang Liu
- Department of Chemistry, National University of Singapore, Singapore 117543, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, Fuzhou 350807, China
| |
Collapse
|
139
|
Study of Morphological Nature of Coronavirus: Causes and Prevention. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2020. [DOI: 10.22207/jpam.14.spl1.34] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The emergence of an unusual Corona virus (COVID-19) flu pandemic starting in China in December 2019, spreading all around the globe is a major threat to public health. The investigations have shown this virus originated from a seafood market in Wuhan. However, the unavailability of medicines for the new disease is a big challenge all around. An attempt has been made in the present article to familiarize about the morphology of the virus. The study of effect of pH, temperature and relative humidity is also depicted. Various preventive measures have also been discussed. The natural dietary measures suggested in the paper would be very beneficial to improve and boost the immunity of the mankind.
Collapse
|
140
|
HE Y, ZHENG C. [Replication and transmission mechanisms of highly pathogenic human coronavirus]. Zhejiang Da Xue Xue Bao Yi Xue Ban 2020; 49:324-339. [PMID: 32762165 PMCID: PMC8800760 DOI: 10.3785/j.issn.1008-9292.2020.03.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 04/03/2020] [Indexed: 11/15/2022]
Abstract
The three known highly pathogenic human coronaviruses are severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome coronavirus (MERS-CoV), and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Human highly pathogenic coronaviruses are composed of non-structural proteins, structural proteins, accessory proteins and ribonucleic acid. Viral particles recognize host receptors via spike glycoprotein (S protein), enter host cells by membrane fusion, replicate in host cells through large replication-transcription complexes, and promote proliferation by interfering with and suppressing the host's immune response. Highly pathogenic human coronaviruses are hosted by humans and vertebrates. Viral particles are transmitted through droplets, contact and aerosols or likely through digestive tract, urine, eyes and other routes. This review discusses the mechanisms of replication and transmission of highly pathogenic human coronaviruses providing basis for future studies on interrupting the transmission and pathogenicity of these pathogenic viruses.
Collapse
Affiliation(s)
| | - Chanying ZHENG
- 郑婵颖(1978-), 女, 博士, 副研究员, 硕士生导师, 主要从事脑功能和蛋白质分子机制研究; E-mail:
;
https://orcid.org/0000-0001-8389-2101
| |
Collapse
|
141
|
Design of a Multiepitope-Based Peptide Vaccine against the E Protein of Human COVID-19: An Immunoinformatics Approach. BIOMED RESEARCH INTERNATIONAL 2020; 2020:2683286. [PMID: 32461973 PMCID: PMC7212276 DOI: 10.1155/2020/2683286] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 04/20/2020] [Indexed: 12/20/2022]
Abstract
Background A new endemic disease has spread across Wuhan City, China, in December 2019. Within few weeks, the World Health Organization (WHO) announced a novel coronavirus designated as coronavirus disease 2019 (COVID-19). In late January 2020, WHO declared the outbreak of a “public-health emergency of international concern” due to the rapid and increasing spread of the disease worldwide. Currently, there is no vaccine or approved treatment for this emerging infection; thus, the objective of this study is to design a multiepitope peptide vaccine against COVID-19 using an immunoinformatics approach. Method Several techniques facilitating the combination of the immunoinformatics approach and comparative genomic approach were used in order to determine the potential peptides for designing the T-cell epitope-based peptide vaccine using the envelope protein of 2019-nCoV as a target. Results Extensive mutations, insertion, and deletion were discovered with comparative sequencing in the COVID-19 strain. Additionally, ten peptides binding to MHC class I and MHC class II were found to be promising candidates for vaccine design with adequate world population coverage of 88.5% and 99.99%, respectively. Conclusion The T-cell epitope-based peptide vaccine was designed for COVID-19 using the envelope protein as an immunogenic target. Nevertheless, the proposed vaccine rapidly needs to be validated clinically in order to ensure its safety and immunogenic profile to help stop this epidemic before it leads to devastating global outbreaks.
Collapse
|
142
|
Tilocca B, Soggiu A, Sanguinetti M, Babini G, De Maio F, Britti D, Zecconi A, Bonizzi L, Urbani A, Roncada P. Immunoinformatic analysis of the SARS-CoV-2 envelope protein as a strategy to assess cross-protection against COVID-19. Microbes Infect 2020; 22:182-187. [PMID: 32446902 PMCID: PMC7241347 DOI: 10.1016/j.micinf.2020.05.013] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 05/19/2020] [Indexed: 12/11/2022]
Abstract
Envelope protein of coronaviruses is a structural protein existing in both monomeric and homo-pentameric form. It has been related to a multitude of roles including virus infection, replication, dissemination and immune response stimulation. In the present study, we employed an immunoinformatic approach to investigate the major immunogenic domains of the SARS-CoV-2 envelope protein and map them among the homologue proteins of coronaviruses with tropism for animal species that are closely inter-related with the human beings population all over the world. Also, when not available, we predicted the envelope protein structural folding and mapped SARS-CoV-2 epitopes. Envelope sequences alignment provides evidence of high sequence homology for some of the investigated virus specimens; while the structural mapping of epitopes resulted in the interesting maintenance of the structural folding and epitope sequence localization also in the envelope proteins scoring a lower alignment score. In line with the One-Health approach, our evidences provide a molecular structural rationale for a potential role of taxonomically related coronaviruses in conferring protection from SARS-CoV-2 infection and identifying potential candidates for the development of diagnostic tools and prophylactic-oriented strategies.
Collapse
Affiliation(s)
- Bruno Tilocca
- Department of Health Science, University "Magna Græcia" of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Alessio Soggiu
- Department of Biomedical, Surgical and Dental Sciences- One Health Unit, University of Milano, Via Celoria n10, 20133, Milano, Italy; Department of Veterinary Medicine, University of Milano, Via dell'Università 6, 26900, Lodi, Italy
| | - Maurizio Sanguinetti
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Roma, Italy; Dipartimento di Scienze di laboratorio e infettivologiche, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli 8, 00168, Roma, Italy
| | - Gabriele Babini
- Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli, Largo A. Gemelli 8, 00168, Roma, Italy
| | - Flavio De Maio
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Roma, Italy; Dipartimento di Scienze di laboratorio e infettivologiche, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli 8, 00168, Roma, Italy
| | - Domenico Britti
- Department of Health Science, University "Magna Græcia" of Catanzaro, Viale Europa, 88100, Catanzaro, Italy
| | - Alfonso Zecconi
- Department of Biomedical, Surgical and Dental Sciences- One Health Unit, University of Milano, Via Celoria n10, 20133, Milano, Italy
| | - Luigi Bonizzi
- Department of Biomedical, Surgical and Dental Sciences- One Health Unit, University of Milano, Via Celoria n10, 20133, Milano, Italy
| | - Andrea Urbani
- Department of Basic Biotechnological Sciences, Intensivological and Perioperative Clinics, Università Cattolica del Sacro Cuore, Largo Francesco Vito 1, 00168, Roma, Italy; Dipartimento di Scienze di laboratorio e infettivologiche, Fondazione Policlinico Universitario Agostino Gemelli, Largo A. Gemelli 8, 00168, Roma, Italy.
| | - Paola Roncada
- Department of Health Science, University "Magna Græcia" of Catanzaro, Viale Europa, 88100, Catanzaro, Italy.
| |
Collapse
|
143
|
Prajapat M, Sarma P, Shekhar N, Avti P, Sinha S, Kaur H, Kumar S, Bhattacharyya A, Kumar H, Bansal S, Medhi B. Drug targets for corona virus: A systematic review. Indian J Pharmacol 2020; 52:56-65. [PMID: 32201449 PMCID: PMC7074424 DOI: 10.4103/ijp.ijp_115_20] [Citation(s) in RCA: 278] [Impact Index Per Article: 55.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 02/23/2020] [Accepted: 02/25/2020] [Indexed: 12/31/2022] Open
Abstract
The 2019-novel coronavirus (nCoV) is a major source of disaster in the 21th century. However, the lack of specific drugs to prevent/treat an attack is a major need at this current point of time. In this regard, we conducted a systematic review to identify major druggable targets in coronavirus (CoV). We searched PubMed and RCSB database with keywords HCoV, NCoV, corona virus, SERS-CoV, MERS-CoV, 2019-nCoV, crystal structure, X-ray crystallography structure, NMR structure, target, and drug target till Feb 3, 2020. The search identified seven major targets (spike protein, envelop protein, membrane protein, protease, nucleocapsid protein, hemagglutinin esterase, and helicase) for which drug design can be considered. There are other 16 nonstructural proteins (NSPs), which can also be considered from the drug design perspective. The major structural proteins and NSPs may serve an important role from drug design perspectives. However, the occurrence of frequent recombination events is a major deterrent factor toward the development of CoV-specific vaccines/drugs.
Collapse
Affiliation(s)
- Manisha Prajapat
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Phulen Sarma
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Nishant Shekhar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pramod Avti
- Department of Biophysics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shweta Sinha
- Department of Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Hardeep Kaur
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Subodh Kumar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anusuya Bhattacharyya
- Departments of Ophthalmology, Government Medical College and Hospital, Chandigarh, India
| | - Harish Kumar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Seema Bansal
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
144
|
Prajapat M, Sarma P, Shekhar N, Avti P, Sinha S, Kaur H, Kumar S, Bhattacharyya A, Kumar H, Bansal S, Medhi B. Drug targets for corona virus: A systematic review. Indian J Pharmacol 2020. [PMID: 32201449 DOI: 10.4103/ijp.ijp.115-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2023] Open
Abstract
The 2019-novel coronavirus (nCoV) is a major source of disaster in the 21th century. However, the lack of specific drugs to prevent/treat an attack is a major need at this current point of time. In this regard, we conducted a systematic review to identify major druggable targets in coronavirus (CoV). We searched PubMed and RCSB database with keywords HCoV, NCoV, corona virus, SERS-CoV, MERS-CoV, 2019-nCoV, crystal structure, X-ray crystallography structure, NMR structure, target, and drug target till Feb 3, 2020. The search identified seven major targets (spike protein, envelop protein, membrane protein, protease, nucleocapsid protein, hemagglutinin esterase, and helicase) for which drug design can be considered. There are other 16 nonstructural proteins (NSPs), which can also be considered from the drug design perspective. The major structural proteins and NSPs may serve an important role from drug design perspectives. However, the occurrence of frequent recombination events is a major deterrent factor toward the development of CoV-specific vaccines/drugs.
Collapse
Affiliation(s)
- Manisha Prajapat
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Phulen Sarma
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Nishant Shekhar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pramod Avti
- Department of Biophysics, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Shweta Sinha
- Department of Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Hardeep Kaur
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Subodh Kumar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Anusuya Bhattacharyya
- Departments of Ophthalmology, Government Medical College and Hospital, Chandigarh, India
| | - Harish Kumar
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Seema Bansal
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Bikash Medhi
- Department of Pharmacology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
145
|
Singh V, Luthra A, Chauhan R, Meena SC. Basic Virology and Pathophysiology of COVID-19. CLINICAL SYNOPSIS OF COVID-19 2020:5-29. [DOI: 10.1007/978-981-15-8681-1_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
146
|
Abstract
BACKGROUND Coronaviruses (CoVs) primarily cause enzootic infections in birds and mammals but, in the last few decades, have shown to be capable of infecting humans as well. The outbreak of severe acute respiratory syndrome (SARS) in 2003 and, more recently, Middle-East respiratory syndrome (MERS) has demonstrated the lethality of CoVs when they cross the species barrier and infect humans. A renewed interest in coronaviral research has led to the discovery of several novel human CoVs and since then much progress has been made in understanding the CoV life cycle. The CoV envelope (E) protein is a small, integral membrane protein involved in several aspects of the virus' life cycle, such as assembly, budding, envelope formation, and pathogenesis. Recent studies have expanded on its structural motifs and topology, its functions as an ion-channelling viroporin, and its interactions with both other CoV proteins and host cell proteins. MAIN BODY This review aims to establish the current knowledge on CoV E by highlighting the recent progress that has been made and comparing it to previous knowledge. It also compares E to other viral proteins of a similar nature to speculate the relevance of these new findings. Good progress has been made but much still remains unknown and this review has identified some gaps in the current knowledge and made suggestions for consideration in future research. CONCLUSIONS The most progress has been made on SARS-CoV E, highlighting specific structural requirements for its functions in the CoV life cycle as well as mechanisms behind its pathogenesis. Data shows that E is involved in critical aspects of the viral life cycle and that CoVs lacking E make promising vaccine candidates. The high mortality rate of certain CoVs, along with their ease of transmission, underpins the need for more research into CoV molecular biology which can aid in the production of effective anti-coronaviral agents for both human CoVs and enzootic CoVs.
Collapse
Affiliation(s)
- Dewald Schoeman
- Molecular Biology and Virology Research Laboratory, Department of Medical Biosciences, University of the Western Cape, Cape Town, South Africa
| | - Burtram C Fielding
- Molecular Biology and Virology Research Laboratory, Department of Medical Biosciences, University of the Western Cape, Cape Town, South Africa.
| |
Collapse
|
147
|
Abstract
Viroporins are short polypeptides encoded by viruses. These small membrane proteins assemble into oligomers that can permeabilize cellular lipid bilayers, disrupting the physiology of the host to the advantage of the virus. Consequently, efforts during the last few decades have been focused towards the discovery of viroporin channel inhibitors, but in general these have not been successful to produce licensed drugs. Viroporins are also involved in viral pathogenesis by engaging in critical interactions with viral proteins, or disrupting normal host cellular pathways through coordinated interactions with host proteins. These protein-protein interactions (PPIs) may become alternative attractive drug targets for the development of antivirals. In this sense, while thus far most antiviral molecules have targeted viral proteins, focus is moving towards targeting host proteins that are essential for virus replication. In principle, this largely would overcome the problem of resistance, with the possibility of using repositioned existing drugs. The precise role of these PPIs, their strain- and host- specificities, and the structural determination of the complexes involved, are areas that will keep the fields of virology and structural biology occupied for years to come. In the present review, we provide an update of the efforts in the characterization of the main PPIs for most viroporins, as well as the role of viroporins in these PPIs interactions.
Collapse
Affiliation(s)
| | - David Bhella
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| |
Collapse
|
148
|
Castaño-Rodriguez C, Honrubia JM, Gutiérrez-Álvarez J, DeDiego ML, Nieto-Torres JL, Jimenez-Guardeño JM, Regla-Nava JA, Fernandez-Delgado R, Verdia-Báguena C, Queralt-Martín M, Kochan G, Perlman S, Aguilella VM, Sola I, Enjuanes L. Role of Severe Acute Respiratory Syndrome Coronavirus Viroporins E, 3a, and 8a in Replication and Pathogenesis. mBio 2018; 9:e02325-17. [PMID: 29789363 PMCID: PMC5964350 DOI: 10.1128/mbio.02325-17] [Citation(s) in RCA: 223] [Impact Index Per Article: 31.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 04/24/2018] [Indexed: 12/24/2022] Open
Abstract
Viroporins are viral proteins with ion channel (IC) activity that play an important role in several processes, including virus replication and pathogenesis. While many coronaviruses (CoVs) encode two viroporins, severe acute respiratory syndrome CoV (SARS-CoV) encodes three: proteins 3a, E, and 8a. Additionally, proteins 3a and E have a PDZ-binding motif (PBM), which can potentially bind over 400 cellular proteins which contain a PDZ domain, making them potentially important for the control of cell function. In the present work, a comparative study of the functional motifs included within the SARS-CoV viroporins was performed, mostly focusing on the roles of the IC and PBM of E and 3a proteins. Our results showed that the full-length E and 3a proteins were required for maximal SARS-CoV replication and virulence, whereas viroporin 8a had only a minor impact on these activities. A virus missing both the E and 3a proteins was not viable, whereas the presence of either protein with a functional PBM restored virus viability. E protein IC activity and the presence of its PBM were necessary for virulence in mice. In contrast, the presence or absence of the homologous motifs in protein 3a did not influence virus pathogenicity. Therefore, dominance of the IC and PBM of protein E over those of protein 3a was demonstrated in the induction of pathogenesis in mice.IMPORTANCE Collectively, these results demonstrate key roles for the ion channel and PBM domains in optimal virus replication and pathogenesis and suggest that the viral viroporins and PBMs are suitable targets for antiviral therapy and for mutation in attenuated SARS-CoV vaccines.
Collapse
Affiliation(s)
- Carlos Castaño-Rodriguez
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Jose M Honrubia
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Javier Gutiérrez-Álvarez
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta L DeDiego
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Jose L Nieto-Torres
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Jose M Jimenez-Guardeño
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Jose A Regla-Nava
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Raul Fernandez-Delgado
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Carmina Verdia-Báguena
- Department of Physics, Laboratory of Molecular Biophysics, Universitat Jaume I, Castelló, Spain
| | - Maria Queralt-Martín
- Eunice Kennedy Shriver NICHD, NIH, Bethesda, Maryland, USA
- Department of Physics, Laboratory of Molecular Biophysics, Universitat Jaume I, Castelló, Spain
| | - Grazyna Kochan
- Immunomodulation Group, Navarrabiomed-Biomedical Research Centre, IdISNA, Pamplona, Navarra, Spain
| | - Stanley Perlman
- Department of Microbiology, University of Iowa, Iowa City, Iowa, USA
| | - Vicente M Aguilella
- Department of Physics, Laboratory of Molecular Biophysics, Universitat Jaume I, Castelló, Spain
| | - Isabel Sola
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| | - Luis Enjuanes
- Department of Molecular and Cell Biology, Centro Nacional de Biotecnología (CNB-CSIC), Campus Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
149
|
Surya W, Li Y, Torres J. Structural model of the SARS coronavirus E channel in LMPG micelles. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1309-1317. [PMID: 29474890 PMCID: PMC7094280 DOI: 10.1016/j.bbamem.2018.02.017] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 02/14/2018] [Accepted: 02/16/2018] [Indexed: 01/27/2023]
Abstract
Coronaviruses (CoV) cause common colds in humans, but are also responsible for the recent Severe Acute, and Middle East, respiratory syndromes (SARS and MERS, respectively). A promising approach for prevention are live attenuated vaccines (LAVs), some of which target the envelope (E) protein, which is a small membrane protein that forms ion channels. Unfortunately, detailed structural information is still limited for SARS-CoV E, and non-existent for other CoV E proteins. Herein, we report a structural model of a SARS-CoV E construct in LMPG micelles with, for the first time, unequivocal intermolecular NOEs. The model corresponding to the detergent-embedded region is consistent with previously obtained orientational restraints obtained in lipid bilayers and in vivo escape mutants. The C-terminal domain is mostly α-helical, and extramembrane intermolecular NOEs suggest interactions that may affect the TM channel conformation.
Collapse
Affiliation(s)
- Wahyu Surya
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Yan Li
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Jaume Torres
- School of Biological Sciences, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
150
|
Stodola JK, Dubois G, Le Coupanec A, Desforges M, Talbot PJ. The OC43 human coronavirus envelope protein is critical for infectious virus production and propagation in neuronal cells and is a determinant of neurovirulence and CNS pathology. Virology 2018; 515:134-149. [PMID: 29287230 PMCID: PMC7118982 DOI: 10.1016/j.virol.2017.12.023] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Revised: 12/08/2017] [Accepted: 12/20/2017] [Indexed: 12/19/2022]
Abstract
The OC43 strain of human coronavirus (HCoV-OC43) is an ubiquitous respiratory tract pathogen possessing neurotropic capacities. Coronavirus structural envelope (E) protein possesses specific motifs involved in protein-protein interaction or in homo-oligomeric ion channel formation, which are known to play various roles including in virion morphology/assembly and in cell response to infection and/or virulence. Making use of recombinant viruses either devoid of the E protein or harboring mutations either in putative transmembrane domain or PDZ-binding motif, we demonstrated that a fully functional HCoV-OC43 E protein is first needed for optimal production of recombinant infectious viruses. Furthermore, HCoV-OC43 infection of human epithelial and neuronal cell lines, of mixed murine primary cultures from the central nervous system and of mouse central nervous system showed that the E protein is critical for efficient and optimal virus replication and propagation, and thereby for neurovirulence.
Collapse
Affiliation(s)
- Jenny K Stodola
- Laboratory of Neuroimmunovirology, INRS-Institut Armand- Frappier, Laval, Québec, Canada
| | - Guillaume Dubois
- Laboratory of Neuroimmunovirology, INRS-Institut Armand- Frappier, Laval, Québec, Canada
| | - Alain Le Coupanec
- Laboratory of Neuroimmunovirology, INRS-Institut Armand- Frappier, Laval, Québec, Canada
| | - Marc Desforges
- Laboratory of Neuroimmunovirology, INRS-Institut Armand- Frappier, Laval, Québec, Canada.
| | - Pierre J Talbot
- Laboratory of Neuroimmunovirology, INRS-Institut Armand- Frappier, Laval, Québec, Canada.
| |
Collapse
|