201
|
Pathangey G, Fadadu PP, Hospodar AR, Abbas AE. Angiotensin-converting enzyme 2 and COVID-19: patients, comorbidities, and therapies. Am J Physiol Lung Cell Mol Physiol 2021; 320:L301-L330. [PMID: 33237815 PMCID: PMC7938645 DOI: 10.1152/ajplung.00259.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 11/19/2020] [Accepted: 11/24/2020] [Indexed: 02/07/2023] Open
Abstract
On March 11, 2020, the World Health Organization declared coronavirus disease 2019 (COVID-19) a pandemic, and the reality of the situation has finally caught up to the widespread reach of the disease. The presentation of the disease is highly variable, ranging from asymptomatic carriers to critical COVID-19. The availability of angiotensin-converting enzyme 2 (ACE2) receptors may reportedly increase the susceptibility and/or disease progression of COVID-19. Comorbidities and risk factors have also been noted to increase COVID-19 susceptibility. In this paper, we hereby review the evidence pertaining to ACE2's relationship to common comorbidities, risk factors, and therapies associated with the susceptibility and severity of COVID-19. We also highlight gaps of knowledge that require further investigation. The primary comorbidities of respiratory disease, cardiovascular disease, renal disease, diabetes, obesity, and hypertension had strong evidence. The secondary risk factors of age, sex, and race/genetics had limited-to-moderate evidence. The tertiary factors of ACE inhibitors and angiotensin II receptor blockers had limited-to-moderate evidence. Ibuprofen and thiazolidinediones had limited evidence.
Collapse
Affiliation(s)
- Girish Pathangey
- William Beaumont School of Medicine, Oakland University, Rochester, Michigan
| | | | | | - Amr E Abbas
- William Beaumont School of Medicine, Oakland University, Rochester, Michigan
- Department of Cardiovascular Medicine, Beaumont Hospital Royal Oak, Royal Oak, Michigan
| |
Collapse
|
202
|
Sundah NR, Natalia A, Liu Y, Ho NRY, Zhao H, Chen Y, Miow QH, Wang Y, Beh DLL, Chew KL, Chan D, Tambyah PA, Ong CWM, Shao H. Catalytic amplification by transition-state molecular switches for direct and sensitive detection of SARS-CoV-2. SCIENCE ADVANCES 2021; 7:7/12/eabe5940. [PMID: 33731349 PMCID: PMC7968834 DOI: 10.1126/sciadv.abe5940] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 01/29/2021] [Indexed: 06/12/2023]
Abstract
Despite the importance of nucleic acid testing in managing the COVID-19 pandemic, current detection approaches remain limited due to their high complexity and extensive processing. Here, we describe a molecular nanotechnology that enables direct and sensitive detection of viral RNA targets in native clinical samples. The technology, termed catalytic amplification by transition-state molecular switch (CATCH), leverages DNA-enzyme hybrid complexes to form a molecular switch. By ratiometric tuning of its constituents, the multicomponent molecular switch is prepared in a hyperresponsive state-the transition state-that can be readily activated upon the binding of sparse RNA targets to turn on substantial enzymatic activity. CATCH thus achieves superior performance (~8 RNA copies/μl), direct fluorescence detection that bypasses all steps of PCR (<1 hour at room temperature), and versatile implementation (high-throughput 96-well format and portable microfluidic assay). When applied for clinical COVID-19 diagnostics, CATCH demonstrated direct and accurate detection in minimally processed patient swab samples.
Collapse
Affiliation(s)
- Noah R Sundah
- Institute for Health Innovation&Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Auginia Natalia
- Institute for Health Innovation&Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Yu Liu
- Institute for Health Innovation&Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Nicholas R Y Ho
- Institute for Health Innovation&Technology, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
| | - Haitao Zhao
- Institute for Health Innovation&Technology, National University of Singapore, Singapore, Singapore
| | - Yuan Chen
- Institute for Health Innovation&Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
| | - Qing Hao Miow
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yu Wang
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Darius L L Beh
- Division of Infectious Diseases, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Ka Lip Chew
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore
| | - Douglas Chan
- Department of Laboratory Medicine, Ng Teng Fong General Hospital, Singapore, Singapore
| | - Paul A Tambyah
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Infectious Diseases, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Catherine W M Ong
- Institute for Health Innovation&Technology, National University of Singapore, Singapore, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Infectious Diseases, Department of Medicine, National University Hospital, Singapore, Singapore
| | - Huilin Shao
- Institute for Health Innovation&Technology, National University of Singapore, Singapore, Singapore.
- Department of Biomedical Engineering, Faculty of Engineering, National University of Singapore, Singapore, Singapore
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
203
|
Na W, Moon H, Song D. A comprehensive review of SARS-CoV-2 genetic mutations and lessons from animal coronavirus recombination in one health perspective. J Microbiol 2021; 59:332-340. [PMID: 33624270 PMCID: PMC7901680 DOI: 10.1007/s12275-021-0660-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/08/2021] [Accepted: 02/15/2021] [Indexed: 12/18/2022]
Abstract
SARS-CoV-2 was originated from zoonotic coronaviruses and confirmed as a novel beta-coronavirus, which causes serious respiratory illness such as pneumonia and lung failure, COVID-19. In this review, we describe the genetic characteristics of SARS-CoV-2, including types of mutation, and molecular epidemiology, highlighting its key difference from animal coronaviruses. We further summarized the current knowledge on clinical, genetic, and pathological features of several animal coronaviruses and compared them with SARS-CoV-2, as well as recent evidences of interspecies transmission and recombination of animal coronaviruses to provide a better understanding of SARS-CoV-2 infection in One Health perspectives. We also discuss the potential wildlife hosts and zoonotic origin of this emerging virus in detail, that may help mitigate the spread and damages caused by the disease.
Collapse
Affiliation(s)
- Woonsung Na
- College of Veterinary Medicine, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyoungjoon Moon
- College of Healthcare & Biotechnology, Semyung University, Jecheon, 27136, Republic of Korea
| | - Daesub Song
- College of Pharmacy, Korea University, Sejong, 30019, Republic of Korea.
| |
Collapse
|
204
|
O'Horo JC, Cerhan JR, Cahn EJ, Bauer PR, Temesgen Z, Ebbert J, Abril A, Abu Saleh OM, Assi M, Berbari EF, Bierle DM, Bosch W, Burger CD, Cano Cevallos EJ, Clements CM, Carmona Porquera EM, Castillo Almeida NE, Challener DW, Chesdachai S, Comba IY, Corsini Campioli CG, Crane SJ, Dababneh AS, Enzler MJ, Fadel HJ, Ganesh R, De Moraes AG, Go JR, Gordon JE, Gurram PR, Guru PK, Halverson EL, Harrison MF, Heaton HA, Hurt R, Kasten MJ, Lee AS, Levy ER, Libertin CR, Mallea JM, Marshall WF, Matcha G, Meehan AM, Franco PM, Morice WG, O'Brien JJ, Oeckler R, Ommen S, Oravec CP, Orenstein R, Ough NJ, Palraj R, Patel BM, Pureza VS, Pickering B, Phelan DM, Razonable RR, Rizza S, Sampathkumar P, Sanghavi DK, Sen A, Siegel JL, Singbartl K, Shah AS, Shweta F, Speicher LL, Suh G, Tabaja H, Tande A, Ting HH, Tontz RC, Vaillant JJ, Vergidis P, Warsame MY, Yetmar ZA, Zomok CCD, Williams AW, Badley AD. Outcomes of COVID-19 With the Mayo Clinic Model of Care and Research. Mayo Clin Proc 2021; 96:601-618. [PMID: 33673913 PMCID: PMC7831394 DOI: 10.1016/j.mayocp.2020.12.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/11/2020] [Accepted: 12/15/2020] [Indexed: 01/02/2023]
Abstract
OBJECTIVE To report the Mayo Clinic experience with coronavirus disease 2019 (COVID-19) related to patient outcomes. METHODS We conducted a retrospective chart review of patients with COVID-19 diagnosed between March 1, 2020, and July 31, 2020, at any of the Mayo Clinic sites. We abstracted pertinent comorbid conditions such as age, sex, body mass index, Charlson Comorbidity Index variables, and treatments received. Factors associated with hospitalization and mortality were assessed in univariate and multivariate models. RESULTS A total of 7891 patients with confirmed COVID-19 infection with research authorization on file received care across the Mayo Clinic sites during the study period. Of these, 7217 patients were adults 18 years or older who were analyzed further. A total of 897 (11.4%) patients required hospitalization, and 354 (4.9%) received care in the intensive care unit (ICU). All hospitalized patients were reviewed by a COVID-19 Treatment Review Panel, and 77.5% (695 of 897) of inpatients received a COVID-19-directed therapy. Overall mortality was 1.2% (94 of 7891), with 7.1% (64 of 897) mortality in hospitalized patients and 11.3% (40 of 354) in patients requiring ICU care. CONCLUSION Mayo Clinic outcomes of patients with COVID-19 infection in the ICU, hospital, and community compare favorably with those reported nationally. This likely reflects the impact of interprofessional multidisciplinary team evaluation, effective leveraging of clinical trials and available treatments, deployment of remote monitoring tools, and maintenance of adequate operating capacity to not require surge adjustments. These best practices can help guide other health care systems with the continuing response to the COVID-19 pandemic.
Collapse
Key Words
- apache iv, acute physiology and chronic health evaluation iv
- ards, acute respiratory distress syndrome
- bmi, body mass index
- cci, charlson comorbidity index
- covid-19, coronavirus disease 2019
- eap, expanded access program
- ecmo, extracorporeal membrane oxygenation
- ehr, electronic health record
- icd-10, international classification of diseases, tenth revision
- icu, intensive care unit
- los, length of stay
- nih, national institutes of health
- or, odds ratio
- sars-cov-2, severe acute respiratory syndrome coronavirus 2
- sofa, sequential organ failure assessment
Collapse
Affiliation(s)
- John Charles O'Horo
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN; Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | - James R Cerhan
- Division of Health Science Research, Mayo Clinic, Rochester, MN
| | - Elliot J Cahn
- Division of Health Science Research, Mayo Clinic, Rochester, MN
| | - Philippe R Bauer
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | | | - Jon Ebbert
- Division of Community Internal Medicine, Mayo Clinic, Rochester, MN
| | - Andy Abril
- Division of Rheumatology, Mayo Clinic, Jacksonville, FL
| | | | - Mariam Assi
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Elie F Berbari
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Dennis M Bierle
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN
| | - Wendelyn Bosch
- Division of Infectious Diseases, Mayo Clinic, Jacksonville, FL
| | - Charles D Burger
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Jacksonville, FL
| | | | | | - Eva M Carmona Porquera
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN; Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN
| | | | | | | | - Isin Y Comba
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | | | - Sarah J Crane
- Division of Community Internal Medicine, Mayo Clinic, Rochester, MN
| | - Ala S Dababneh
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Mark J Enzler
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Hind J Fadel
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Ravindra Ganesh
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN
| | | | - John R Go
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Joel E Gordon
- Department of Family Medicine, Mayo Clinic Health System, Mankato, MN
| | - Pooja R Gurram
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Pramod K Guru
- Department of Critical Care Medicine, Mayo Clinic, Jacksonville, FL
| | | | | | | | - Ryan Hurt
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN
| | - Mary J Kasten
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Augustine S Lee
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Jacksonville, FL
| | - Emily R Levy
- Division of Pediatric Critical Care Medicine, Mayo Clinic, Rochester, MN; Division of Pediatric Infectious Diseases, Mayo Clinic, Rochester, MN
| | | | - Jorge M Mallea
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Jacksonville, FL
| | | | - Gautam Matcha
- Department of Internal Medicine, Mayo Clinic, Jacksonville, FL
| | - Anne M Meehan
- Division of Hospital Medicine, Mayo Clinic, Rochester, MN
| | | | - William G Morice
- Department of Laboratory Medicine Pathology, Mayo Clinic, Rochester, MN
| | - Jennifer J O'Brien
- Department of Laboratory Medicine Pathology, Mayo Clinic, Jacksonville, FL
| | - Richard Oeckler
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN; Division of Infectious Diseases, Mayo Clinic, Scottsdale, AZ
| | - Steve Ommen
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN
| | | | | | - Natalie J Ough
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Raj Palraj
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Bhavesh M Patel
- Department of Critical Care Medicine, Mayo Clinic, Scottsdale, AZ
| | - Vincent S Pureza
- Division of Hospital Internal Medicine, Mayo Clinic, Rochester, MN
| | - Brian Pickering
- Division of Intensive Care, Department of Anesthesia, Mayo Clinic, Rochester, MN
| | - David M Phelan
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | | | - Stacey Rizza
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | | | | | - Ayan Sen
- Department of Critical Care Medicine, Mayo Clinic, Scottsdale, AZ
| | | | - Kai Singbartl
- Department of Critical Care, Mayo Clinic, Rochester, MN
| | - Aditya S Shah
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Fnu Shweta
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Leigh L Speicher
- Division of General Internal Medicine, Mayo Clinic, Jacksonville, FL
| | - Gina Suh
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Hussam Tabaja
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Aaron Tande
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN
| | - Henry H Ting
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL
| | - Russell C Tontz
- Division of Occupational Medicine, Mayo Clinic Health System, Mankato, MN
| | | | | | | | | | | | - Amy W Williams
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN
| | - Andrew D Badley
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN; Department of Molecular Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
205
|
Izzedine H, Jhaveri KD, Perazella MA. Genes, COVID-19 and phenotype. Clin Kidney J 2021; 14:1485-1487. [PMID: 34221375 PMCID: PMC7928999 DOI: 10.1093/ckj/sfab044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 02/11/2021] [Indexed: 12/02/2022] Open
Affiliation(s)
- Hassan Izzedine
- Department of Nephrology, Peupliers Private Hospital, Ramsay Générale de Santé, Paris, France
| | - Kenar D Jhaveri
- Department of Medicine, Donald and Barbara School of Medicine at Hofstra/Northwell, Northwell Health, Great Neck, NY, USA
| | | |
Collapse
|
206
|
Jabarpour M, Dehghan M, Afsharipour G, Hajipour Abaee E, Mangolian Shahrbabaki P, Ahmadinejad M, Maazallahi M. The Impact of COVID-19 Outbreak on Nosocomial Infection Rate: A Case of Iran. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2021; 2021:6650920. [PMID: 33680220 PMCID: PMC7905999 DOI: 10.1155/2021/6650920] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/02/2021] [Accepted: 02/10/2021] [Indexed: 01/17/2023]
Abstract
BACKGROUND Coronavirus disease-19 (COVID-19) is a new type of coronavirus that has caused a global pandemic. The disease is highly contagious, and all people are susceptible to the disease. Therefore, extensive measures were taken to prevent the spread of the disease at the community and hospitals. This study aimed to investigate the impact of COVID-19 outbreak on nosocomial infection rate. METHODS This cross-sectional study was conducted in an educational hospital, southeast Iran. The nosocomial infection rates of critical/intensive care units (CCU/ICUs) and medical-surgical units were assessed during and before the COVID-19 outbreak. RESULTS There was a 19.75-point decrease in the total rate of nosocomial infection during the COVID-19 outbreak (P = 0.02). In addition, there was a 39.12-point decrease in the total rate of CCU/ICUs' nosocomial infection during the COVID-19 outbreak (P < 0.001). A 19.23-point decrease was also observed in the total rate of medical-surgical units' nosocomial infection during the COVID-19 outbreak (P = 0.13). All kinds of CCU/ICUs' nosocomial infections had between 31.22- and 100-point decreases during the COVID-19 outbreak. Among medical-surgical units, 33.33- and 30.70-point decreases were observed only in UTI and SSI, respectively, during the COVID-19 outbreak, while BSI had a 40-point increase during the COVID-19 outbreak. CONCLUSIONS Proper implementation of infection control protocols during the COVID-19 pandemic seems to reduce nosocomial infections.
Collapse
Affiliation(s)
- Maryam Jabarpour
- Clinical Research Unit, Shahid Bahonar Academic Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahlagha Dehghan
- Nursing Research Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Giti Afsharipour
- Clinical Research Unit, Shahid Bahonar Academic Center, Kerman University of Medical Sciences, Kerman, Iran
| | - Elham Hajipour Abaee
- Clinical Research Unit, Shahid Bahonar Academic Center, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Mehdi Ahmadinejad
- Fellow of Critical Care Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mahboobeh Maazallahi
- Department of Critical Care Nursing, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
207
|
Ali AM, Kunugi H. Propolis, Bee Honey, and Their Components Protect against Coronavirus Disease 2019 (COVID-19): A Review of In Silico, In Vitro, and Clinical Studies. Molecules 2021; 26:1232. [PMID: 33669054 PMCID: PMC7956496 DOI: 10.3390/molecules26051232] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/19/2021] [Accepted: 02/20/2021] [Indexed: 12/14/2022] Open
Abstract
Despite the virulence and high fatality of coronavirus disease 2019 (COVID-19), no specific antiviral treatment exists until the current moment. Natural agents with immune-promoting potentials such as bee products are being explored as possible treatments. Bee honey and propolis are rich in bioactive compounds that express strong antimicrobial, bactericidal, antiviral, anti-inflammatory, immunomodulatory, and antioxidant activities. This review examined the literature for the anti-COVID-19 effects of bee honey and propolis, with the aim of optimizing the use of these handy products as prophylactic or adjuvant treatments for people infected with severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2). Molecular simulations show that flavonoids in propolis and honey (e.g., rutin, naringin, caffeic acid phenyl ester, luteolin, and artepillin C) may inhibit viral spike fusion in host cells, viral-host interactions that trigger the cytokine storm, and viral replication. Similar to the potent antiviral drug remdesivir, rutin, propolis ethanolic extract, and propolis liposomes inhibited non-structural proteins of SARS-CoV-2 in vitro, and these compounds along with naringin inhibited SARS-CoV-2 infection in Vero E6 cells. Propolis extracts delivered by nanocarriers exhibit better antiviral effects against SARS-CoV-2 than ethanolic extracts. In line, hospitalized COVID-19 patients receiving green Brazilian propolis or a combination of honey and Nigella sativa exhibited earlier viral clearance, symptom recovery, discharge from the hospital as well as less mortality than counterparts receiving standard care alone. Thus, the use of bee products as an adjuvant treatment for COVID-19 may produce beneficial effects. Implications for treatment outcomes and issues to be considered in future studies are discussed.
Collapse
Affiliation(s)
- Amira Mohammed Ali
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-0031, Japan;
- Department of Psychiatric Nursing and Mental Health, Faculty of Nursing, Alexandria University, Alexandria 21527, Egypt
| | - Hiroshi Kunugi
- Department of Mental Disorder Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo 187-0031, Japan;
- Department of Psychiatry, Teikyo University School of Medicine, Tokyo 173-8605, Japan
| |
Collapse
|
208
|
Abstract
The ongoing pandemic of the new severe acute respiratory syndrome coronavirus (SARS-CoV-2) has caused more than one million deaths, overwhelmed many public health systems, and led to a worldwide economic recession. This has raised an unprecedented need to develop antiviral drugs and vaccines, which requires profound knowledge of the fundamental pathology of the virus, including its entry, replication, and release from host cells. The genome of coronaviruses comprises around 30 kb of positive single-stranded RNA, representing one of the largest RNA genomes of viruses. The 5′ part of the genome encodes a large polyprotein, PP1ab, which gives rise to 16 non-structural proteins (nsp1– nsp16). Two proteases encoded in nsp3 and nsp5 cleave the polyprotein into individual proteins. Most nsps belong to the viral replicase complex that promotes replication of the viral genome and translation of structural proteins by producing subgenomic mRNAs. The replicase complexes are found on double-membrane vesicles (DMVs) that contain viral double-stranded RNA. Expression of a small subset of viral proteins, including nsp3 and nsp4, is sufficient to induce formation of these DMVs in human cells, suggesting that both proteins deform host membranes into such structures. We will discuss the formation of DMVs and provide an overview of other membrane remodeling processes that are induced by coronaviruses.
Collapse
Affiliation(s)
- Jagan Mohan
- Membrane Biochemistry and Transport, Institut Pasteur, UMR3691 CNRS, F-75015, Paris, France
| | - Thomas Wollert
- Membrane Biochemistry and Transport, Institut Pasteur, UMR3691 CNRS, F-75015, Paris, France
| |
Collapse
|
209
|
Traversi D, Pulliero A, Izzotti A, Franchitti E, Iacoviello L, Gianfagna F, Gialluisi A, Izzi B, Agodi A, Barchitta M, Calabrò GE, Hoxhaj I, Sassano M, Sbrogiò LG, Del Sole A, Marchiori F, Pitini E, Migliara G, Marzuillo C, De Vito C, Tamburro M, Sammarco ML, Ripabelli G, Villari P, Boccia S. Precision Medicine and Public Health: New Challenges for Effective and Sustainable Health. J Pers Med 2021; 11:135. [PMID: 33669364 PMCID: PMC7920275 DOI: 10.3390/jpm11020135] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/11/2021] [Accepted: 02/14/2021] [Indexed: 02/06/2023] Open
Abstract
The development of high-throughput omics technologies represents an unmissable opportunity for evidence-based prevention of adverse effects on human health. However, the applicability and access to multi-omics tests are limited. In Italy, this is due to the rapid increase of knowledge and the high levels of skill and economic investment initially necessary. The fields of human genetics and public health have highlighted the relevance of an implementation strategy at a national level in Italy, including integration in sanitary regulations and governance instruments. In this review, the emerging field of public health genomics is discussed, including the polygenic scores approach, epigenetic modulation, nutrigenomics, and microbiomes implications. Moreover, the Italian state of implementation is presented. The omics sciences have important implications for the prevention of both communicable and noncommunicable diseases, especially because they can be used to assess the health status during the whole course of life. An effective population health gain is possible if omics tools are implemented for each person after a preliminary assessment of effectiveness in the medium to long term.
Collapse
Affiliation(s)
- Deborah Traversi
- Department of Public Health and Pediatrics, University of Torino, Piazza Polonia 94, 10126 Torino, Italy;
| | - Alessandra Pulliero
- Department of Health Sciences School of Medicine, University of Genoa, 16132 Genova, Italy;
| | - Alberto Izzotti
- Department of Experimental Medicine, University of Genoa, 16132 Genova, Italy;
- IRCCS Ospedale Policlinico San Martino, 161632 Genova, Italy
| | - Elena Franchitti
- Department of Public Health and Pediatrics, University of Torino, Piazza Polonia 94, 10126 Torino, Italy;
| | - Licia Iacoviello
- Research Center in Epidemiology and Preventive Medicine (EPIMED), Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (L.I.); (F.G.)
- Department of Epidemiology and Prevention, IRCCS NEUROMED, 86077 Pozzilli, Italy; (A.G.); (B.I.)
| | - Francesco Gianfagna
- Research Center in Epidemiology and Preventive Medicine (EPIMED), Department of Medicine and Surgery, University of Insubria, 21100 Varese, Italy; (L.I.); (F.G.)
- Mediterranea Cardiocentro, 80122 Napoli, Italy
| | - Alessandro Gialluisi
- Department of Epidemiology and Prevention, IRCCS NEUROMED, 86077 Pozzilli, Italy; (A.G.); (B.I.)
| | - Benedetta Izzi
- Department of Epidemiology and Prevention, IRCCS NEUROMED, 86077 Pozzilli, Italy; (A.G.); (B.I.)
| | - Antonella Agodi
- Department of Medical and Surgical Sciences and Advanced Technologies “GF Ingrassia”, University of Catania, 95123 Catania, Italy; (A.A.); (M.B.)
| | - Martina Barchitta
- Department of Medical and Surgical Sciences and Advanced Technologies “GF Ingrassia”, University of Catania, 95123 Catania, Italy; (A.A.); (M.B.)
| | - Giovanna Elisa Calabrò
- Section of Hygiene, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.E.C.); (I.H.); (M.S.); (S.B.)
| | - Ilda Hoxhaj
- Section of Hygiene, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.E.C.); (I.H.); (M.S.); (S.B.)
| | - Michele Sassano
- Section of Hygiene, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.E.C.); (I.H.); (M.S.); (S.B.)
| | - Luca Gino Sbrogiò
- Dipartimento di Prevenzione, Az. ULSS3 Serenissima, 30174 Venezia, Italy;
| | | | | | - Erica Pitini
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Roma, Italy; (E.P.); (G.M.); (C.M.); (C.D.V.); (P.V.)
| | - Giuseppe Migliara
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Roma, Italy; (E.P.); (G.M.); (C.M.); (C.D.V.); (P.V.)
| | - Carolina Marzuillo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Roma, Italy; (E.P.); (G.M.); (C.M.); (C.D.V.); (P.V.)
| | - Corrado De Vito
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Roma, Italy; (E.P.); (G.M.); (C.M.); (C.D.V.); (P.V.)
| | - Manuela Tamburro
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, 86100 Campobasso, Italy; (M.T.); (M.L.S.); (G.R.)
| | - Michela Lucia Sammarco
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, 86100 Campobasso, Italy; (M.T.); (M.L.S.); (G.R.)
| | - Giancarlo Ripabelli
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, University of Molise, 86100 Campobasso, Italy; (M.T.); (M.L.S.); (G.R.)
| | - Paolo Villari
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00185 Roma, Italy; (E.P.); (G.M.); (C.M.); (C.D.V.); (P.V.)
| | - Stefania Boccia
- Section of Hygiene, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, 00168 Roma, Italy; (G.E.C.); (I.H.); (M.S.); (S.B.)
- Department of Woman and Child Health and Public Health-Public Health Area, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00168 Roma, Italy
| |
Collapse
|
210
|
Genomic-informed pathogen surveillance in Africa: opportunities and challenges. THE LANCET. INFECTIOUS DISEASES 2021; 21:e281-e289. [PMID: 33587898 PMCID: PMC7906676 DOI: 10.1016/s1473-3099(20)30939-7] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/18/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022]
Abstract
The ongoing COVID-19 pandemic has highlighted the need to incorporate pathogen genomics for enhanced disease surveillance and outbreak management in Africa. The genomics of SARS-CoV-2 has been instrumental to the timely development of diagnostics and vaccines and in elucidating transmission dynamics. Global disease control programmes, including those for tuberculosis, malaria, HIV, foodborne pathogens, and antimicrobial resistance, also recommend genomics-based surveillance as an integral strategy towards control and elimination of these diseases. Despite the potential benefits, capacity remains low for many public health programmes in Africa. The COVID-19 pandemic presents an opportunity to reassess and strengthen surveillance systems and potentially integrate emerging technologies for preparedness of future epidemics and control of endemic diseases. We discuss opportunities and challenges for integrating pathogen genomics into public health surveillance systems in Africa. Improving accessibility through the creation of functional continent-wide networks, building multipathogen sequencing cores, training a critical mass of local experts, development of standards and policies to facilitate best practices for data sharing, and establishing a community of practice of genomics experts are all needed to use genomics for improved disease surveillance in Africa. Coordination and leadership are also crucial, which the Africa Centres for Disease Control and Prevention seeks to provide through its institute for pathogen genomics.
Collapse
|
211
|
Flores-Alanis A, Cruz-Rangel A, Rodríguez-Gómez F, González J, Torres-Guerrero CA, Delgado G, Cravioto A, Morales-Espinosa R. Molecular Epidemiology Surveillance of SARS-CoV-2: Mutations and Genetic Diversity One Year after Emerging. Pathogens 2021; 10:184. [PMID: 33572190 PMCID: PMC7915391 DOI: 10.3390/pathogens10020184] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/02/2021] [Accepted: 02/06/2021] [Indexed: 12/16/2022] Open
Abstract
In December 2019, the first cases of the novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) were identified in the city of Wuhan, China. Since then, it has spread worldwide with new mutations being reported. The aim of the present study was to monitor the changes in genetic diversity and track non-synonymous substitutions (dN) that could be implicated in the fitness of SARS-CoV-2 and its spread in different regions between December 2019 and November 2020. We analyzed 2213 complete genomes from six geographical regions worldwide, which were downloaded from GenBank and GISAID databases. Although SARS-CoV-2 presented low genetic diversity, there has been an increase over time, with the presence of several hotspot mutations throughout its genome. We identified seven frequent mutations that resulted in dN substitutions. Two of them, C14408T>P323L and A23403G>D614G, located in the nsp12 and Spike protein, respectively, emerged early in the pandemic and showed a considerable increase in frequency over time. Two other mutations, A1163T>I120F in nsp2 and G22992A>S477N in the Spike protein, emerged recently and have spread in Oceania and Europe. There were associations of P323L, D614G, R203K and G204R substitutions with disease severity. Continuous molecular surveillance of SARS-CoV-2 will be necessary to detect and describe the transmission dynamics of new variants of the virus with clinical relevance. This information is important to improve programs to control the virus.
Collapse
Affiliation(s)
- Alejandro Flores-Alanis
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico; (A.F.-A.); (G.D.); (A.C.)
| | - Armando Cruz-Rangel
- Laboratorio de Bioquímica de Enfermedades Crónicas, Instituto Nacional de Medicina Genómica, Mexico City 14610, Mexico;
| | - Flor Rodríguez-Gómez
- Departamento de Ciencias Computacionales, Centro Universitario de Ciencias Exactas e Ingenierías, Universidad de Guadalajara, Guadalajara 44430, Jalisco, Mexico;
| | - James González
- Departamento de Biología Celular, Facultad de Ciencias, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico;
| | | | - Gabriela Delgado
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico; (A.F.-A.); (G.D.); (A.C.)
| | - Alejandro Cravioto
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico; (A.F.-A.); (G.D.); (A.C.)
| | - Rosario Morales-Espinosa
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Mexico City 04360, Mexico; (A.F.-A.); (G.D.); (A.C.)
| |
Collapse
|
212
|
Borghesi A, Golemi S, Carapella N, Zigliani A, Farina D, Maroldi R. Lombardy, Northern Italy: COVID-19 second wave less severe and deadly than the first? A preliminary investigation. Infect Dis (Lond) 2021; 53:370-375. [PMID: 33560897 DOI: 10.1080/23744235.2021.1884745] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The much-heralded second wave of coronavirus disease (COVID-19) has arrived in Italy. Right now, one of the main questions about COVID-19 is whether the second wave is less severe and deadly than the first wave. In order to answer this challenging question, we decided to evaluate the chest X-ray (CXR) severity of COVID-19 pneumonia, the mechanical ventilation (MV) use, the patient outcome, and certain clinical/laboratory data during the second wave and compare them with those of the first wave. METHODS During the two COVID-19 waves two independent groups of hospitalised patients were selected. The first group consisted of the first 100 COVID-19 patients admitted to our hospital during the first wave. The second group consisted of another 100 consecutive COVID-19 patients admitted to our hospital during the second wave. We enlisted only Caucasian male patients over the age of fifty for whom the final outcome was available. For each patient, the CXR severity of COVID-19 pneumonia, the MV use, the patient outcome, comorbidities, corticosteroid use, and C-reactive protein (CRP) levels were considered. Nonparametric statistical tests were used to compare the data obtained from the two waves. RESULTS The CXR severity of COVID-19 pneumonia, the in-hospital mortality, and CRP levels were significantly higher in the first wave than in the second wave (p ≤ .041). Although not statistically significant, the frequency of MV use was higher in the first wave. CONCLUSIONS This preliminary investigation seems to confirm that the COVID-19 second wave is less severe and deadly than the first wave.
Collapse
Affiliation(s)
- Andrea Borghesi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Salvatore Golemi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Nicola Carapella
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Angelo Zigliani
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Davide Farina
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Roberto Maroldi
- Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| |
Collapse
|
213
|
Chaibun T, Puenpa J, Ngamdee T, Boonapatcharoen N, Athamanolap P, O'Mullane AP, Vongpunsawad S, Poovorawan Y, Lee SY, Lertanantawong B. Rapid electrochemical detection of coronavirus SARS-CoV-2. Nat Commun 2021; 12:802. [PMID: 33547323 PMCID: PMC7864991 DOI: 10.1038/s41467-021-21121-7] [Citation(s) in RCA: 267] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/11/2021] [Indexed: 12/20/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a highly contagious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Diagnosis of COVID-19 depends on quantitative reverse transcription PCR (qRT-PCR), which is time-consuming and requires expensive instrumentation. Here, we report an ultrasensitive electrochemical biosensor based on isothermal rolling circle amplification (RCA) for rapid detection of SARS-CoV-2. The assay involves the hybridization of the RCA amplicons with probes that were functionalized with redox active labels that are detectable by an electrochemical biosensor. The one-step sandwich hybridization assay could detect as low as 1 copy/μL of N and S genes, in less than 2 h. Sensor evaluation with 106 clinical samples, including 41 SARS-CoV-2 positive and 9 samples positive for other respiratory viruses, gave a 100% concordance result with qRT-PCR, with complete correlation between the biosensor current signals and quantitation cycle (Cq) values. In summary, this biosensor could be used as an on-site, real-time diagnostic test for COVID-19.
Collapse
Affiliation(s)
- Thanyarat Chaibun
- Biosensors Laboratory, Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| | - Jiratchaya Puenpa
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Tatchanun Ngamdee
- Department of Biotechnology, School of Bioresources and Technology, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Nimaradee Boonapatcharoen
- Pilot Plant Development and Training Institute (PDTI), King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Pornpat Athamanolap
- Biosensors Laboratory, Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand
| | - Anthony Peter O'Mullane
- School of Chemistry and Physics, Queensland University of Technology (QUT), Brisbane, QLD, Australia
| | - Sompong Vongpunsawad
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Yong Poovorawan
- Center of Excellence in Clinical Virology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Su Yin Lee
- Faculty of Applied Sciences, AIMST University, Bedong, Kedah, Malaysia
- Centre of Excellence for Omics-Driven Computational Biodiscovery (COMBio), AIMST University, Bedong, Kedah, Malaysia
| | - Benchaporn Lertanantawong
- Biosensors Laboratory, Department of Biomedical Engineering, Faculty of Engineering, Mahidol University, Nakhon Pathom, Thailand.
| |
Collapse
|
214
|
Ko K, Nagashima S, E. B, Ouoba S, Akita T, Sugiyama A, Ohisa M, Sakaguchi T, Tahara H, Ohge H, Ohdan H, Kubo T, Kishita E, Kuwabara M, Takahashi K, Tanaka J. Molecular characterization and the mutation pattern of SARS-CoV-2 during first and second wave outbreaks in Hiroshima, Japan. PLoS One 2021; 16:e0246383. [PMID: 33544733 PMCID: PMC7864667 DOI: 10.1371/journal.pone.0246383] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/18/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND In this study, we performed molecular characterization of SARS-CoV-2 strains in Hiroshima and its mutation pattern between the first and second waves of the outbreak. METHOD A total of 55 nasal swab samples from the first wave in Hiroshima and 13 from the second wave were examined quantitatively by RT-qPCR and qualitatively by nested PCR using specific primers. Four samples from each wave underwent next-generation sequencing and phylogenetic tree analysis including controls and all sequences retrieved in Japan from GISAID and GenBank. Subsequently, mutations were examined. RESULTS Viral load ranged 7.85 × 101-1.42 × 108 copies/ml. Of 68 samples, one was Asian type-O, 65 were European type-GR, and 2 were undetectable. Phylogenetic tree analysis indicated that Japan was infected with various Asian strains (L, S, V, O) from January through April. By second week of March, European strains (G, GH, GR) had appeared, and GR strains became predominant after mid-March. The first case in Hiroshima was classified as Asian strain O, and the rest were GR strains. Then, second wave of GR strains appeared independently with 11-15 base mutations. Comparing the first- and second-wave GR strains, mutation rate was 1.17-1.36 × 10-3 base substitutions per site per year; in addition, amino acid changes occurred at S1361P and P3371S in ORF1a, A314V in ORF1b, and P151L in N. All seven GR strains were D614G variants with R202K and G203R mutations in N. A single-nucleotide insertion in ORF8 that causes a defect in ORF8 protein was found in one isolate (S66) from the second wave. CONCLUSION Our findings reveal the evolutionary hierarchy of SARS-CoV-2 in Japan. The predominant D614G variants and a new form of ORF8 deletion in Hiroshima provide the clue for role of viral factor in local outbreaks of SARS-CoV-2.
Collapse
Affiliation(s)
- Ko Ko
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Shintaro Nagashima
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Bunthen E.
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Payment Certification Agency, Ministry of Health, Phnom Penh, Cambodia
| | - Serge Ouoba
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- Unité de Recherche Clinique de Nanoro (URCN), Nanoro, Burkina Faso
| | - Tomoyuki Akita
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Aya Sugiyama
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Masayuki Ohisa
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Takemasa Sakaguchi
- Department of Virology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hidetoshi Tahara
- Department of Cellular and Molecular Biology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroki Ohge
- Department of Infectious Diseases, Hiroshima University Hospital, Hiroshima, Japan
| | - Hideki Ohdan
- Department of Gastroenterological and Transplant Surgery, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Tatsuhiko Kubo
- Department of Public Health and Health Policy, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Eisaku Kishita
- Hiroshima Prefecture Health and Welfare Bureau, Hiroshima, Japan
| | - Masao Kuwabara
- Hiroshima Prefectural Center for Disease Control and Prevention, Hiroshima, Japan
| | - Kazuaki Takahashi
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Junko Tanaka
- Department of Epidemiology, Infectious Disease Control and Prevention, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
- * E-mail:
| |
Collapse
|
215
|
Dumonteil E, Fusco D, Drouin A, Herrera C. Genomic Signatures of SARS-CoV-2 Associated with Patient Mortality. Viruses 2021; 13:227. [PMID: 33540576 PMCID: PMC7912856 DOI: 10.3390/v13020227] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [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: 01/12/2021] [Accepted: 01/27/2021] [Indexed: 12/20/2022] Open
Abstract
Infections with SARS-CoV-2 can progress toward multiple clinical outcomes, and the identification of factors associated with disease severity would represent a major advance to guide care and improve prognosis. We tested for associations between SARS-CoV-2 genomic variants from an international cohort of 2508 patients and mortality rates. Findings were validated in a second cohort. Phylogenetic analysis of SARS-CoV-2 genome sequences revealed four well-resolved clades which had significantly different mortality rates, even after adjusting for patient demographic and geographic characteristics. We further identified ten single-nucleotide polymorphisms (SNPs) in the SARS-CoV-2 genome that were associated with patient mortality. Three SNPs remained associated with mortality in a generalized linear model (GLM) that also included patient age, sex, geographic region, and month of sample collection. Multiple SNPs were confirmed in the validation cohort. These SNPs represent targets to assess the mechanisms underlying COVID-19 disease severity and warrant straightforward validation in functional studies.
Collapse
Affiliation(s)
- Eric Dumonteil
- Department of Tropical Medicine, Vector-Borne and Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA; (D.F.); (C.H.)
| | - Dahlene Fusco
- Department of Tropical Medicine, Vector-Borne and Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA; (D.F.); (C.H.)
- Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA;
| | - Arnaud Drouin
- Department of Medicine, School of Medicine, Tulane University, New Orleans, LA 70112, USA;
- Department of Pathology, School of Medicine, Tulane University, New Orleans, LA 70112, USA
| | - Claudia Herrera
- Department of Tropical Medicine, Vector-Borne and Infectious Disease Research Center, School of Public Health and Tropical Medicine, Tulane University, New Orleans, LA 70112, USA; (D.F.); (C.H.)
| |
Collapse
|
216
|
Islam MT, Alam ARU, Sakib N, Hasan MS, Chakrovarty T, Tawyabur M, Islam OK, Al-Emran HM, Jahid MIK, Anwar Hossain M. A rapid and cost-effective multiplex ARMS-PCR method for the simultaneous genotyping of the circulating SARS-CoV-2 phylogenetic clades. J Med Virol 2021; 93:2962-2970. [PMID: 33491822 PMCID: PMC8014803 DOI: 10.1002/jmv.26818] [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: 12/26/2020] [Revised: 01/15/2021] [Accepted: 01/20/2021] [Indexed: 01/09/2023]
Abstract
Tracing the globally circulating severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2) phylogenetic clades by high‐throughput sequencing is costly, time‐consuming, and labor‐intensive. We here propose a rapid, simple, and cost‐effective amplification refractory mutation system (ARMS)‐based multiplex reverse‐transcription polymerase chain reaction (PCR) assay to identify six distinct phylogenetic clades: S, L, V, G, GH, and GR. Our multiplex PCR is designed in a mutually exclusive way to identify V–S and G–GH–GR clade variants separately. The pentaplex assay included all five variants and the quadruplex comprised of the triplex variants alongside either V or S clade mutations that created two separate subsets. The procedure was optimized with 0.2–0.6 µM primer concentration, 56–60°C annealing temperature, and 3–5 ng/µl complementary DNA to validate on 24 COVID‐19‐positive samples. Targeted Sanger sequencing further confirmed the presence of the clade‐featured mutations with another set of primers. This multiplex ARMS‐PCR assay is a fast, low‐cost alternative and convenient to discriminate the circulating phylogenetic clades of SARS‐CoV‐2. Multiplex ARMS‐PCR (amplification refractory mutation system‐polymerase chain reaction) method for genotyping major severe acute respiratory syndrome coronavirus 2 (SARS‐CoV‐2 clades). Identify the mutated region of circulating phylogenetically SARS‐CoV‐2 clades. PCR conditions were optimized and validated to identify V–S and G–GH–GR clade.
Collapse
Affiliation(s)
- Mohammad Tanvir Islam
- Department of Microbiology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Asm Rubayet Ul Alam
- Department of Microbiology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Najmuj Sakib
- Department of Microbiology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Mohammad Shazid Hasan
- Department of Microbiology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Tanay Chakrovarty
- Department of Microbiology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Mohammad Tawyabur
- Department of Microbiology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Ovinu Kibria Islam
- Department of Microbiology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Hassan M Al-Emran
- Department of Biomedical Engineering, Jashore University of Science and Technology, Jashore, Bangladesh
| | | | - Mohammad Anwar Hossain
- Department of Microbiology, Jashore University of Science and Technology, Jashore, Bangladesh.,Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
217
|
Tzachor A, Rozen O, Khatib S, Jensen S, Avni D. Photosynthetically Controlled Spirulina, but Not Solar Spirulina, Inhibits TNF-α Secretion: Potential Implications for COVID-19-Related Cytokine Storm Therapy. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2021; 23:149-155. [PMID: 33566210 PMCID: PMC7874025 DOI: 10.1007/s10126-021-10020-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 01/13/2021] [Indexed: 05/30/2023]
Abstract
An array of infections, including the novel coronavirus (SARS-CoV-2), trigger macrophage activation syndrome (MAS) and subsequently hypercytokinemia, commonly referred to as a cytokine storm (CS). It is postulated that CS is mainly responsible for critical COVID-19 cases, including acute respiratory distress syndrome (ARDS). Recognizing the therapeutic potential of Spirulina blue-green algae (Arthrospira platensis), in this in vitro stimulation study, LPS-activated macrophages and monocytes were treated with aqueous extracts of Spirulina, cultivated in either natural or controlled light conditions. We report that an extract of photosynthetically controlled Spirulina (LED Spirulina), at a concentration of 0.1 µg/mL, decreases macrophage and monocyte-induced TNF-α secretion levels by over 70% and 40%, respectively. We propose prompt in vivo studies in animal models and human subjects to determine the putative effectiveness of a natural, algae-based treatment for viral CS and ARDS, and explore the potential of a novel anti-TNF-α therapy.
Collapse
Affiliation(s)
- Asaf Tzachor
- Centre for the Study of Existential Risk & Cambridge Global Food Security Research Center, University of Cambridge, Cambridge, UK
| | - Or Rozen
- Sphingolipids, Active Metabolites and Immune Modulation Laboratory, MIGAL - Galilee Research Institute, Kiryat Shemona, Israel
| | - Soliman Khatib
- Natural compounds and analytical chemistry Laboratory, MIGAL - Galilee Research Institute and Tel Hai college, Kiryat Shemona, Israel
| | - Sophie Jensen
- MATIS - Food and Biotech Research and Development, Reykjavík, Iceland
| | - Dorit Avni
- Sphingolipids, Active Metabolites and Immune Modulation Laboratory, MIGAL - Galilee Research Institute, Kiryat Shemona, Israel.
| |
Collapse
|
218
|
Kotaki T, Xie X, Shi PY, Kameoka M. A PCR amplicon-based SARS-CoV-2 replicon for antiviral evaluation. Sci Rep 2021; 11:2229. [PMID: 33500537 PMCID: PMC7838314 DOI: 10.1038/s41598-021-82055-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Accepted: 01/14/2021] [Indexed: 01/08/2023] Open
Abstract
The development of specific antiviral compounds to SARS-CoV-2 is an urgent task. One of the obstacles for the antiviral development is the requirement of biocontainment because infectious SARS-CoV-2 must be handled in a biosafety level-3 laboratory. Replicon, a non-infectious self-replicative viral RNA, could be a safe and effective tool for antiviral evaluation. Herein, we generated a PCR-based SARS-CoV-2 replicon. Eight fragments covering the entire SARS-CoV-2 genome except S, E, and M genes were amplified with HiBiT-tag sequence by PCR. The amplicons were ligated and in vitro transcribed to RNA. The cells electroporated with the replicon RNA showed more than 3000 times higher luminescence than MOCK control cells at 24 h post-electroporation, indicating robust translation and RNA replication of the replicon. The replication was drastically inhibited by remdesivir, an RNA polymerase inhibitor for SARS-CoV-2. The IC50 of remdesivir in this study was 0.29 μM, generally consistent to the IC50 obtained using infectious SARS-CoV-2 in a previous study (0.77 μM). Taken together, this system could be applied to the safe and effective antiviral evaluation without using infectious SARS-CoV-2. Because this is a PCR-based and transient replicon system, further improvement including the establishment of stable cell line must be achieved.
Collapse
Affiliation(s)
- Tomohiro Kotaki
- Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Masanori Kameoka
- Department of Public Health, Kobe University Graduate School of Health Sciences, 7-10-2 Tomogaoka, Suma-ku, Kobe, Hyogo, 654-0142, Japan.
| |
Collapse
|
219
|
Vilar S, Isom DG. One Year of SARS-CoV-2: How Much Has the Virus Changed? BIOLOGY 2021; 10:91. [PMID: 33530355 PMCID: PMC7911924 DOI: 10.3390/biology10020091] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/15/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a worldwide crisis with profound effects on both public health and the economy. In order to combat the COVID-19 pandemic, research groups have shared viral genome sequence data through the Global Initiative on Sharing All Influenza Data (GISAID). Over the past year, ≈290,000 full SARS-CoV-2 proteome sequences have been deposited in the GISAID. Here, we used these sequences to assess the rate of nonsynonymous mutants over the entire viral proteome. Our analysis shows that SARS-CoV-2 proteins are mutating at substantially different rates, with most of the viral proteins exhibiting little mutational variability. As anticipated, our calculations capture previously reported mutations that arose in the first months of the pandemic, such as D614G (Spike), P323L (NSP12), and R203K/G204R (Nucleocapsid), but they also identify more recent mutations, such as A222V and L18F (Spike) and A220V (Nucleocapsid), among others. Our comprehensive temporal and geographical analyses show two distinct periods with different proteome mutation rates: December 2019 to July 2020 and August to December 2020. Notably, some mutation rates differ by geography, primarily during the latter half of 2020 in Europe. Furthermore, our structure-based molecular analysis provides an exhaustive assessment of SARS-CoV-2 mutation rates in the context of the current set of 3D structures available for SARS-CoV-2 proteins. This emerging sequence-to-structure insight is beginning to illuminate the site-specific mutational (in)tolerance of SARS-CoV-2 proteins as the virus continues to spread around the globe.
Collapse
Affiliation(s)
- Santiago Vilar
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Daniel G. Isom
- Department of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL 33136, USA
- Institute for Data Science and Computing, University of Miami, Coral Gables, FL 33146, USA
| |
Collapse
|
220
|
Janik E, Bartos M, Niemcewicz M, Gorniak L, Bijak M. SARS-CoV-2: Outline, Prevention, and Decontamination. Pathogens 2021; 10:114. [PMID: 33498679 PMCID: PMC7911301 DOI: 10.3390/pathogens10020114] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 02/06/2023] Open
Abstract
The new coronavirus began to spread around the world in late 2019. Initially, it was found only in China, but in the following days there were reported cases of infections in other countries. Subsequently, based on taxonomy, phylogeny, and accepted practice, the virus was officially designated as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). As a result of the rapid spread of SARS-CoV-2 in different countries around the world, on March 11, 2020, the World Health Organization (WHO) announced a status change in the disease caused by this coronavirus-from an epidemic to a pandemic disease. Although the world is taking unprecedented efforts to control the spread of SARS-CoV-2, the number of confirmed cases is rising. Therefore, effective preventive measures are needed in order to limit the spread of illness. The prevention measures are mainly based on information on the virus transmission routes, its environmental stability, and persistence on commonly touched surfaces. Social distancing, mask usage, and good hygiene practice are the most important recommendations for general public. Healthcare professionals who are directly involved in SARS-CoV-2 patients care are more exposed to virus infection and additional protection measures are necessary, including protective suits, aprons, face shields, goggles, and gloves. Due to the stability of SARS-CoV-2 on different surfaces, such as glass, paper, or wood, proper disinfection is crucial. Several studies have shown that despite the virus's stability, it is sensitive to various disinfectants, such as ethanol, isopropanol, sodium hypochlorite, or hydrogen peroxide. These findings underline the importance of having comprehensive knowledge about SARS-CoV-2 and multidirectional strategies in order to limit the spread of the virus. This review is a summary of the most important information about SARS-CoV-2, such as its stability on different surfaces, protection strategies, and decontamination options.
Collapse
Affiliation(s)
- Edyta Janik
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland. (E.J.); (M.N.); (L.G.)
| | - Maciej Bartos
- Department of Biodiversity Studies and Bioeducation, Faculty of Biology and Environmental Protection, University of Lodz, Banacha 1/3, 90-237 Lodz, Poland.
| | - Marcin Niemcewicz
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland. (E.J.); (M.N.); (L.G.)
| | - Leslaw Gorniak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland. (E.J.); (M.N.); (L.G.)
| | - Michal Bijak
- Biohazard Prevention Centre, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland. (E.J.); (M.N.); (L.G.)
| |
Collapse
|
221
|
Varahachalam SP, Lahooti B, Chamaneh M, Bagchi S, Chhibber T, Morris K, Bolanos JF, Kim NY, Kaushik A. Nanomedicine for the SARS-CoV-2: State-of-the-Art and Future Prospects. Int J Nanomedicine 2021; 16:539-560. [PMID: 33519200 PMCID: PMC7837559 DOI: 10.2147/ijn.s283686] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/25/2020] [Indexed: 01/08/2023] Open
Abstract
The newly emerged ribonucleic acid (RNA) enveloped human beta-coronavirus, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection caused the COVID-19 pandemic, severely affects the respiratory system, and may lead to death. Lacking effective diagnostics and therapies made this pandemic challenging to manage since the SARS-CoV-2 transmits via human-to-human, enters via ACE2 and TMPSSR2 receptors, and damages organs rich in host cells, spreads via symptomatic carriers and is prominent in an immune-compromised population. New SARS-CoV-2 informatics (structure, strains, like-cycles, functional sites) motivated bio-pharma experts to investigate novel therapeutic agents that act to recognize, inhibit, and knockdown combinations of drugs, vaccines, and antibodies, have been optimized to manage COVID-19. However, successful targeted delivery of these agents to avoid off-targeting and unnecessary drug ingestion is very challenging. To overcome these obstacles, this mini-review projects nanomedicine technology, a pharmacologically relevant cargo of size within 10 to 200 nm, for site-specific delivery of a therapeutic agent to recognize and eradicate the SARS-CoV-2, and improving the human immune system. Such combinational therapy based on compartmentalization controls the delivery and releases of a drug optimized based on patient genomic profile and medical history. Nanotechnology could help combat COVID-19 via various methods such as avoiding viral contamination and spraying by developing personal protective equipment (PPE) to increase the protection of healthcare workers and produce effective antiviral disinfectants surface coatings capable of inactivating and preventing the virus from spreading. To quickly recognize the infection or immunological response, design highly accurate and sensitive nano-based sensors. Development of new drugs with improved activity, reduced toxicity, and sustained release to the lungs, as well as tissue targets; and development of nano-based immunizations to improve humoral and cellular immune responses. The desired and controlled features of suggested personalized therapeutics, nanomedicine, is a potential therapy to manage COVID-19 successfully. The state-of-the-art nanomedicine, challenges, and prospects of nanomedicine are carefully and critically discussed in this report, which may serve as a key platform for scholars to investigate the role of nanomedicine for higher efficacy to manage the COVID-19 pandemic.
Collapse
Affiliation(s)
- Sree Pooja Varahachalam
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX79106, USA
| | - Behnaz Lahooti
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX79106, USA
| | - Masoumeh Chamaneh
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX79106, USA
| | - Sounak Bagchi
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX79106, USA
| | - Tanya Chhibber
- Department of Pharmaceutical Sciences, Jerry H. Hodge School of Pharmacy, Texas Tech University Health Sciences Center (TTUHSC), Amarillo, TX79106, USA
| | - Kevin Morris
- Maharashtra University of Health Sciences (MUHS), Nashik, Maharashtra422004, India
| | - Joe F Bolanos
- Facultad De Ciencias De La Salud “Dr.Luis Edmundo Vasquez” Santa Tecla, Universidad Dr. Jose Matias Delgado, Cd Merliot, El Salvador
| | - Nam-Young Kim
- RFIC Bio Center, Department of Electronics Engineering, Kwangwoon University, Seoul01897, South Korea
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Natural Sciences, Division of Sciences, Art, and Mathematics, Florida Polytechnic University, Lakeland, FL3385, USA
| |
Collapse
|
222
|
Chilamakuri R, Agarwal S. COVID-19: Characteristics and Therapeutics. Cells 2021; 10:206. [PMID: 33494237 PMCID: PMC7909801 DOI: 10.3390/cells10020206] [Citation(s) in RCA: 167] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 01/11/2021] [Accepted: 01/18/2021] [Indexed: 02/07/2023] Open
Abstract
Novel coronavirus (COVID-19 or 2019-nCoV or SARS-CoV-2), which suddenly emerged in December 2019 is still haunting the entire human race and has affected not only the healthcare system but also the global socioeconomic balances. COVID-19 was quickly designated as a global pandemic by the World Health Organization as there have been about 98.0 million confirmed cases and about 2.0 million confirmed deaths, as of January 2021. Although, our understanding of COVID-19 has significantly increased since its outbreak, and multiple treatment approaches and pharmacological interventions have been tested or are currently under development to mitigate its risk-factors. Recently, some vaccine candidates showed around 95% clinical efficacy, and now receiving emergency use approvals in different countries. US FDA recently approved BNT162 and mRNA-1273 vaccines developed by Pfizer/BioNTech and Moderna Inc. for emergency use and vaccination in the USA. In this review, we present a succinct overview of the SARS-CoV-2 virus structure, molecular mechanisms of infection, COVID-19 epidemiology, diagnosis, and clinical manifestations. We also systematize different treatment strategies and clinical trials initiated after the pandemic outbreak, based on viral infection and replication mechanisms. Additionally, we reviewed the novel pharmacological intervention approaches and vaccine development strategies against COVID-19. We speculate that the current pandemic emergency will trigger detailed studies of coronaviruses, their mechanism of infection, development of systematic drug repurposing approaches, and novel drug discoveries for current and future pandemic outbreaks.
Collapse
Affiliation(s)
| | - Saurabh Agarwal
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, 8000 Utopia Parkway, Queens, NY 11439, USA;
| |
Collapse
|
223
|
Abstract
SARS-CoV-2, with an infection fatality rate between 0.5 and 1%, has spread to all corners of the globe and infected millions of people. While vaccination is essential to protect against the virus and halt community transmission, rapidly making and delivering safe and efficacious vaccines presents unique development, manufacturing, supply chain, delivery, and post-market surveillance challenges. Despite the large number of vaccines in or entering the clinic, it is unclear how many candidates will meet regulatory requirements and which vaccine strategy will most effectively lead to sustained, population-wide immunity. Interviews with experts from biopharmaceutical companies, regulatory and multilateral organizations, non-profit foundations, and academic research groups, complemented with extensive literature review, informed the development of a framework for understanding the factors leading to population-wide immunity against SARS-CoV-2, in particular considering the role of vaccines. This paper presents a systems-level modeling framework to guide the development of analytical tools aimed at informing time-critical decisions to make vaccines globally and equitably accessible. Such a framework can be used for scenario planning and evaluating tradeoffs across access strategies. It highlights the diverse and powerful ways in which data can be used to evaluate future risks and strategically allocate limited resources.
Collapse
|
224
|
Shagufta, Ahmad I. The race to treat COVID-19: Potential therapeutic agents for the prevention and treatment of SARS-CoV-2. Eur J Med Chem 2021; 213:113157. [PMID: 33486200 PMCID: PMC7802596 DOI: 10.1016/j.ejmech.2021.113157] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 12/10/2020] [Accepted: 01/02/2021] [Indexed: 12/13/2022]
Abstract
The unforeseen emergence of coronavirus disease 2019 (COVID-19), a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) at the Wuhan province of China in December 2019, subsequently its abrupt spread across the world has severely affected human life. In a short span of time, COVID-19 has sacked more than one million human lives and marked as a severe global pandemic, which is drastically accountable for the adverse effect directly to the human society, particularly the health care system and the economy. The unavailability of approved and effective drugs or vaccines against COVID-19 further created conditions more adverse and terrifying. To win the war against this pandemic within time there is a desperate need for the most adequate therapeutic treatment, which can be achieved by the collaborative research work among scientists worldwide. In continuation of our efforts to support the scientific community, a review has been presented which discusses the structure and the activity of numerous molecules exhibiting promising SARS-CoV-2 and other CoVs inhibition activities. Furthermore, this review offers an overview of the structure, a plausible mechanism of action of SARS-CoV-2, and crucial structural features substantial to inhibit the primary virus-based and host-based targets involved in SARS-CoV-2 treatment. We anticipate optimistically that this perspective will provide the reader and researcher’s better understanding regarding COVID-19 and pave the path in the direction of COVID-19 drug discovery and development paradigm.
Collapse
Affiliation(s)
- Shagufta
- Department of Mathematics and Natural Sciences, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, United Arab Emirates.
| | - Irshad Ahmad
- Department of Mathematics and Natural Sciences, School of Arts and Sciences, American University of Ras Al Khaimah, Ras Al Khaimah, United Arab Emirates
| |
Collapse
|
225
|
On the Application of Advanced Machine Learning Methods to Analyze Enhanced, Multimodal Data from Persons Infected with COVID-19. COMPUTATION 2021. [DOI: 10.3390/computation9010004] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The current COVID-19 pandemic, caused by the rapid worldwide spread of the SARS-CoV-2 virus, is having severe consequences for human health and the world economy. The virus affects different individuals differently, with many infected patients showing only mild symptoms, and others showing critical illness. To lessen the impact of the epidemic, one problem is to determine which factors play an important role in a patient’s progression of the disease. Here, we construct an enhanced COVID-19 structured dataset from more than one source, using natural language processing to add local weather conditions and country-specific research sentiment. The enhanced structured dataset contains 301,363 samples and 43 features, and we applied both machine learning algorithms and deep learning algorithms on it so as to forecast patient’s survival probability. In addition, we import alignment sequence data to improve the performance of the model. Application of Extreme Gradient Boosting (XGBoost) on the enhanced structured dataset achieves 97% accuracy in predicting patient’s survival; with climatic factors, and then age, showing the most importance. Similarly, the application of a Multi-Layer Perceptron (MLP) achieves 98% accuracy. This work suggests that enhancing the available data, mostly basic information on patients, so as to include additional, potentially important features, such as weather conditions, is useful. The explored models suggest that textual weather descriptions can improve outcome forecast.
Collapse
|
226
|
Zaide G, Cohen-Gihon I, Israeli O, Stein D, Shifman O, Weiss S, Simon I, Laskar O, Beth-Din A, Zvi A. Mutation Profile of SARS-CoV-2 Genome Sequences Originating from Eight Israeli Patient Isolates. Microbiol Resour Announc 2021; 10:e01387-20. [PMID: 33414330 PMCID: PMC8407753 DOI: 10.1128/mra.01387-20] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 12/10/2020] [Indexed: 12/17/2022] Open
Abstract
We report the genome sequences and the identification of genetic variations in eight clinical samples of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Samples were collected from nasopharyngeal swabs of symptomatic and asymptomatic individuals from five care homes for elderly and infirm persons in Israel. The sequences obtained are valuable, as they carry a newly reported nonsynonymous substitution located within the nucleoprotein open reading frame.
Collapse
Affiliation(s)
- Galia Zaide
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Inbar Cohen-Gihon
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ofir Israeli
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Dana Stein
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Ohad Shifman
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Shay Weiss
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Irit Simon
- Department of Biotechnology, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Orly Laskar
- Department of Infectious Diseases, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Adi Beth-Din
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| | - Anat Zvi
- Department of Biochemistry and Molecular Genetics, Israel Institute for Biological Research, Ness-Ziona, Israel
| |
Collapse
|
227
|
Bui CHT, Yeung HW, Ho JCW, Leung CYH, Hui KPY, Perera RAPM, Webby RJ, Schultz-Cherry SL, Nicholls JM, Peiris JSM, Chan MCW. Tropism of SARS-CoV-2, SARS-CoV and influenza virus in canine tissue explants. J Infect Dis 2021; 224:821-830. [PMID: 33395484 PMCID: PMC7799041 DOI: 10.1093/infdis/jiab002] [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: 10/19/2020] [Accepted: 01/04/2021] [Indexed: 01/04/2023] Open
Abstract
Background Human spillovers of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to dogs and the emergence of a highly contagious avian-origin H3N2 canine influenza virus have raised concerns on the role of dogs in the spread of SARS-CoV-2 and their susceptibility to existing human and avian influenza viruses, which might result in further reassortment. Methods We systematically studied the replication kinetics of SARS-CoV-2, SARS-CoV, influenza A viruses of H1, H3, H5, H7, and H9 subtypes, and influenza B viruses of Yamagata-like and Victoria-like lineages in ex vivo canine nasal cavity, soft palate, trachea, and lung tissue explant cultures and examined ACE2 and sialic acid (SA) receptor distribution in these tissues. Results There was limited productive replication of SARS-CoV-2 in canine nasal cavity and SARS-CoV in canine nasal cavity, soft palate, and lung, with unexpectedly high ACE2 levels in canine nasal cavity and soft palate. Canine tissues were susceptible to a wide range of human and avian influenza viruses, which matched with the abundance of both human and avian SA receptors. Conclusions Existence of suitable receptors and tropism for the same tissue foster virus adaptation and reassortment. Continuous surveillance in dog populations should be conducted given the many chances for spillover during outbreaks.
Collapse
Affiliation(s)
- Christine H T Bui
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - H W Yeung
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - John C W Ho
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Connie Y H Leung
- Centre for Comparative Medicine Research, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kenrie P Y Hui
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ranawaka A P M Perera
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Richard J Webby
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - Stacey L Schultz-Cherry
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, Tennessee, USA
| | - John M Nicholls
- Department of Pathology, Queen Mary Hospital, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - J S Malik Peiris
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Michael C W Chan
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
228
|
Chugh A, Khera D, Khasbage S, Singh S. Does bacille calmette–Guérin vaccination provides protection against COVID-19: A systematic review and meta-analysis. Indian J Community Med 2021; 46:592-599. [PMID: 35068716 PMCID: PMC8729290 DOI: 10.4103/ijcm.ijcm_952_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 06/14/2021] [Indexed: 11/18/2022] Open
Abstract
Background: Lower morbidity and mortality in few geographic locations on the globe suffering with SARS-CoV-2 has been associated with the existing or previously followed long-standing Bacille Calmette–Guérin (BCG) vaccination policy among infants. However, does it hold true that today after years of BCG vaccination, few adults have better prognosis or is it just confounding due to differential disease burden, population density, testing facilities, or improper reporting. The purpose was to evaluate and correlate this effect systematically. Methods: Detailed electronic search for randomized controlled trials (RCTs) and observational studies in PubMed, Cochrane Library, and ClinicalTrials.gov for eligible studies was performed. Results: One hundred and fourteen studies were yielded on search strategy and 28 observational studies were finally included for analysis. From our results, we can say that BCG vaccination causes a decrease in COVID-19 incidence and mortality. However, these results must be interpreted cautiously as lot of confounding factors were present in included studies, which can affect the outcome. Conclusion: The evidence of BCG vaccination for the protection against COVID-19 cannot be ruled out as evidence from many studies support the hypothesis, but the evidence of well-conducted RCTs and observational studies can strengthen the evidence. Registration Number: PROSPERO (International Prospective Register of Systematic Reviews) database (CRD42020204466).
Collapse
|
229
|
Ghazy RM, Almaghraby A, Shaaban R, Kamal A, Beshir H, Moursi A, Ramadan A, Taha SHN. A systematic review and meta-analysis on chloroquine and hydroxychloroquine as monotherapy or combined with azithromycin in COVID-19 treatment. Sci Rep 2020; 10:22139. [PMID: 33335141 PMCID: PMC7746770 DOI: 10.1038/s41598-020-77748-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 11/04/2020] [Indexed: 12/15/2022] Open
Abstract
Many recent studies have investigated the role of either Chloroquine (CQ) or Hydroxychloroquine (HCQ) alone or in combination with azithromycin (AZM) in the management of the emerging coronavirus. This systematic review and meta-analysis of either published or preprint observational studies or randomized control trials (RCT) aimed to assess mortality rate, duration of hospital stay, need for mechanical ventilation (MV), virologic cure rate (VQR), time to a negative viral polymerase chain reaction (PCR), radiological progression, experiencing drug side effects, and clinical worsening. A search of the online database through June 2020 was performed and examined the reference lists of pertinent articles for in-vivo studies only. Pooled relative risks (RRs), standard mean differences of 95% confidence intervals (CIs) were calculated with the random-effects model. Mortality was not different between the standard care (SC) and HCQ groups (RR = 0.99, 95% CI 0.61-1.59, I2 = 82%), meta-regression analysis proved that mortality was significantly different across the studies from different countries. However, mortality among the HCQ + AZM was significantly higher than among the SC (RR = 1.8, 95% CI 1.19-2.27, I2 = 70%). The duration of hospital stay in days was shorter in the SC in comparison with the HCQ group (standard mean difference = 0.57, 95% CI 0.20-0.94, I2 = 92%), or the HCQ + AZM (standard mean difference = 0.77, 95% CI 0.46-1.08, I2 = 81). Overall VQR, and that at days 4, 10, and 14 among patients exposed to HCQ did not differ significantly from the SC [(RR = 0.92, 95% CI 0.69-1.23, I2 = 67%), (RR = 1.11, 95% CI 0.26-4.69, I2 = 85%), (RR = 1.21, 95% CI 0.70-2.01, I2 = 95%), and (RR = 0.98, 95% CI 0.76-1.27, I2 = 85% )] respectively. Exposure to HCQ + AZM did not improve the VQR as well (RR = 3.23, 95% CI 0.70-14.97, I2 = 58%). The need for MV was not significantly different between the SC and HCQ (RR = 1.5, 95% CI 0.78-2.89, I2 = 81%), or HCQ + AZM (RR = 1.27, 95% CI 0.7-2.13, I2 = 88%). Side effects were more reported in the HCQ group than in the SC (RR = 3.14, 95% CI 1.58-6.24, I2 = 0). Radiological improvement and clinical worsening were not statistically different between HCQ and SC [(RR = 1.11, 95% CI 0.74-1.65, I2 = 45%) and (RR = 1.28, 95% CI 0.33-4.99), I2 = 54%] respectively. Despite the scarcity of published data of good quality, the effectiveness and safety of either HCQ alone or in combination with AZM in treating COVID-19 cannot be assured. Future high-quality RCTs need to be carried out.PROSPERO registration: CRD42020192084.
Collapse
Affiliation(s)
- Ramy Mohamed Ghazy
- Tropical Health Department, High Institute of Public Health, Alexandria University, Alexandria, Egypt
| | - Abdallah Almaghraby
- Department of Cardiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| | - Ramy Shaaban
- Department of Instructional Technology and Learning Sciences, Utah State University, Logan, USA
| | - Ahmed Kamal
- Hepatology Unit, Department of Internal Medicine, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Hatem Beshir
- Department of Cardiothoracic Surgery, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Cardiothoracic Surgery, Amreya General Hospital, Egyptian Ministry of Health and Population, Alexandria, Egypt
| | - Amr Moursi
- Department of Neurosurgery, NHS Tayside Trust, London, UK
| | - Ahmed Ramadan
- Department of Medical Information and Data Science, DataClin CRO, Cairo, Egypt
| | - Sarah Hamed N Taha
- Department of Forensic Medicine and Clinical Toxicology, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
230
|
Ghosh R, Chakraborty A, Biswas A, Chowdhuri S. Depicting the inhibitory potential of polyphenols from Isatis indigotica root against the main protease of SARS CoV-2 using computational approaches. J Biomol Struct Dyn 2020; 40:4110-4121. [PMID: 33292085 PMCID: PMC7738210 DOI: 10.1080/07391102.2020.1858164] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The pandemic disease COVID-19, caused by SARS CoV-2, has created a global crisis. Presently, researchers across the globe are in a quest to identify/develop drugs or vaccines by targeting different non-structural proteins (Nsps) of SARS CoV-2. One such important drug target is Nsp5/main protease (Mpro) which plays a critical role in the viral replication. This cysteine protease/Mpro of SARS CoV-2 has high sequence similarity with the same protease from SARS CoV-1. Previously, it has been shown experimentally that eight polyphenols derived from the root of Isatis indigotica show inhibitory effect on the cleavage/catalytic activity of the SARS CoV-1 Mpro. But whether these polyphenols exhibit any inhibitory effect on SARS CoV-2 Mpro is unclear. To explore this possibility, here, we have adopted various computational approaches. Polyphenols that qualified the pharmacological parameters (indigo, sinigrin, hesperetin and daidzein) and two well-known Mpro inhibitors (N3 and lopinavir) were subjected to molecular docking studies. Two of them (sinigrin and hesperetin) were selected by comparing their binding affinities with N3 and lopinavir. Sinigrin and hesperetin interacted with the two most important catalytic residues of Mpro (His41 and Cys145). Molecular dynamics studies further revealed that these two Mpro-polyphenol complexes are more stable and experience less conformational fluctuations than Mpro-N3/lopinavir complex. The Mpro-hesperetin complex was more compact and less expanded than Mpro-sinigrin complex. These findings were additionally validated by MM-GBSA analysis. As a whole, our study revealed that these two polyphenols may be potent SARS CoV-2 Mpro inhibitors and may possibly be considered for COVID-19 treatment.
Collapse
Affiliation(s)
- Rajesh Ghosh
- School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, India
| | - Ayon Chakraborty
- School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, India
| | - Ashis Biswas
- School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, India
| | - Snehasis Chowdhuri
- School of Basic Sciences, Indian Institute of Technology Bhubaneswar, Bhubaneswar, India
| |
Collapse
|
231
|
Haji-Maghsoudi S, Sadeghifar M, Roshanaei G, Mahjub H. The Impact of Control Measures and Holiday Seasons on Incidence and Mortality Rate of COVID-19 in Iran. J Res Health Sci 2020; 20:e00500. [PMID: 33424009 PMCID: PMC8695787 DOI: 10.34172/jrhs.2020.35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/15/2020] [Accepted: 11/30/2020] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Preventive measures on the COVID-19 pandemic is an effective way to control its spread. We aimed to investigate the effect of control measures and holiday seasons on the incidence and mortality rate of COVID-19 in Iran. STUDY DESIGN An observational study. METHODS The daily data of confirmed new cases and deaths in Iran were taken from the Johns Hopkins University COVID-19 database. We calculated weekly data from 19 Feb to 6 Oct 2020. To estimate the impact of control measures and holiday seasons on the incidence rate of new cases and deaths, an autoregressive hidden Markov model (ARHMM) with two hidden states fitted the data. The hidden states of the fitted model can distinguish the peak period from the non-peak period. RESULTS The control measures with a delay of one-week and two-week had a decreasing effect on the new cases in the peak and non-peak periods, respectively (P=0.005). The holiday season with a two-week delay increased the total number of new cases in the peak periods (P=0.031). The peak period for the occurrence of COVID-19 was estimated at 3 weeks. In the peak period of mortality, the control measures with a three-week delay decreased the COVID-19 mortality (P=0.010). The expected duration of staying in the peak period of mortality was around 6 weeks. CONCLUSION When an increasing trend was seen in the country, the control measures could decline the incidence and mortality related to COVID-19. Implementation of official restrictions on holiday seasons could prevent an upward trend of incidence for COVID-19 during the peak period.
Collapse
Affiliation(s)
- Saiedeh Haji-Maghsoudi
- Department of Biostatistics, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Majid Sadeghifar
- Department of Statistics, Faculty of Basic Sciences, Bu-Ali Sina University, Hamadan, Iran
| | - Ghodratollah Roshanaei
- Department of Biostatistics, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
- Modeling of Noncommunicable Diseases Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hossein Mahjub
- Department of Biostatistics, School of Public Health, Hamadan University of Medical Sciences, Hamadan, Iran
- Research Center for Health Sciences, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
232
|
Baum A, Ajithdoss D, Copin R, Zhou A, Lanza K, Negron N, Ni M, Wei Y, Mohammadi K, Musser B, Atwal GS, Oyejide A, Goez-Gazi Y, Dutton J, Clemmons E, Staples HM, Bartley C, Klaffke B, Alfson K, Gazi M, Gonzalez O, Dick E, Carrion R, Pessaint L, Porto M, Cook A, Brown R, Ali V, Greenhouse J, Taylor T, Andersen H, Lewis MG, Stahl N, Murphy AJ, Yancopoulos GD, Kyratsous CA. REGN-COV2 antibodies prevent and treat SARS-CoV-2 infection in rhesus macaques and hamsters. Science 2020; 370:1110-1115. [PMID: 33037066 PMCID: PMC7857396 DOI: 10.1126/science.abe2402] [Citation(s) in RCA: 415] [Impact Index Per Article: 83.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 10/07/2020] [Indexed: 01/06/2023]
Abstract
An urgent global quest for effective therapies to prevent and treat coronavirus disease 2019 (COVID-19) is ongoing. We previously described REGN-COV2, a cocktail of two potent neutralizing antibodies (REGN10987 and REGN10933) that targets nonoverlapping epitopes on the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike protein. In this report, we evaluate the in vivo efficacy of this antibody cocktail in both rhesus macaques, which may model mild disease, and golden hamsters, which may model more severe disease. We demonstrate that REGN-COV-2 can greatly reduce virus load in the lower and upper airways and decrease virus-induced pathological sequelae when administered prophylactically or therapeutically in rhesus macaques. Similarly, administration in hamsters limits weight loss and decreases lung titers and evidence of pneumonia in the lungs. Our results provide evidence of the therapeutic potential of this antibody cocktail.
Collapse
Affiliation(s)
- Alina Baum
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Richard Copin
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Anbo Zhou
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Kathryn Lanza
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Nicole Negron
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Min Ni
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | - Yi Wei
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | - Bret Musser
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | | | - Yenny Goez-Gazi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - John Dutton
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Elizabeth Clemmons
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Hilary M Staples
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Carmen Bartley
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Benjamin Klaffke
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Kendra Alfson
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Michal Gazi
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Olga Gonzalez
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Edward Dick
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | - Ricardo Carrion
- Southwest National Primate Research Center, Texas Biomedical Research Institute, San Antonio, TX 78245, USA
| | | | | | | | | | | | | | | | | | | | - Neil Stahl
- Regeneron Pharmaceuticals, Inc., Tarrytown, NY 10591, USA
| | | | | | | |
Collapse
|
233
|
Why Is the COVID-19 Curve on a Decline in Pakistan? Disaster Med Public Health Prep 2020; 16:849-850. [PMID: 33208217 PMCID: PMC7943944 DOI: 10.1017/dmp.2020.453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
234
|
Rahman MS, Islam MR, Alam ASMRU, Islam I, Hoque MN, Akter S, Rahaman MM, Sultana M, Hossain MA. Evolutionary dynamics of SARS-CoV-2 nucleocapsid protein and its consequences. J Med Virol 2020; 93:2177-2195. [PMID: 33095454 DOI: 10.1002/jmv.26626] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/17/2020] [Accepted: 10/20/2020] [Indexed: 12/21/2022]
Abstract
The emerged novel coronavirus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has created a global health crisis that warrants an accurate and detailed characterization of the rapidly evolving viral genome for understanding its epidemiology, pathogenesis, and containment. Here, we explored 61,485 sequences of the nucleocapsid (N) protein, a potent diagnostic and prophylactic target, for identifying the mutations to review their roles in real-time polymerase chain reaction based diagnosis and observe consequent impacts. Compared to the Wuhan reference strain, a total of 1034 unique nucleotide mutations were identified in the mutant strains (49.15%, n = 30,221) globally. Of these mutations, 367 occupy primer binding sites including the 3'-end mismatch to the primer-pair of 11 well-characterized primer sets. Noteworthily, CDC (USA) recommended the N2 primer set contained a lower mismatch than the other primer sets. Moreover, 684 amino acid (aa) substitutions were located across 317 (75.66% of total aa) unique positions including 82, 21, and 83 of those in the RNA binding N-terminal domain (NTD), SR-rich region, and C-terminal dimerization domain, respectively. Moreover, 11 in-frame deletions, mostly (n = 10) within the highly flexible linker region, were revealed, and the rest was within the NTD region. Furthermore, we predicted the possible consequence of high-frequency mutations (≥20) and deletions on the tertiary structure of the N protein. Remarkably, we observed that a high frequency (67.94% of mutated sequences) co-occuring mutations (R203K and G204R) destabilized and decreased overall structural flexibility. The N protein of SARS-CoV-2 comprises an average of 1.2 mutations per strain compared to 4.4 and 0.4 in Middle East respiratory syndrome-related coronavirus and SARS-CoV, respectively. Despite being proposed as the alternative target to spike protein for vaccine and therapeutics, the ongoing evolution of the N protein may challenge these endeavors, thus needing further immunoinformatics analyses. Therefore, continuous monitoring is required for tracing the ongoing evolution of the SARS-CoV-2 N protein in prophylactic and diagnostic interventions.
Collapse
Affiliation(s)
| | - M Rafiul Islam
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
| | - A S M Rubayet Ul Alam
- Department of Microbiology, Jashore University of Science and Technology, Jashore, Bangladesh
| | - Israt Islam
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
| | - M Nazmul Hoque
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh.,Department of Gynecology, Obstetrics and Reproductive Health, Bangabandhu Sheikh Mujibur Rahman Agricultural University, Gazipur, Bangladesh
| | - Salma Akter
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh.,Department of Microbiology, Jahangirnagar University, Savar, Dhaka, Bangladesh
| | | | - Munawar Sultana
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
| | - M Anwar Hossain
- Department of Microbiology, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
235
|
Huang Y, Lyu X, Li D, Wang L, Wang Y, Zou W, Wei Y, Wu X. A cohort study of 676 patients indicates D-dimer is a critical risk factor for the mortality of COVID-19. PLoS One 2020; 15:e0242045. [PMID: 33166991 PMCID: PMC7652321 DOI: 10.1371/journal.pone.0242045] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/26/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus Disease 2019 (COVID-19) has recently become a public emergency and a worldwide pandemic. However, the information on the risk factors associated with the mortality of COVID-19 and of their prognostic potential is limited. In this retrospective study, the clinical characteristics, treatment and outcome data were collected and analyzed from 676 COVID-19 patients stratified into 140 non-survivors and 536 survivors. We found that the levels of Dimerized plasmin fragment D (D-dimer), C-reactive protein (CRP), lactate dehydrogenase (LDH), procalcitonin (PCT) were significantly higher in non-survivals on admission (non-survivors vs. survivors: D-Dimer ≥ 0.5 mg/L, 83.2% vs. 44.9%, P<0.01; CRP ≥10 mg/L, 50.4% vs. 6.0%, P<0.01; LDH ≥ 250 U/L, 73.8% vs. 20.1%, P<0.01; PCT ≥ 0.5 ng/ml, 27.7% vs. 1.8%, P<0.01). Moreover, dynamic tracking showed D-dimer kept increasing in non-survivors, while CRP, LDH and PCT remained relatively stable after admission. D-dimer has the highest C-index to predict in-hospital mortality, and patients with D-dimer levels ≥0.5 mg/L had a higher incidence of mortality (Hazard Ratio: 4.39, P<0.01). Our study suggested D-dimer could be a potent marker to predict the mortality of COVID-19, which may be helpful for the management of patients.
Collapse
Affiliation(s)
- Yongsheng Huang
- School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyu Lyu
- Department of Endocrinology, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Li
- National Cancer Center/ Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Wang
- School of Basic Medicine, Peking Union Medical College, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Beijing, China
| | - Yujun Wang
- Department of Critical care medicine, the Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wenbin Zou
- Department of Thoracic Surgery, TongJi Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingxin Wei
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- * E-mail: (XW); (YW)
| | - Xiaowei Wu
- Department of Thoracic Surgery, TongJi Hospital, TongJi Medical College, Huazhong University of Science and Technology, Wuhan, China
- * E-mail: (XW); (YW)
| |
Collapse
|
236
|
Díaz FJ, Aguilar-Jiménez W, Flórez-Álvarez L, Valencia G, Laiton-Donato K, Franco-Muñoz C, Álvarez-Díaz D, Mercado-Reyes M, Rugeles MT. Isolation and characterization of an early SARS-CoV-2 isolate from the 2020 epidemic in Medellín, Colombia. BIOMEDICA : REVISTA DEL INSTITUTO NACIONAL DE SALUD 2020; 40:148-158. [PMID: 33152198 PMCID: PMC7676823 DOI: 10.7705/biomedica.5834] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 10/15/2020] [Indexed: 11/24/2022]
Abstract
Introduction: SARS-CoV-2 has been identified as the new coronavirus causing an outbreak of acute respiratory disease in China in December, 2019. This disease, currently named COVID-19, has been declared as a pandemic by the World Health Organization (WHO). The first case of COVID-19 in Colombia was reported on March 6, 2020. Here we characterize an early SARS-CoV-2 isolate from the pandemic recovered in April, 2020. Objective: To describe the isolation and characterization of an early SARS-CoV-2 isolate from the epidemic in Colombia. Materials and methods: A nasopharyngeal specimen from a COVID-19 positive patient was inoculated on different cell lines. To confirm the presence of SARS-CoV-2 on cultures we used qRT-PCR, indirect immunofluorescence assay, transmission and scanning electron microscopy, and next-generation sequencing. Results: We determined the isolation of SARS-CoV-2 in Vero-E6 cells by the appearance of the cytopathic effect three days post-infection and confirmed it by the positive results in the qRT-PCR and the immunofluorescence with convalescent serum. Transmission and scanning electron microscopy images obtained from infected cells showed the presence of structures compatible with SARS-CoV-2. Finally, a complete genome sequence obtained by next-generation sequencing allowed classifying the isolate as B.1.5 lineage. Conclusion: The evidence presented in this article confirms the first isolation of SARSCoV-2 in Colombia. In addition, it shows that this strain behaves in cell culture in a similar way to that reported in the literature for other isolates and that its genetic composition is consistent with the predominant variant in the world. Finally, points out the importance of viral isolation for the detection of neutralizing antibodies, for the genotypic and phenotypic characterization of the strain and for testing compounds with antiviral potential.
Collapse
Affiliation(s)
- Francisco J Díaz
- Grupo de Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.
| | - Wbeimar Aguilar-Jiménez
- Grupo de Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.
| | - Lizdany Flórez-Álvarez
- Grupo de Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.
| | - Gladys Valencia
- Ayudas Diagnósticas de Laboratorio Clínico, ADILAB, Medellín, Colombia.
| | | | - Carlos Franco-Muñoz
- Unidad de Secuenciación Genómica, Instituto Nacional de Salud, Bogotá, D.C., Colombia.
| | - Diego Álvarez-Díaz
- Unidad de Secuenciación Genómica, Instituto Nacional de Salud, Bogotá, D.C., Colombia.
| | - Marcela Mercado-Reyes
- Unidad de Secuenciación Genómica, Instituto Nacional de Salud, Bogotá, D.C., Colombia.
| | - María Teresa Rugeles
- Grupo de Inmunovirología, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.
| |
Collapse
|
237
|
Testing the Accuracy of the ARIMA Models in Forecasting the Spreading of COVID-19 and the Associated Mortality Rate. ACTA ACUST UNITED AC 2020; 56:medicina56110566. [PMID: 33121072 PMCID: PMC7694177 DOI: 10.3390/medicina56110566] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 01/15/2023]
Abstract
Background and objectives: The current pandemic of SARS-CoV-2 has not only changed, but also affected the lives of tens of millions of people around the world in these last nine to ten months. Although the situation is stable to some extent within the developed countries, approximately one million have already died as a consequence of the unique symptomatology that these people displayed. Thus, the need to develop an effective strategy for monitoring, restricting, but especially for predicting the evolution of COVID-19 is urgent, especially in middle-class countries such as Romania. Material and Methods: Therefore, autoregressive integrated moving average (ARIMA) models have been created, aiming to predict the epidemiological course of COVID-19 in Romania by using two statistical software (STATGRAPHICS Centurion (v.18.1.13) and IBM SPSS (v.20.0.0)). To increase the accuracy, we collected data between the established interval (1 March, 31 August) from the official website of the Romanian Government and the World Health Organization. Results: Several ARIMA models were generated from which ARIMA (1,2,1), ARIMA (3,2,2), ARIMA (3,1,3), ARIMA (3,2,2), ARIMA (3,1,3), ARIMA (2,2,2) and ARIMA (1,2,1) were considered the best models. For this, we took into account the lowest value of mean absolute percentage error (MAPE) for March, April, May, June, July, and August (MAPEMarch = 9.3225, MAPEApril = 0.975287, MAPEMay = 0.227675, MAPEJune = 0.161412, MAPEJuly = 0.243285, MAPEAugust = 0.163873, MAPEMarch – August = 2.29175 for STATGRAPHICS Centurion (v.18.1.13) and MAPEMarch = 57.505, MAPEApril = 1.152, MAPEMay = 0.259, MAPEJune = 0.185, MAPEJuly = 0.307, MAPEAugust = 0.194, and MAPEMarch – August = 6.013 for IBM SPSS (v.20.0.0) respectively. Conclusions: This study demonstrates that ARIMA is a useful statistical model for making predictions and provides an idea of the epidemiological status of the country of interest.
Collapse
|
238
|
Mohapatra PR, Mishra B. Upcoming SARS-CoV-2 vaccine: expectations and reality. Postgrad Med J 2020; 98:e75-e76. [PMID: 33087532 DOI: 10.1136/postgradmedj-2020-138821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/30/2020] [Indexed: 11/04/2022]
Affiliation(s)
- Prasanta R Mohapatra
- Pulmonary Medicine & Critical Care, All India Institute of Medical Sciences Bhubaneswar, Bhubaneswar, India
| | - Baijayantimala Mishra
- Microbiology, All India Institute of Medical Sciences - Bhubaneswar, Bhubaneswar, India
| |
Collapse
|
239
|
Xu Y, Xu Z, Liu X, Cai L, Zheng H, Huang Y, Zhou L, Huang L, Ling Y, Deng L, Li J, Chen S, Liu D, Lin Z, Zhou L, He W, Zhong N, Liu X, Li Y. Clinical Findings of COVID-19 Patients Admitted to Intensive Care Units in Guangdong Province, China: A Multicenter, Retrospective, Observational Study. Front Med (Lausanne) 2020; 7:576457. [PMID: 33195325 PMCID: PMC7604321 DOI: 10.3389/fmed.2020.576457] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/03/2020] [Indexed: 02/05/2023] Open
Abstract
Background: Information about critically ill patients with coronavirus disease 2019 (COVID-19) in China but outside of Wuhan is scarce. We aimed to describe the clinical features, treatment, and outcomes of patients with COVID-19 admitted to the intensive care unit (ICU) in Guangdong Province. Methods: In this multicenter, retrospective, observational study, we enrolled consecutive patients with COVID-19 who were admitted to seven ICUs in Guangdong Province. Demographic data, symptoms, laboratory findings, comorbidities, treatment, and outcomes were collected. Data were compared between patients with and without intubation. Results: A total of 45 COVID-19 patients required ICU admission in the study hospitals [mean age 56.7 ± 15.4 years, 29 males (64.4%)]. The most common symptoms at onset were fever and cough. Most patients presented with lymphopenia and elevated lactate dehydrogenase. Treatment with antiviral drugs was initiated in all patients. Thirty-six patients (80%) developed acute respiratory distress syndrome at ICU admission, and 15 (33.3%) septic shock. Twenty patients (44.4%) were intubated, and 10 (22.2%) received extracorporeal membrane oxygenation. The 60-day mortality was 4.4% (2 of 45). Conclusion: COVID-19 patients admitted to ICU were characterized by fever, lymphopenia, acute respiratory failure, and multiple organ dysfunction. The mortality of ICU patients in Guangdong Province was relatively low with a small sample size.
Collapse
Affiliation(s)
- Yonghao Xu
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhiheng Xu
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xuesong Liu
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lihua Cai
- Department of Critical Care Medicine, Dongguan People's Hospital, Dongguan, China
| | - Haichong Zheng
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yongbo Huang
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lixin Zhou
- Department of Critical Care Medicine, Foshan First People's Hospital, Foshan, China
| | - Linxi Huang
- Department of Critical Care Medicine, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Yun Ling
- Department of Critical Care Medicine, Huizhou Municipal Central Hospital, Huizhou, China
| | - Liehua Deng
- Department of Critical Care Medicine, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Jianwei Li
- Department of Critical Care Medicine, Zhongshan City People's Hospital, Zhongshan, China
| | - Sibei Chen
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Dongdong Liu
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zhimin Lin
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liang Zhou
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weiqun He
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Nanshan Zhong
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoqing Liu
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yimin Li
- State Key Laboratory of Respiratory Diseases, Department of Critical Care Medicine, Guangzhou Institute of Respiratory Health, First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
240
|
Berrio A, Gartner V, Wray GA. Positive selection within the genomes of SARS-CoV-2 and other Coronaviruses independent of impact on protein function. PeerJ 2020; 8:e10234. [PMID: 33088633 PMCID: PMC7571416 DOI: 10.7717/peerj.10234] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 10/04/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The emergence of a novel coronavirus (SARS-CoV-2) associated with severe acute respiratory disease (COVID-19) has prompted efforts to understand the genetic basis for its unique characteristics and its jump from non-primate hosts to humans. Tests for positive selection can identify apparently nonrandom patterns of mutation accumulation within genomes, highlighting regions where molecular function may have changed during the origin of a species. Several recent studies of the SARS-CoV-2 genome have identified signals of conservation and positive selection within the gene encoding Spike protein based on the ratio of synonymous to nonsynonymous substitution. Such tests cannot, however, detect changes in the function of RNA molecules. METHODS Here we apply a test for branch-specific oversubstitution of mutations within narrow windows of the genome without reference to the genetic code. RESULTS We recapitulate the finding that the gene encoding Spike protein has been a target of both purifying and positive selection. In addition, we find other likely targets of positive selection within the genome of SARS-CoV-2, specifically within the genes encoding Nsp4 and Nsp16. Homology-directed modeling indicates no change in either Nsp4 or Nsp16 protein structure relative to the most recent common ancestor. These SARS-CoV-2-specific mutations may affect molecular processes mediated by the positive or negative RNA molecules, including transcription, translation, RNA stability, and evasion of the host innate immune system. Our results highlight the importance of considering mutations in viral genomes not only from the perspective of their impact on protein structure, but also how they may impact other molecular processes critical to the viral life cycle.
Collapse
Affiliation(s)
| | - Valerie Gartner
- Department of Biology, Duke University, Durham, NC, USA
- University Program in Genetics and Genomics, Duke University, Durham, NC, USA
| | - Gregory A. Wray
- Department of Biology, Duke University, Durham, NC, USA
- Center for Genomic and Computational Biology, Duke University, Durham, NC, USA
| |
Collapse
|
241
|
Elizondo V, Harkins GW, Mabvakure B, Smidt S, Zappile P, Marier C, Maurano M, Perez V, Mazza N, Beloso C, Ifran S, Fernandez M, Santini A, Perez V, Estevez V, Nin M, Manrique G, Perez L, Ross F, Boschi S, Noel Zubillaga M, Balleste R, Dellicour S, Heguy A, Duerr R. SARS-CoV-2 genomic characterization and clinical manifestation of the COVID-19 outbreak in Uruguay. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2020:2020.10.08.20208546. [PMID: 33052352 PMCID: PMC7553156 DOI: 10.1101/2020.10.08.20208546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
COVID-19 is a respiratory illness caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and declared by the World Health Organization a global public health emergency. Among the severe outbreaks across South America, Uruguay has become known for curtailing SARS-CoV-2 exceptionally well. To understand the SARS-CoV-2 introductions, local transmissions, and associations with genomic and clinical parameters in Uruguay, we sequenced the viral genomes of 44 outpatients and inpatients in a private healthcare system in its capital, Montevideo, from March to May 2020. We performed a phylogeographic analysis using sequences from our cohort and other studies that indicate a minimum of 23 independent introductions into Uruguay, resulting in five major transmission clusters. Our data suggest that most introductions resulting in chains of transmission originate from other South American countries, with the earliest seeding of the virus in late February 2020, weeks before the borders were closed to all non-citizens and a partial lockdown implemented. Genetic analyses suggest a dominance of S and G clades (G, GH, GR) that make up >90% of the viral strains in our study. In our cohort, lethal outcome of SARS-CoV-2 infection significantly correlated with arterial hypertension, kidney failure, and ICU admission (FDR < 0.01), but not with any mutation in a structural or non-structural protein, such as the spike D614G mutation. Our study contributes genetic, phylodynamic, and clinical correlation data about the exceptionally well-curbed SARS-CoV-2 outbreak in Uruguay, which furthers the understanding of disease patterns and regional aspects of the pandemic in Latin America.
Collapse
Affiliation(s)
- Victoria Elizondo
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Gordon W. Harkins
- South African Medical Research Council Capacity Development Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| | - Batsirai Mabvakure
- Department of Medicine, Johns Hopkins School of Medicine, Baltimore, Maryland, United States of America
| | - Sabine Smidt
- South African Medical Research Council Capacity Development Unit, South African National Bioinformatics Institute, University of the Western Cape, Bellville, South Africa
| | - Paul Zappile
- Genome Technology Center, Office for Science and Research, NYU Langone Health, New York, New York, United States of America
| | - Christian Marier
- Genome Technology Center, Office for Science and Research, NYU Langone Health, New York, New York, United States of America
| | - Matthew Maurano
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, United States of America
| | - Victoria Perez
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
- Departamento de Desarrollo Biotecnológico, Instituto de Higiene, Facultad de Medicina, Udelar, Montevideo, Uruguay
| | - Natalia Mazza
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Carolina Beloso
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
- Departamento de Biodiversidad y Genética. Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Silvana Ifran
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Mariana Fernandez
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Andrea Santini
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Veronica Perez
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Veronica Estevez
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Matilde Nin
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Gonzalo Manrique
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Leticia Perez
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Fabiana Ross
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Susana Boschi
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Maria Noel Zubillaga
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Raquel Balleste
- Laboratorio de Biología Molecular, Asociación Española Primera en Salud, Montevideo, Uruguay
| | - Simon Dellicour
- Spatial Epidemiology Lab. (SpELL), Université Libre de Bruxelles, Bruxelles, Belgium
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Leuven, Belgium
| | - Adriana Heguy
- Genome Technology Center, Office for Science and Research, NYU Langone Health, New York, New York, United States of America
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, United States of America
| | - Ralf Duerr
- Department of Pathology, NYU Grossman School of Medicine, New York, New York, United States of America
| |
Collapse
|
242
|
Karamitros T, Papadopoulou G, Bousali M, Mexias A, Tsiodras S, Mentis A. SARS-CoV-2 exhibits intra-host genomic plasticity and low-frequency polymorphic quasispecies. J Clin Virol 2020; 131:104585. [PMID: 32818852 PMCID: PMC7418792 DOI: 10.1016/j.jcv.2020.104585] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 08/05/2020] [Accepted: 08/09/2020] [Indexed: 12/19/2022]
Abstract
In December 2019, an outbreak of atypical pneumonia (Coronavirus disease 2019 -COVID-19) associated with a novel coronavirus (SARS-CoV-2) was reported in Wuhan city, Hubei province, China. The outbreak was traced to a seafood wholesale market and human to human transmission was confirmed. The rapid spread and the death toll of the new epidemic warrants immediate intervention. The intra-host genomic variability of SARS-CoV-2 plays a pivotal role in the development of effective antiviral agents and vaccines, as well as in the design of accurate diagnostics. We analyzed NGS data derived from clinical samples of three Chinese patients infected with SARS-CoV-2, in order to identify small- and large-scale intra-host variations in the viral genome. We identified tens of low- or higher- frequency single nucleotide variations (SNVs) with variable density across the viral genome, affecting 7 out of 10 protein-coding viral genes. The majority of these SNVs (72/104) corresponded to missense changes. The annotation of the identified SNVs but also of all currently circulating strain variations revealed colocalization of intra-host as well as strain specific SNVs with primers and probes currently used in molecular diagnostics assays. Moreover, we de-novo assembled the viral genome, in order to isolate and validate intra-host structural variations and recombination breakpoints. The bioinformatics analysis disclosed genomic rearrangements over poly-A / poly-U regions located in ORF1ab and spike (S) gene, including a potential recombination hot-spot within S gene. Our results highlight the intra-host genomic diversity and plasticity of SARS-CoV-2, pointing out genomic regions that are prone to alterations. The isolated SNVs and genomic rearrangements reflect the intra-patient capacity of the polymorphic quasispecies, which may arise rapidly during the outbreak, allowing immunological escape of the virus, offering resistance to anti-viral drugs and affecting the sensitivity of the molecular diagnostics assays.
Collapse
Affiliation(s)
- Timokratis Karamitros
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece.
| | - Gethsimani Papadopoulou
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Maria Bousali
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Anastasios Mexias
- Bioinformatics and Applied Genomics Unit, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| | - Sotirios Tsiodras
- 4(th) Academic Department of Internal Medicine, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Andreas Mentis
- Public Health Laboratories, Department of Microbiology, Hellenic Pasteur Institute, Athens, Greece
| |
Collapse
|
243
|
Li S, Li S, Disoma C, Zheng R, Zhou M, Razzaq A, Liu P, Zhou Y, Dong Z, Du A, Peng J, Hu L, Huang J, Feng P, Jiang T, Xia Z. SARS‐CoV‐2: Mechanism of infection and emerging technologies for future prospects. Rev Med Virol 2020; 31:e2168. [DOI: 10.1002/rmv.2168] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 08/23/2020] [Accepted: 08/30/2020] [Indexed: 01/08/2023]
Affiliation(s)
- Shiqin Li
- Department of Cell Biology School of Life Sciences Central South University Changsha China
| | - Sijia Li
- Department of Cell Biology School of Life Sciences Central South University Changsha China
| | - Cyrollah Disoma
- Department of Cell Biology School of Life Sciences Central South University Changsha China
| | - Rong Zheng
- Department of Cell Biology School of Life Sciences Central South University Changsha China
| | - Mei Zhou
- Department of Cell Biology School of Life Sciences Central South University Changsha China
| | - Aroona Razzaq
- Department of Cell Biology School of Life Sciences Central South University Changsha China
| | - Pinjia Liu
- Department of Cell Biology School of Life Sciences Central South University Changsha China
| | - Yuzheng Zhou
- Department of Cell Biology School of Life Sciences Central South University Changsha China
- Section of Infection and Immunity Herman Ostrow School of Dentistry University of Southern California Los Angeles California USA
| | - Zijun Dong
- Department of Cell Biology School of Life Sciences Central South University Changsha China
| | - Ashuai Du
- Department of Cell Biology School of Life Sciences Central South University Changsha China
| | - Jian Peng
- Department of General Surgery Xiangya Hospital Central South University Changsha China
| | - Liqiang Hu
- The First Hospital of Changsha University of South China Changsha China
| | - Jufang Huang
- Department of Anatomy and Neurobiology School of Basic Medical Sciences Central South University Changsha China
- School of Life Sciences Central South University Changsha China
| | - Pinghui Feng
- Section of Infection and Immunity Herman Ostrow School of Dentistry University of Southern California Los Angeles California USA
| | - Taijiao Jiang
- Center for Systems Medicine Institute of Basic Medical Sciences Chinese Academy of Medical Sciences & Peking Union Medical College Beijing China
- Suzhou Institute of Systems Medicine Suzhou Jiangsu China
| | - Zanxian Xia
- Department of Cell Biology School of Life Sciences Central South University Changsha China
- Hunan Key Laboratory of Medical Genetics & Center for Medical Genetics School of Life Sciences Hunan Key Laboratory of Animal Models for Human Diseases Central South University Changsha China
| |
Collapse
|
244
|
Pehote G, Vij N. Autophagy Augmentation to Alleviate Immune Response Dysfunction, and Resolve Respiratory and COVID-19 Exacerbations. Cells 2020; 9:cells9091952. [PMID: 32847034 PMCID: PMC7565665 DOI: 10.3390/cells9091952] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
The preservation of cellular homeostasis requires the synthesis of new proteins (proteostasis) and organelles, and the effective removal of misfolded or impaired proteins and cellular debris. This cellular homeostasis involves two key proteostasis mechanisms, the ubiquitin proteasome system and the autophagy–lysosome pathway. These catabolic pathways have been known to be involved in respiratory exacerbations and the pathogenesis of various lung diseases, such as chronic obstructive pulmonary disease (COPD), cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), acute lung injury (ALI), acute respiratory distress syndrome (ARDS), and coronavirus disease-2019 (COVID-19). Briefly, proteostasis and autophagy processes are known to decline over time with age, cigarette or biomass smoke exposure, and/or influenced by underlying genetic factors, resulting in the accumulation of misfolded proteins and cellular debris, elevating apoptosis and cellular senescence, and initiating the pathogenesis of acute or chronic lung disease. Moreover, autophagic dysfunction results in an impaired microbial clearance, post-bacterial and/or viral infection(s) which contribute to the initiation of acute and recurrent respiratory exacerbations as well as the progression of chronic obstructive and restrictive lung diseases. In addition, the autophagic dysfunction-mediated cystic fibrosis transmembrane conductance regulator (CFTR) immune response impairment further exacerbates the lung disease. Recent studies demonstrate the therapeutic potential of novel autophagy augmentation strategies, in alleviating the pathogenesis of chronic obstructive or restrictive lung diseases and exacerbations such as those commonly seen in COPD, CF, ALI/ARDS and COVID-19.
Collapse
Affiliation(s)
- Garrett Pehote
- Michigan State University College of Osteopathic Medicine, East Lansing, MI 48823, USA;
| | - Neeraj Vij
- Department of Pediatrics and Pulmonary Medicine, the Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- PRECISION THERANOSTICS INC, Baltimore, MD 21202, USA
- VIJ BIOTECH, Baltimore, MD 21202, USA
- Correspondence: or ; Tel.: +1-240-623-0757
| |
Collapse
|
245
|
Isabel S, Graña-Miraglia L, Gutierrez JM, Bundalovic-Torma C, Groves HE, Isabel MR, Eshaghi A, Patel SN, Gubbay JB, Poutanen T, Guttman DS, Poutanen SM. Evolutionary and structural analyses of SARS-CoV-2 D614G spike protein mutation now documented worldwide. Sci Rep 2020; 10:14031. [PMID: 32820179 PMCID: PMC7441380 DOI: 10.1038/s41598-020-70827-z] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/30/2020] [Indexed: 11/08/2022] Open
Abstract
The COVID-19 pandemic, caused by the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), was declared on March 11, 2020 by the World Health Organization. As of the 31st of May, 2020, there have been more than 6 million COVID-19 cases diagnosed worldwide and over 370,000 deaths, according to Johns Hopkins. Thousands of SARS-CoV-2 strains have been sequenced to date, providing a valuable opportunity to investigate the evolution of the virus on a global scale. We performed a phylogenetic analysis of over 1,225 SARS-CoV-2 genomes spanning from late December 2019 to mid-March 2020. We identified a missense mutation, D614G, in the spike protein of SARS-CoV-2, which has emerged as a predominant clade in Europe (954 of 1,449 (66%) sequences) and is spreading worldwide (1,237 of 2,795 (44%) sequences). Molecular dating analysis estimated the emergence of this clade around mid-to-late January (10-25 January) 2020. We also applied structural bioinformatics to assess the potential impact of D614G on the virulence and epidemiology of SARS-CoV-2. In silico analyses on the spike protein structure suggests that the mutation is most likely neutral to protein function as it relates to its interaction with the human ACE2 receptor. The lack of clinical metadata available prevented our investigation of association between viral clade and disease severity phenotype. Future work that can leverage clinical outcome data with both viral and human genomic diversity is needed to monitor the pandemic.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Angiotensin-Converting Enzyme 2
- Base Sequence
- Betacoronavirus/chemistry
- Betacoronavirus/pathogenicity
- COVID-19
- Child
- Child, Preschool
- Computer Simulation
- Coronavirus Infections/epidemiology
- Coronavirus Infections/virology
- Evolution, Molecular
- Female
- Genome, Viral/genetics
- Humans
- Infant
- Male
- Middle Aged
- Mutation, Missense
- Pandemics
- Peptidyl-Dipeptidase A/metabolism
- Phylogeny
- Pneumonia, Viral/epidemiology
- Pneumonia, Viral/virology
- Protein Conformation
- SARS-CoV-2
- Spike Glycoprotein, Coronavirus/chemistry
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/metabolism
- Virulence/genetics
- Young Adult
Collapse
Affiliation(s)
- Sandra Isabel
- Infectious Diseases Division, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada.
| | - Lucía Graña-Miraglia
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | | | - Cedoljub Bundalovic-Torma
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Helen E Groves
- Infectious Diseases Division, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
| | - Marc R Isabel
- Département de mathématique, Université Laval, Québec City, Canada
| | | | - Samir N Patel
- Public Health Ontario, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Jonathan B Gubbay
- Infectious Diseases Division, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON, Canada
- Public Health Ontario, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | | | - David S Guttman
- Centre for the Analysis of Genome Evolution and Function, University of Toronto, Toronto, ON, Canada
- Department of Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Susan M Poutanen
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
- Department of Microbiology, University Health System/Sinai Health, Toronto, ON, Canada
| |
Collapse
|