1
|
Abbasian MH, Rahimian K, Mahmanzar M, Bayat S, Kuehu DL, Sisakht MM, Moradi B, Deng Y. Comparative Atlas of SARS-CoV-2 Substitution Mutations: A Focus on Iranian Strains Amidst Global Trends. Viruses 2024; 16:1331. [PMID: 39205305 PMCID: PMC11359407 DOI: 10.3390/v16081331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 08/12/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a new emerging coronavirus that caused coronavirus disease 2019 (COVID-19). Whole-genome tracking of SARS-CoV-2 enhanced our understanding of the mechanism of the disease, control, and prevention of COVID-19. METHODS we analyzed 3368 SARS-CoV-2 protein sequences from Iran and compared them with 15.6 million global sequences in the GISAID database, using the Wuhan-Hu-1 strain as a reference. RESULTS Our investigation revealed that NSP12-P323L, ORF9c-G50N, NSP14-I42V, membrane-A63T, Q19E, and NSP3-G489S were found to be the most frequent mutations among Iranian SARS-CoV-2 sequences. Furthermore, it was observed that more than 94% of the SARS-CoV-2 genome, including NSP7, NSP8, NSP9, NSP10, NSP11, and ORF8, had no mutations when compared to the Wuhan-Hu-1 strain. Finally, our data indicated that the ORF3a-T24I, NSP3-G489S, NSP5-P132H, NSP14-I42V, envelope-T9I, nucleocapsid-D3L, membrane-Q19E, and membrane-A63T mutations might be responsible factors for the surge in the SARS-CoV-2 Omicron variant wave in Iran. CONCLUSIONS real-time genomic surveillance is crucial for detecting new SARS-CoV-2 variants, updating diagnostic tools, designing vaccines, and understanding adaptation to new environments.
Collapse
Affiliation(s)
- Mohammad Hadi Abbasian
- Department of Medical Genetics, National Institute for Genetic Engineering and Biotechnology, Tehran 1497716316, Iran;
| | - Karim Rahimian
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran 14174, Iran;
| | - Mohammadamin Mahmanzar
- Department of Bioinformatics, Kish International Campus University of Tehran, Kish 7941639982, Iran;
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA;
| | - Saleha Bayat
- Department of Biology & Research Center for Animal Development Applied Biology, Mashhad Branch, Islamic Azad University, Mashhad 9187147578, Iran;
| | - Donna Lee Kuehu
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA;
| | - Mahsa Mollapour Sisakht
- Faculty of Pharmacy, Biotechnology Research Center, Tehran University of Medical Sciences, Tehran 1936893813, Iran;
| | - Bahman Moradi
- Department of Biology, Faculty of Sciences, Shahid Bahonar University of Kerman, Kerman 7616913439, Iran;
| | - Youping Deng
- Department of Quantitative Health Sciences, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, HI 96813, USA;
| |
Collapse
|
2
|
Valerio TL, Anastácio R, da Silva SS, de Oliveira CC, Vidotti M. An overview of electrochemical biosensors used for COVID-19 detection. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:2164-2176. [PMID: 38536084 DOI: 10.1039/d3ay02042h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
This short review presents the latest advances in the field of electrochemical biosensors, focusing particularly on impedimetric biosensors for the direct measurement of analytes. As a source of study we have chosen to describe these advances in the latest global health crisis originated from the COVID-19 pandemic, initiated by the SARS-CoV-2 virus. In this period, the necessity for swift and precise detection methods has grown rapidly due to an imminent need for the development of an analytical method to identify and isolate infected patients as an attempt to control the spreading of the disease. Traditional approaches such as the enzyme-linked immunosorbent assay (ELISA), were extensively used during the SARS-CoV-2 pandemic, but their drawbacks, including slow response time, became evident. In this context, the potential of electrochemical biosensors as an alternative for COVID-19 detection was emphasized. These biosensors merge electrochemical technology with bioreceptors, offering benefits such as rapidity, accuracy, portability, and real-time result provision. Additionally, we present instances of electrochemical biosensors modified with conductive polymers, eliminating the necessity for an electrochemical probe. The adaptability of the developed materials and devices facilitated the prompt production of electrochemical biosensors during the pandemic, creating opportunities for broader applications in infectious disease diagnosis.
Collapse
Affiliation(s)
- Tatiana Lima Valerio
- Grupo de Pesquisa em Macromoléculas e Interfaces, Universidade Federal Do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil.
| | - Raquel Anastácio
- Grupo de Pesquisa em Macromoléculas e Interfaces, Universidade Federal Do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil.
| | - Stella Schuster da Silva
- Laboratório de Células Inflamatórias e Neoplásicas (LCIN) e Laboratório de Investigação de Polissacarídeos Sulfatados (LIPS), Universidade Federal Do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil
| | - Carolina Camargo de Oliveira
- Laboratório de Células Inflamatórias e Neoplásicas (LCIN) e Laboratório de Investigação de Polissacarídeos Sulfatados (LIPS), Universidade Federal Do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil
| | - Marcio Vidotti
- Grupo de Pesquisa em Macromoléculas e Interfaces, Universidade Federal Do Paraná (UFPR), 81531-980, Curitiba, PR, Brazil.
| |
Collapse
|
3
|
Sarkar A, Ghosh TA, Bandyopadhyay B, Maiti S, Panja AS. Prediction of Prospective Mutational Landscape of SARS-CoV-2 Spike ssRNA and Evolutionary Basis of Its Host Interaction. Mol Biotechnol 2024:10.1007/s12033-024-01146-1. [PMID: 38619800 DOI: 10.1007/s12033-024-01146-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 03/14/2024] [Indexed: 04/16/2024]
Abstract
Booster doses are crucial against severe COVID-19, as rapid virus mutations and variant emergence prolong the pandemic crisis. The virus's quick evolution, short generation-time, and adaptive changes impact virulence and evolvability, helping predictions about variant of concerns' (VOCs') landscapes. Here, in this study, we used a new computational algorithm, to predict the mutational pattern in SARS-CoV-2 ssRNA, proteomics, structural identification, mutation stability, and functional correlation, as well as immune escape mechanisms. Interestingly, the sequence diversity of SARS Coronavirus-2 has demonstrated a predominance of G- > A and C- > U substitutions. The best validation statistics are explored here in seven homologous models of the expected mutant SARS-CoV-2 spike ssRNA and employed for hACE2 and IgG interactions. The interactome profile of SARS-CoV-2 spike with hACE2 and IgG revealed a strong correlation between phylogeny and divergence time. The systematic adaptation of SARS-CoV-2 spike ssRNA influences infectivity and immune escape. Data suggest higher propensity of Adenine rich sequence promotes MHC system avoidance, preferred by A-rich codons. Phylogenetic data revealed the evolution of SARS-CoV-2 lineages' epidemiology. Our findings may unveil processes governing the genesis of immune-resistant variants, prompting a critical reassessment of the coronavirus mutation rate and exploration of hypotheses beyond mechanical aspects.
Collapse
Affiliation(s)
- Aniket Sarkar
- Post Graduate Department of Biotechnology, Oriental Institute of Science and Technology, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Trijit Arka Ghosh
- Department of Computer Application, Burdwan Institute of Management and Computer Science, The University of Burdwan, Dewandighi, Burdwan, West Bengal, 713102, India
| | - Bidyut Bandyopadhyay
- Post Graduate Department of Biotechnology, Oriental Institute of Science and Technology, Vidyasagar University, Midnapore, West Bengal, 721102, India
| | - Smarajit Maiti
- Department of Medical Laboratory Technology, Haldia Institute of Health Sciences, ICARE Complex, Haldia, West Bengal, 721657, India
| | - Anindya Sundar Panja
- Post Graduate Department of Biotechnology, Molecular Informatics Laboratory, Oriental Institute of Science and Technology, Vidyasagar University, Midnapore, West Bengal, 721102, India.
| |
Collapse
|
4
|
Rak A, Isakova-Sivak I, Rudenko L. Overview of Nucleocapsid-Targeting Vaccines against COVID-19. Vaccines (Basel) 2023; 11:1810. [PMID: 38140214 PMCID: PMC10747980 DOI: 10.3390/vaccines11121810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
The new SARS-CoV-2 coronavirus, which emerged in late 2019, is a highly variable causative agent of COVID-19, a contagious respiratory disease with potentially severe complications. Vaccination is considered the most effective measure to prevent the spread and complications of this infection. Spike (S) protein-based vaccines were very successful in preventing COVID-19 caused by the ancestral SARS-CoV-2 strain; however, their efficacy was significantly reduced when coronavirus variants antigenically different from the original strain emerged in circulation. This is due to the high variability of this major viral antigen caused by escape from the immunity caused by the infection or vaccination with spike-targeting vaccines. The nucleocapsid protein (N) is a much more conserved SARS-CoV-2 antigen than the spike protein and has therefore attracted the attention of scientists as a promising target for broad-spectrum vaccine development. Here, we summarized the current data on various N-based COVID-19 vaccines that have been tested in animal challenge models or clinical trials. Despite the high conservatism of the N protein, escape mutations gradually occurring in the N sequence can affect its protective properties. During the three years of the pandemic, at least 12 mutations have arisen in the N sequence, affecting more than 40 known immunogenic T-cell epitopes, so the antigenicity of the N protein of recent SARS-CoV-2 variants may be altered. This fact should be taken into account as a limitation in the development of cross-reactive vaccines based on N-protein.
Collapse
Affiliation(s)
- Alexandra Rak
- Department of Virology, Institute of Experimental Medicine, St. Petersburg 197022, Russia; (I.I.-S.); (L.R.)
| | | | | |
Collapse
|
5
|
Arya R, Tripathi P, Nayak K, Ganesh J, Bihani SC, Ghosh B, Prashar V, Kumar M. Insights into the evolution of mutations in SARS-CoV-2 non-spike proteins. Microb Pathog 2023; 185:106460. [PMID: 37995880 DOI: 10.1016/j.micpath.2023.106460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 10/16/2023] [Accepted: 11/17/2023] [Indexed: 11/25/2023]
Abstract
The COVID-19 pandemic has been driven by the emergence of SARS-CoV-2 variants with mutations across all the viral proteins. Although mutations in the spike protein have received significant attention, understanding the prevalence and potential impact of mutations in other viral proteins is essential for comprehending the evolution of SARS-CoV-2. Here, we conducted a comprehensive analysis of approximately 14 million sequences of SARS-CoV-2 deposited in the GISAID database until December 2022 to identify prevalent mutations in the non-spike proteins at the global and country levels. Additionally, we evaluated the energetics of each mutation to better understand their impact on protein stability. While the consequences of many mutations remain unclear, we discuss potential structural and functional significance of some mutations. Our study highlights the ongoing evolutionary process of SARS-CoV-2 and underscores the importance of understanding changes in non-spike proteins.
Collapse
Affiliation(s)
- Rimanshee Arya
- Protein Crystallography Section, Bhabha Atomic Research Centre, Mumbai, 400085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Preeti Tripathi
- Protein Crystallography Section, Bhabha Atomic Research Centre, Mumbai, 400085, India
| | - Karthik Nayak
- Protein Crystallography Section, Bhabha Atomic Research Centre, Mumbai, 400085, India; School of Chemical Sciences, UM-DAE Centre for Excellence in Basic Sciences, University of Mumbai, Vidyanagari, Mumbai, 400098, India
| | - Janani Ganesh
- Protein Crystallography Section, Bhabha Atomic Research Centre, Mumbai, 400085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Subhash C Bihani
- Protein Crystallography Section, Bhabha Atomic Research Centre, Mumbai, 400085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Biplab Ghosh
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India; Beamline Development & Application Section, Bhabha Atomic Research Centre, Mumbai, 400085, India
| | - Vishal Prashar
- Protein Crystallography Section, Bhabha Atomic Research Centre, Mumbai, 400085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India.
| | - Mukesh Kumar
- Protein Crystallography Section, Bhabha Atomic Research Centre, Mumbai, 400085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India.
| |
Collapse
|
6
|
Liu L, Ross NM, Handorf EA, Meeker CR, Chen G, Baldwin D, Vijayvergia N. Incidence of asymptomatic COVID-19 positivity in cancer patients and effects on therapy. J Cancer Res Clin Oncol 2023; 149:3243-3247. [PMID: 35904602 PMCID: PMC9334983 DOI: 10.1007/s00432-022-04231-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 07/19/2022] [Indexed: 11/24/2022]
Abstract
PURPOSE The coronavirus disease 2019 (COVID-19) pandemic is posing unprecedented challenges for patient care, especially for cancer patients. This study looks at asymptomatic (AS) COVID-19 positivity in cancer patients and its effects on their care. METHODS We conducted a retrospective chart review of AS patients testing positive for COVID-19 upon screening at Fox Chase Cancer Center between January 2020 and September 2020. Relationships between positive tests and demographics, clinical characteristics, and treatment delays were investigated using conditional logistic regression or Mantel-Haenszel tests. RESULTS Among 4143 AS patients who underwent COVID-19 testing, 25 (0.6%) were COVID-19 positive (cases) and these were matched to 50 controls. The median age was lower in the cases compared to that of the controls (64 vs 70 years old, p = 0.04). Of the cases, 10 patients (40%) never underwent their planned oncologic intervention [6/10 (60%) did not require the planned intervention once deemed okay to proceed]. Of the controls, only 1 patient (2%) did not undergo the planned intervention. Of these 15 COVID-19 positive patients who underwent the planned intervention, 11 (73.3%) had a delay related to COVID-19, with a mean delay duration of 18 days (range: 0-49, SD: 16.72). CONCLUSION Cancer patients had lower incidence of AS COVID-19 than general population. Delays that occur due to AS COVID screening are not very long and serve as a tool to limit spread of virus. Further studies will be important in addressing delays in cancer care and concerns of patient safety as the pandemic continues.
Collapse
Affiliation(s)
- Lisa Liu
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19128, USA
| | - Nicole M Ross
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19128, USA
| | - Elizabeth A Handorf
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19128, USA
| | - Caitlin R Meeker
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19128, USA
| | - Giana Chen
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19128, USA
| | - Donald Baldwin
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19128, USA
| | - Namrata Vijayvergia
- Department of Medical Oncology, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA, 19128, USA.
| |
Collapse
|
7
|
Bills C, Xie X, Shi PY. The multiple roles of nsp6 in the molecular pathogenesis of SARS-CoV-2. Antiviral Res 2023; 213:105590. [PMID: 37003304 PMCID: PMC10063458 DOI: 10.1016/j.antiviral.2023.105590] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/19/2023] [Accepted: 03/27/2023] [Indexed: 04/03/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to evolve and adapt after its emergence in late 2019. As the causative agent of the coronavirus disease 2019 (COVID-19), the replication and pathogenesis of SARS-CoV-2 have been extensively studied by the research community for vaccine and therapeutics development. Given the importance of viral spike protein in viral infection/transmission and vaccine development, the scientific community has thus far primarily focused on studying the structure, function, and evolution of the spike protein. Other viral proteins are understudied. To fill in this knowledge gap, a few recent studies have identified nonstructural protein 6 (nsp6) as a major contributor to SARS-CoV-2 replication through the formation of replication organelles, antagonism of interferon type I (IFN-I) responses, and NLRP3 inflammasome activation (a major factor of severe disease in COVID-19 patients). Here, we review the most recent progress on the multiple roles of nsp6 in modulating SARS-CoV-2 replication and pathogenesis.
Collapse
Affiliation(s)
- Cody Bills
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Xuping Xie
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Pei-Yong Shi
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, Texas, USA; Institute for Human Infection and Immunity, University of Texas Medical Branch, Galveston, Texas, USA; World Reference Center of Emerging Viruses and Arboviruses, University of Texas Medical Branch, Galveston, Texas, USA; Center for Biodefense and Emerging Infectious Diseases, University of Texas Medical Branch, Galveston, Texas, USA; Sealy Institute for Drug Discovery, University of Texas Medical Branch, Galveston, Texas, USA.
| |
Collapse
|
8
|
Yi D, Chen X, Wang H, Song Q, Zhang L, Li P, Ye W, Chen J, Li F, Yi D, Wu Y. COVID-19 epidemic and public health interventions in Shanghai, China: Statistical analysis of transmission, correlation and conversion. Front Public Health 2023; 10:1076248. [PMID: 36703835 PMCID: PMC9871588 DOI: 10.3389/fpubh.2022.1076248] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 12/16/2022] [Indexed: 01/12/2023] Open
Abstract
Background The Shanghai COVID-19 epidemic is an important example of a local outbreak and of the implementation of normalized prevention and disease control strategies. The precise impact of public health interventions on epidemic prevention and control is unknown. Methods We collected information on COVID-19 patients reported in Shanghai, China, from January 30 to May 31, 2022. These newly added cases were classified as local confirmed cases, local asymptomatic infections, imported confirmed cases and imported asymptomatic infections. We used polynomial fitting correlation analysis and illustrated the time lag plot in the correlation analysis of local and imported cases. Analyzing the conversion of asymptomatic infections to confirmed cases, we proposed a new measure of the conversion rate (C r ). In the evolution of epidemic transmission and the analysis of intervention effects, we calculated the effective reproduction number (R t ). Additionally, we used simulated predictions of public health interventions in transmission, correlation, and conversion analyses. Results (1) The overall level of R t in the first three stages was higher than the epidemic threshold. After the implementation of public health intervention measures in the third stage, R t decreased rapidly, and the overall R t level in the last three stages was lower than the epidemic threshold. The longer the public health interventions were delayed, the more cases that were expected and the later the epidemic was expected to end. (2) In the correlation analysis, the outbreak in Shanghai was characterized by double peaks. (3) In the conversion analysis, when the incubation period was short (3 or 7 days), the conversion rate fluctuated smoothly and did not reflect the effect of the intervention. When the incubation period was extended (10 and 14 days), the conversion rate fluctuated in each period, being higher in the first five stages and lower in the sixth stage. Conclusion Effective public health interventions helped slow the spread of COVID-19 in Shanghai, shorten the outbreak duration, and protect the healthcare system from stress. Our research can serve as a positive guideline for addressing infectious disease prevention and control in China and other countries and regions.
Collapse
Affiliation(s)
- Dali Yi
- Department of Health Statistics, College of Preventive Medicine, Army Medical University, Chongqing, China,Department of Health Education, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Xicheng Chen
- Department of Health Statistics, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Haojia Wang
- Department of Health Statistics, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Qiuyue Song
- Department of Health Statistics, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Ling Zhang
- Department of Health Education, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Pengpeng Li
- Department of Health Statistics, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Wei Ye
- Department of Health Statistics, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Jia Chen
- Department of Health Statistics, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Fang Li
- Department of Health Statistics, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Dong Yi
- Department of Health Statistics, College of Preventive Medicine, Army Medical University, Chongqing, China
| | - Yazhou Wu
- Department of Health Statistics, College of Preventive Medicine, Army Medical University, Chongqing, China,*Correspondence: Yazhou Wu ✉
| |
Collapse
|
9
|
Zeng Z, Wu T, Lin Z, Luo L, Lin Z, Guan W, Liang J, Yu M, Guan P, He W, Liu Z, Lu G, Xie P, Chen C, Lau EHY, Yang Z, Hon C, He J. Containment of SARS-CoV-2 Delta strain in Guangzhou, China by quarantine and social distancing: a modelling study. Sci Rep 2022; 12:21096. [PMID: 36473881 PMCID: PMC9727161 DOI: 10.1038/s41598-022-21674-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 09/29/2022] [Indexed: 12/12/2022] Open
Abstract
China detected the first case of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection with Delta variant in May 2021. We assessed control strategies against this variant of concern. We constructed a robust transmission model to assess the effectiveness of interventions against the Delta variant in Guangzhou with initial quarantine/isolation, followed by social distancing. We also assessed the effectiveness of alternative strategies and that against potentially more infectious variants. The effective reproduction number (Rt) fell below 1 when the average daily number of close contacts was reduced to ≤ 7 and quarantine/isolation was implemented on average at the same day of symptom onset in Guangzhou. Simulations showed that the outbreak could still be contained when quarantine is implemented on average 1 day after symptom onset while the average daily number of close contacts was reduced to ≤ 9 per person one week after the outbreak's beginning. Early quarantine and reduction of close contacts were found to be important for containment of the outbreaks. Early implementation of quarantine/isolation along with social distancing measures could effectively suppress spread of the Delta and more infectious variants.
Collapse
Affiliation(s)
- Zhiqi Zeng
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120 People’s Republic of China ,grid.410737.60000 0000 8653 1072Guangzhou key laboratory for clinical rapid diagnosis and early warning of infectious diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, People’s Republic of China
| | - Tong Wu
- grid.259384.10000 0000 8945 4455Macao Institute of Systems Engineering, Macao University of Science and Technology, Macau SAR, People’s Republic of China ,Singou Technology (Macau) Ltd, Macau SAR, People’s Republic of China
| | - Zhijie Lin
- grid.259384.10000 0000 8945 4455Macao Institute of Systems Engineering, Macao University of Science and Technology, Macau SAR, People’s Republic of China
| | - Lei Luo
- grid.508371.80000 0004 1774 3337Guangzhou Center for Disease Control and Prevention, Guangzhou, Guangdong People’s Republic of China
| | - Zhengshi Lin
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120 People’s Republic of China
| | - Wenda Guan
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120 People’s Republic of China
| | - Jingyi Liang
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120 People’s Republic of China
| | - Minfei Yu
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120 People’s Republic of China
| | - Peikun Guan
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120 People’s Republic of China
| | - Wei He
- grid.259384.10000 0000 8945 4455Macao Institute of Systems Engineering, Macao University of Science and Technology, Macau SAR, People’s Republic of China
| | - Zige Liu
- grid.259384.10000 0000 8945 4455Macao Institute of Systems Engineering, Macao University of Science and Technology, Macau SAR, People’s Republic of China
| | - Guibin Lu
- grid.259384.10000 0000 8945 4455Macao Institute of Systems Engineering, Macao University of Science and Technology, Macau SAR, People’s Republic of China
| | - Peifang Xie
- grid.218292.20000 0000 8571 108XFaculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500 People’s Republic of China
| | - Canxiong Chen
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120 People’s Republic of China
| | - Eric H. Y. Lau
- grid.194645.b0000000121742757School of Public Health, Li Ka Shing, Faculty of Medicine, The University of Hong Kong, 7 Sassoon Road, Pokfulam, Hong Kong People’s Republic of China ,Laboratory of Data Discovery for Health, Tai Po, Hong Kong People’s Republic of China
| | - Zifeng Yang
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120 People’s Republic of China ,grid.410737.60000 0000 8653 1072Guangzhou key laboratory for clinical rapid diagnosis and early warning of infectious diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, People’s Republic of China ,Guangzhou Laboratory, Guangzhou, People’s Republic of China
| | - Chitin Hon
- grid.259384.10000 0000 8945 4455Macao Institute of Systems Engineering, Macao University of Science and Technology, Macau SAR, People’s Republic of China ,Guangzhou Laboratory, Guangzhou, People’s Republic of China
| | - Jianxing He
- grid.470124.4State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120 People’s Republic of China
| |
Collapse
|
10
|
Abavisani M, Rahimian K, Kodori M, Khayami R, Mollapour Sisakht M, Mahmanzar M, Meshkat Z. In silico analysis of the substitution mutations and evolutionary trends of the SARS-CoV-2 structural proteins in Asia. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:1299-1307. [PMID: 36474565 PMCID: PMC9699957 DOI: 10.22038/ijbms.2022.66649.14620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 10/01/2022] [Indexed: 12/23/2022]
Abstract
OBJECTIVES To address a highly mutable pathogen, mutations must be evaluated. SARS-CoV-2 involves changing infectivity, mortality, and treatment and vaccination susceptibility resulting from mutations. MATERIALS AND METHODS We investigated the Asian and worldwide samples of amino-acid sequences (AASs) for envelope (E), membrane (M), nucleocapsid (N), and spike (S) proteins from the announcement of the new coronavirus 2019 (COVID-19) up to January 2022. Sequence alignment to the Wuhan-2019 virus permits tracking mutations in Asian and global samples. Furthermore, we explored the evolutionary tendencies of structural protein mutations and compared the results between Asia and the globe. RESULTS The mutation analyses indicated that 5.81%, 70.63%, 26.59%, and 3.36% of Asian S, E, M, and N samples did not display any mutation. Additionally, the most relative mutations among the S, E, M, and N AASs occurred in the regions of 508 to 635 AA, 7 to 14 AA, 66 to 88 AA, and 164 to 205 AA in both Asian and total samples. D614G, T9I, I82T, and R203M were inferred as the most frequent mutations in S, E, M, and N AASs. Timeline research showed that substitution mutation in the location of 614 among Asian and total S AASs was detected from January 2020. CONCLUSION N protein was the most non-conserved protein, and the most prevalent mutations in S, E, M, and N AASs were D614G, T9I, I82T, and R203M. Screening structural protein mutations is a robust approach for developing drugs, vaccines, and more specific diagnostic tools.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran,Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Karim Rahimian
- Bioinformatics and Computational Omics Lab (BioCOOL), Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University
| | | | - Reza Khayami
- Non communicable Diseases Research Center, Bam University of Medical sciences, Bam, Iran
| | - Mahsa Mollapour Sisakht
- Department of Medical Genetics and Molecular Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadamin Mahmanzar
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands
| | - Zahra Meshkat
- Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran ,Corresponding author: Zahra Meshkat. Department of Microbiology and Virology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran. Tel: +98-51-38002313; +98-51-38012453;
| |
Collapse
|
11
|
Quantitatively evaluate the impact of domestic aviation control measures on the spread of COVID-19 in China. Sci Rep 2022; 12:17600. [PMID: 36266307 PMCID: PMC9584274 DOI: 10.1038/s41598-022-21355-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/26/2022] [Indexed: 01/13/2023] Open
Abstract
To quantitatively evaluate the impact of domestic aviation control measures on the spread of COVID-19 in China. The number of international flights from March to September 2019 simulated the number of flights from March to September 2020 without implementing aviation control measures. In addition, the proportion of asymptomatic persons and the delay in case reporting were adjusted to estimate the prevalence of each country during the same period and calculate the estimated imported cases. The estimated imported cases were assigned each day with weight, and the estimated daily reported cases were obtained based on the actual daily number of domestic cases in China. Effective Reproduction Number ([Formula: see text]) was calculated based on delayed distribution, Basic Reproductive Number ([Formula: see text]) distribution, and generation time distribution were reported in previous studies. Gaussian Process was used to estimate the effect of time-varying on [Formula: see text], and the estimated [Formula: see text] was compared with the actual [Formula: see text]. The estimated imported cases increased significantly compared with the actual number of imported cases. The estimated imported cases were mainly concentrated in North America and Europe from March to April and gradually increased in many East Asian countries from May to September. The difference between predicted [Formula: see text] and actual [Formula: see text] was statistically significant. The estimated imported cases and the estimated [Formula: see text] have increased compared to the actual situation. This paper quantitatively proves that Chinese aviation control measures significantly suppress the COVID-19 epidemic, which is conducive to promoting and applying this measure.
Collapse
|
12
|
Daher-Nashif S, Al-Anany R, Ali M, Erradi K, Farag E, Abdallah AM, Emara MM. COVID-19 exit strategy during vaccine implementation: a balance between social distancing and herd immunity. Arch Virol 2022; 167:1773-1783. [PMID: 35723757 PMCID: PMC9208258 DOI: 10.1007/s00705-022-05495-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 04/05/2022] [Indexed: 11/09/2022]
Abstract
Currently, health authorities around the world are struggling to limit the spread of COVID-19. Since the beginning of the pandemic, social distancing has been the most important strategy used by most countries to control disease spread by flattening and elongating the epidemic curve. Another strategy, herd immunity, was also applied by some countries through relaxed control measures that allow the free spread of natural infection to build up solid immunity within the population. In 2021, COVID-19 vaccination was introduced with tremendous effort as a promising strategy for limiting the spread of disease. Therefore, in this review, we present the current knowledge about social distancing, herd immunity strategies, and aspects of their implementation to control the COVID-19 pandemic in the presence of the newly developed vaccines. Finally, we suggest a short-term option for controlling the pandemic during vaccine application.
Collapse
Affiliation(s)
- Suhad Daher-Nashif
- Population Medicine Department, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Rania Al-Anany
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, Doha, Qatar
- Public Health Department, Health Protection and Communicable Diseases, Ministry of Public Health, Doha, Qatar
| | - Menatalla Ali
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Khadija Erradi
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Elmoubasher Farag
- Public Health Department, Health Protection and Communicable Diseases, Ministry of Public Health, Doha, Qatar
| | - Abdallah M Abdallah
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Mohamed M Emara
- Basic Medical Sciences Department, College of Medicine, QU Health, Qatar University, Doha, Qatar.
| |
Collapse
|
13
|
Gilner J, Kansal N, Biggio JR, Delaney S, Grotegut CA, Hardy E, Hirshberg A, Kachikis A, LaCourse SM, Martin J, Metz TD, Miller ES, Norton ME, Sinkey R, Sobhani NC, Son SL, Srinivas S, Tita A, Werner EF, Hughes BL. Universal Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Testing for Obstetric Inpatient Units Across the United States. Clin Infect Dis 2022; 75:e322-e328. [PMID: 34791093 PMCID: PMC8689996 DOI: 10.1093/cid/ciab955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Indexed: 12/01/2022] Open
Abstract
BACKGROUND The purpose of this study was to estimate prevalence of asymptomatic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection among patients admitted to obstetric inpatient units throughout the United States as detected by universal screening. We sought to describe the relationship between obstetric inpatient asymptomatic infection rates and publicly available surrounding community infection rates. METHODS A cross-sectional study in which medical centers reported rates of positive SARS-CoV-2 testing in asymptomatic pregnant and immediate postpartum patients over a 1-3-month time span in 2020. Publicly reported SARS-CoV-2 case rates from the relevant county and state for each center were collected from the COVID Act Now dashboard and the COVID Tracking Project for correlation analysis. RESULTS Data were collected from 9 health centers, encompassing 18 hospitals. Participating health centers were located in Alabama, California, Illinois, Louisiana, New Jersey, North Carolina, Pennsylvania, Rhode Island, Utah, and Washington State. Each hospital had an active policy for universal SARS-CoV-2 testing on obstetric inpatient units. A total of 10 147 SARS-CoV-2 tests were administered, of which 124 were positive (1.2%). Positivity rates varied by site, ranging from 0-3.2%. While SARS-CoV-2 infection rates were lower in asymptomatic obstetric inpatient groups than the surrounding communities, there was a positive correlation between positivity rates in obstetric inpatient units and their surrounding county (P=.003, r=.782) and state (P=.007, r=.708). CONCLUSIONS Given the correlation between community and obstetric inpatient rates, the necessity of SARS-CoV-2-related healthcare resource utilization in obstetric inpatient units may be best informed by surrounding community infection rates.
Collapse
Affiliation(s)
- Jennifer Gilner
- Department of Obstetrics and Gynecology, Duke University, Durham, North Carolina, USA
| | - Namita Kansal
- Department of Obstetrics and Gynecology, Duke University, Durham, North Carolina, USA
| | - Joseph R Biggio
- Section of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Women’s Service Line, Ochsner Health, New Orleans, Louisiana, USA
| | - Shani Delaney
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Chad A Grotegut
- Division of Maternal-Fetal Medicine, Duke University, Durham, North Carolina, USA
| | - Erica Hardy
- Departments of Medicine and Obstetrics and Gynecology, Division of Infectious Disease, Women & Infants Hospital, Providence, Rhode Island, USA
| | - Adi Hirshberg
- Division of Maternal Fetal Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alisa Kachikis
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Sylvia M LaCourse
- Department of Obstetrics and Gynecology, University of Washington, Seattle, Washington, USA
| | - Jane Martin
- Section of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Women’s Service Line, Ochsner Health, New Orleans, Louisiana, USA
| | - Torri D Metz
- University of Utah Health, Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Salt Lake City, Utah, USA
| | - Emily S Miller
- Department of Obstetrics and Gynecology, Division of Maternal Fetal Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Mary E Norton
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, California, USA
| | - Rachel Sinkey
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama, USAand
| | - Nasim C Sobhani
- Division of Maternal Fetal Medicine, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California, San Francisco, California, USA
| | - Shannon L Son
- University of Utah Health, Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, Salt Lake City, Utah, USA
| | - Sindhu Srinivas
- Division of Maternal Fetal Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alan Tita
- Division of Maternal-Fetal Medicine, Department of Obstetrics and Gynecology, University of Alabama at Birmingham, Birmingham, Alabama, USAand
| | - Erika F Werner
- Department of Obstetrics and Gynecology, Tufts Medical Center, North Dartmouth, Massachusetts, USA
| | - Brenna L Hughes
- Department of Obstetrics and Gynecology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
14
|
Brynjolfsson SF, Sigurgrimsdottir H, Gudlaugsson O, Kristjansson M, Kristinsson KG, Ludviksson BR. Determining SARS-CoV-2 non-infectivity state-A brief overview. Front Public Health 2022; 10:934242. [PMID: 36033758 PMCID: PMC9412020 DOI: 10.3389/fpubh.2022.934242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/18/2022] [Indexed: 01/25/2023] Open
Abstract
From the beginning of the COVID-19 pandemic, it has claimed over 6 million lives, and globally the pandemic rages with detrimental consequences, with the emergence of new more infectious and possibly virulent variants. A clinical obstacle in this battle has been to determine when an infected individual has reached a non-infectious state. Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) can be transmitted under diverse circumstances, and various rules and regulations, along with different testing methods, have been applied in an attempt to confine the transmission. However, that has proven to be a difficult task. In this review, we take together recently published data on infectivity and transmission of SARS-CoV-2 and have combined it with the clinical experience that physicians in Iceland have accumulated from the pandemic. In addition, we suggest guidelines for determining when patients with COVID-19 reach a non-infectious state based on a combination of clinical experience, scientific data, and proficient use of available tests. This review has addressed some of the questions regarding contagiousness and immunity against SARS-CoV-2.
Collapse
Affiliation(s)
- Siggeir F. Brynjolfsson
- Department of Immunology, Landspitali—The National University Hospital of Iceland, Reykjavik, Iceland,Department of Medicine, Faculty of Medicine, University of Iceland, Reykjavik, Iceland,*Correspondence: Siggeir F. Brynjolfsson
| | - Hildur Sigurgrimsdottir
- Department of Immunology, Landspitali—The National University Hospital of Iceland, Reykjavik, Iceland,Department of Medicine, Faculty of Medicine, University of Iceland, Reykjavik, Iceland
| | - Olafur Gudlaugsson
- Department of Infectious Diseases, Landspitali—The National University Hospital of Iceland, Reykjavik, Iceland
| | - Mar Kristjansson
- Department of Medicine, Faculty of Medicine, University of Iceland, Reykjavik, Iceland,Department of Infectious Diseases, Landspitali—The National University Hospital of Iceland, Reykjavik, Iceland
| | - Karl G. Kristinsson
- Department of Medicine, Faculty of Medicine, University of Iceland, Reykjavik, Iceland,Department of Clinical Microbiology, Landspitali—The National University Hospital of Iceland, Reykjavik, Iceland
| | - Bjorn R. Ludviksson
- Department of Immunology, Landspitali—The National University Hospital of Iceland, Reykjavik, Iceland,Department of Medicine, Faculty of Medicine, University of Iceland, Reykjavik, Iceland,Bjorn R. Ludviksson
| |
Collapse
|
15
|
Cer RZ, Voegtly LJ, Adhikari BN, Pike BL, Lueder MR, Glang LA, Malagon F, Ana ES, Regeimbal JM, Potts-Szoke MF, Schully KL, Smith DR, Bishop-Lilly KA. Genomic and virologic characterization of samples from a shipboard outbreak of COVID-19 reveals distinct variants within limited temporospatial parameters. Front Microbiol 2022; 13:960932. [PMID: 36033872 PMCID: PMC9399806 DOI: 10.3389/fmicb.2022.960932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 07/05/2022] [Indexed: 11/13/2022] Open
Abstract
Early in the pandemic, in March of 2020, an outbreak of COVID-19 occurred aboard the aircraft carrier USS Theodore Roosevelt (CVN-71), during deployment in the Western Pacific. Out of the crew of 4,779 personnel, 1,331 service members were suspected or confirmed to be infected with SARS-CoV-2. The demographic, epidemiologic, and laboratory findings of service members from subsequent investigations have characterized the outbreak as widespread transmission of virus with relatively mild symptoms and asymptomatic infection among mostly young healthy adults. At the time, there was no available vaccination against COVID-19 and there was very limited knowledge regarding SARS-CoV-2 mutation, dispersal, and transmission patterns among service members in a shipboard environment. Since that time, other shipboard outbreaks from which data can be extracted have occurred, but these later shipboard outbreaks have occurred largely in settings where the majority of the crew were vaccinated, thereby limiting spread of the virus, shortening duration of the outbreaks, and minimizing evolution of the virus within those close quarters settings. On the other hand, since the outbreak on the CVN-71 occurred prior to widespread vaccination, it continued over the course of roughly two months, infecting more than 25% of the crew. In order to better understand genetic variability and potential transmission dynamics of COVID-19 in a shipboard environment of immunologically naïve, healthy individuals, we performed whole-genome sequencing and virus culture from eighteen COVID-19-positive swabs collected over the course of one week. Using the unique variants identified in those genomes, we detected seven discrete groups of individuals within the population aboard CVN-71 infected with viruses of distinct genomic signature. This is in stark contrast to a recent outbreak aboard another U.S. Navy ship with >98% vaccinated crew after a port visit in Reykjavik, Iceland, where the outbreak lasted only approximately 2 weeks and the virus was clonal. Taken together, these results demonstrate the utility of sequencing from complex clinical samples for molecular epidemiology and they also suggest that a high rate of vaccination among a population in close communities may greatly reduce spread, thereby restricting evolution of the virus.
Collapse
Affiliation(s)
- Regina Z. Cer
- Department of Genomics and Bioinformatics, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
| | - Logan J. Voegtly
- Department of Genomics and Bioinformatics, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
- Leidos, Reston, VA, United States
| | - Bishwo N. Adhikari
- Department of Genomics and Bioinformatics, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
- Defense Threat Reduction Agency, Ft. Belvoir, VA, United States
| | - Brian L. Pike
- Department of Operations, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
| | - Matthew R. Lueder
- Department of Genomics and Bioinformatics, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
- Leidos, Reston, VA, United States
| | - Lindsay A. Glang
- Department of Genomics and Bioinformatics, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
- Leidos, Reston, VA, United States
| | - Francisco Malagon
- Department of Genomics and Bioinformatics, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
- Leidos, Reston, VA, United States
| | - Ernesto Santa Ana
- Department of Operations, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
| | - James M. Regeimbal
- Department of Operations, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
| | - Maria F. Potts-Szoke
- Department of Operations, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
| | - Kevin L. Schully
- The Austere Environments Consortium for Enhanced Sepsis Outcomes (ACESO), Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
| | - Darci R. Smith
- Department of Microbiology and Immunology, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
| | - Kimberly A. Bishop-Lilly
- Department of Genomics and Bioinformatics, Biological Defense Research Directorate, Naval Medical Research Center, Fort Detrick, MD, United States
- *Correspondence: Kimberly A. Bishop-Lilly,
| |
Collapse
|
16
|
Goes LR, Siqueira JD, Garrido MM, Alves BM, Cicala C, Arthos J, Viola JPB, Soares MA. Evidence of recurrent selection of mutations commonly found in SARS-CoV-2 variants of concern in viruses infecting immunocompromised patients. Front Microbiol 2022; 13:946549. [PMID: 35958144 PMCID: PMC9362983 DOI: 10.3389/fmicb.2022.946549] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Chronically immunosuppressed patients infected with SARS-CoV-2 often experience prolonged virus shedding, and may pave the way to the emergence of mutations that render viral variants of concern (VOC) able to escape immune responses induced by natural infection or by vaccination. We report herein a SARS-CoV-2+ cancer patient from the beginning of the COVID-19 pandemic whose virus quasispecies across multiple timepoints carried several immune escape mutations found in more contemporary VOC, such as alpha, delta and omicron, that appeared to be selected for during infection. We hypothesize that immunosuppressed patients may represent the source of VOC seen throughout the COVID-19 pandemics.
Collapse
Affiliation(s)
- Livia R. Goes
- Oncovirology Program, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
- *Correspondence: Livia R. Goes,
| | - Juliana D. Siqueira
- Oncovirology Program, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Marianne M. Garrido
- Hospital Infection Control Committee, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Brunna M. Alves
- Oncovirology Program, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Claudia Cicala
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - James Arthos
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - João P. B. Viola
- Immunology and Tumor Biology Program, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
| | - Marcelo A. Soares
- Oncovirology Program, Brazilian National Cancer Institute, Rio de Janeiro, Brazil
- Department of Genetics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- Marcelo A. Soares,
| |
Collapse
|
17
|
Vassilaki N, Papadimitriou K, Ioannidis A, Papandreou NC, Milona RS, Iconomidou VA, Chatzipanagiotou S. SARS-CoV-2 Amino Acid Mutations Detection in Greek Patients Infected in the First Wave of the Pandemic. Microorganisms 2022; 10:microorganisms10071430. [PMID: 35889149 PMCID: PMC9322066 DOI: 10.3390/microorganisms10071430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel virus that belongs to the Coronoviridae family, emerged in December 2019, causing the COVID-19 pandemic in March 2020. Unlike previous SARS and Middle East respiratory syndrome (MERS) outbreaks, this virus has a higher transmissibility rate, albeit a lower case fatality rate, which results in accumulation of a significant number of mutations and a faster evolution rate. Genomic studies on the mutation rate of the virus, as well as the identification of mutations that prevail and their impact on disease severity, are of great importance for pandemic surveillance and vaccine and drug development. Here, we aim to identify mutations on the SARS-CoV-2 viral genome and their effect on the proteins they are located in, in Greek patients infected in the first wave of the pandemic. To this end, we perform SARS-CoV-2 amplicon-based NGS sequencing on nasopharyngeal swab samples from Greek patients and bioinformatic analysis of the results. Although SARS-CoV-2 is considered genetically stable, we discover a variety of mutations on the viral genome. In detail, 18 mutations are detected in total on 10 SARS-CoV-2 isolates. The mutations are located on ORF1ab, S protein, M protein, ORF3a and ORF7a. Sixteen are also detected in patients from other regions around the world, and two are identified for the first time in the present study. Most of them result in amino acid substitutions. These substitutions are analyzed using computational tools, and the results indicate minor or major impact on the proteins’ structural stability, which could probably affect viral transmissibility and pathogenesis. The correlation of these variations with the viral load levels is examined, and their implication for disease severity and the biology of the virus are discussed.
Collapse
Affiliation(s)
- Niki Vassilaki
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521 Athens, Greece; (N.V.); (R.S.M.)
| | - Konstantinos Papadimitriou
- Laboratory of Food Quality Control and Hygiene, Department of Food Science and Human Nutrition, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece;
| | - Anastasios Ioannidis
- Department of Nursing, Faculty of Health Sciences, University of Peloponnese, Sehi Area, 22100 Tripoli, Greece;
| | - Nikos C. Papandreou
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, Panepistimiopolis, 15701 Athens, Greece; (N.C.P.); (V.A.I.)
| | - Raphaela S. Milona
- Laboratory of Molecular Virology, Hellenic Pasteur Institute, 127 Vasilissis Sofias Avenue, 11521 Athens, Greece; (N.V.); (R.S.M.)
| | - Vassiliki A. Iconomidou
- Section of Cell Biology and Biophysics, Department of Biology, School of Science, National and Kapodistrian University of Athens, Panepistimiopolis, 15701 Athens, Greece; (N.C.P.); (V.A.I.)
| | - Stylianos Chatzipanagiotou
- Department of Medical Biopathology, Eginition Hospital, Athens Medical School, National and Kapodistrian University of Athens, 72–74 Vasilissis Sofias Avenue, 11528 Athens, Greece
- Correspondence:
| |
Collapse
|
18
|
Gao K, Wang R, Chen J, Cheng L, Frishcosy J, Huzumi Y, Qiu Y, Schluckbier T, Wei X, Wei GW. Methodology-Centered Review of Molecular Modeling, Simulation, and Prediction of SARS-CoV-2. Chem Rev 2022; 122:11287-11368. [PMID: 35594413 PMCID: PMC9159519 DOI: 10.1021/acs.chemrev.1c00965] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Despite tremendous efforts in the past two years, our understanding of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), virus-host interactions, immune response, virulence, transmission, and evolution is still very limited. This limitation calls for further in-depth investigation. Computational studies have become an indispensable component in combating coronavirus disease 2019 (COVID-19) due to their low cost, their efficiency, and the fact that they are free from safety and ethical constraints. Additionally, the mechanism that governs the global evolution and transmission of SARS-CoV-2 cannot be revealed from individual experiments and was discovered by integrating genotyping of massive viral sequences, biophysical modeling of protein-protein interactions, deep mutational data, deep learning, and advanced mathematics. There exists a tsunami of literature on the molecular modeling, simulations, and predictions of SARS-CoV-2 and related developments of drugs, vaccines, antibodies, and diagnostics. To provide readers with a quick update about this literature, we present a comprehensive and systematic methodology-centered review. Aspects such as molecular biophysics, bioinformatics, cheminformatics, machine learning, and mathematics are discussed. This review will be beneficial to researchers who are looking for ways to contribute to SARS-CoV-2 studies and those who are interested in the status of the field.
Collapse
Affiliation(s)
- Kaifu Gao
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Rui Wang
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jiahui Chen
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Limei Cheng
- Clinical
Pharmacology and Pharmacometrics, Bristol
Myers Squibb, Princeton, New Jersey 08536, United States
| | - Jaclyn Frishcosy
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuta Huzumi
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Yuchi Qiu
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Tom Schluckbier
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Xiaoqi Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
| | - Guo-Wei Wei
- Department
of Mathematics, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Electrical and Computer Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Biochemistry and Molecular Biology, Michigan
State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
19
|
Liu L, Ross NM, Handorf EA, Meeker CR, Chen G, Baldwin D, Vijayvergia N. Incidence of asymptomatic COVID-19 positivity in cancer patients and effects on therapy. RESEARCH SQUARE 2022:rs.3.rs-1785577. [PMID: 35794894 PMCID: PMC9258301 DOI: 10.21203/rs.3.rs-1785577/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Purpose The coronavirus disease 2019 (COVID-19) pandemic is posing unprecedented challenges for patient care, especially for cancer patients. This study looks at asymptomatic (AS) COVID-19 positivity in cancer patients and its effects on their care. Methods We conducted a retrospective chart review of AS patients testing positive for COVID-19 upon screening at Fox Chase Cancer Center between January 2020 and September 2020. Relationships between positive tests and demographics, clinical characteristics, and treatment delays were investigated using conditional logistic regression or Mantel-Haenszel tests. Results Among 4143 AS patients who underwent COVID-19 testing, 25 (0.6%) were COVID-19 positive (cases) and these were matched to 50 controls. The median age was lower in the cases compared to that of the controls (64 vs 70 years old, p = 0.04). Of the cases, 10 patients (40%) never underwent their planned oncologic intervention [6/10 (60%) did not require the planned intervention once deemed okay to proceed]. Of the controls, only 1 patient (2%) did not undergo the planned intervention. Of these 15 COVID-19 positive patients who underwent the planned intervention, 11 (73.3%) had a delay related to COVID-19, with a mean delay duration of 18 days (range: 0-49, SD: 16.72). Conclusion Cancer patients had lower incidence of AS COVID-19 than general population. Delays that occur due to AS COVID screening are not very long and serve as a tool to limit spread of virus. Further studies will be important in addressing delays in cancer care and concerns of patient safety as the pandemic continues.
Collapse
|
20
|
Markarian NM, Galli G, Patel D, Hemmings M, Nagpal P, Berghuis AM, Abrahamyan L, Vidal SM. Identifying Markers of Emerging SARS-CoV-2 Variants in Patients With Secondary Immunodeficiency. Front Microbiol 2022; 13:933983. [PMID: 35847101 PMCID: PMC9283111 DOI: 10.3389/fmicb.2022.933983] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/31/2022] [Indexed: 12/03/2022] Open
Abstract
Since the end of 2019, the world has been challenged by the coronavirus disease 2019 (COVID-19) pandemic. With COVID-19 cases rising globally, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to evolve, resulting in the emergence of variants of interest (VOI) and of concern (VOC). Of the hundreds of millions infected, immunodeficient patients are one of the vulnerable cohorts that are most susceptible to this virus. These individuals include those with preexisting health conditions and/or those undergoing immunosuppressive treatment (secondary immunodeficiency). In these cases, several researchers have reported chronic infections in the presence of anti-COVID-19 treatments that may potentially lead to the evolution of the virus within the host. Such variations occurred in a variety of viral proteins, including key structural ones involved in pathogenesis such as spike proteins. Tracking and comparing such mutations with those arisen in the general population may provide information about functional sites within the SARS-CoV-2 genome. In this study, we reviewed the current literature regarding the specific features of SARS-CoV-2 evolution in immunocompromised patients and identified recurrent de novo amino acid changes in virus isolates of these patients that can potentially play an important role in SARS-CoV-2 pathogenesis and evolution.
Collapse
Affiliation(s)
- Nathan M. Markarian
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
- Swine and Poultry Infectious Diseases Research Center and Research Group on Infectious Diseases in Production Animals, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Gaël Galli
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, QC, Canada
- CNRS, ImmunoConcEpT, UMR 5164, Université de Bordeaux, Bordeaux, France
- CHU de Bordeaux, FHU ACRONIM, Centre National de Référence des Maladies Auto-Immunes et Systémiques Rares Est/Sud-Ouest, Bordeaux, France
| | - Dhanesh Patel
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
| | - Mark Hemmings
- Department of Biochemistry, McGill University, Montréal, QC, Canada
| | - Priya Nagpal
- Department of Pharmacology, McGill University, Montréal, QC, Canada
| | | | - Levon Abrahamyan
- Swine and Poultry Infectious Diseases Research Center and Research Group on Infectious Diseases in Production Animals, Faculty of Veterinary Medicine, University of Montreal, Saint-Hyacinthe, QC, Canada
| | - Silvia M. Vidal
- Department of Human Genetics, McGill University, Montréal, QC, Canada
- McGill University Research Centre on Complex Traits, Montréal, QC, Canada
| |
Collapse
|
21
|
Prince T, Dong X, Penrice-Randal R, Randle N, Hartley C, Goldswain H, Jones B, Semple MG, Baillie JK, Openshaw PJM, Turtle L, Hughes GL, Anderson ER, Patterson EI, Druce J, Screaton G, Carroll MW, Stewart JP, Hiscox JA. Analysis of SARS-CoV-2 in Nasopharyngeal Samples from Patients with COVID-19 Illustrates Population Variation and Diverse Phenotypes, Placing the Growth Properties of Variants of Concern in Context with Other Lineages. mSphere 2022; 7:e0091321. [PMID: 35491827 PMCID: PMC9241508 DOI: 10.1128/msphere.00913-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 03/18/2022] [Indexed: 01/02/2023] Open
Abstract
New variants of SARS-CoV-2 are continuing to emerge and dominate the global sequence landscapes. Several variants have been labeled variants of concern (VOCs) because they may have a transmission advantage, increased risk of morbidity and/or mortality, or immune evasion upon a background of prior infection or vaccination. Placing the VOCs in context with the underlying variability of SARS-CoV-2 is essential in understanding virus evolution and selection pressures. Dominant genome sequences and the population genetics of SARS-CoV-2 in nasopharyngeal swabs from hospitalized patients were characterized. Nonsynonymous changes at a minor variant level were identified. These populations were generally preserved when isolates were amplified in cell culture. To place the Alpha, Beta, Delta, and Omicron VOCs in context, their growth was compared to clinical isolates of different lineages from earlier in the pandemic. The data indicated that the growth in cell culture of the Beta variant was more than that of the other variants in Vero E6 cells but not in hACE2-A549 cells. Looking at each time point, Beta grew more than the other VOCs in hACE2-A549 cells at 24 to 48 h postinfection. At 72 h postinfection there was no difference in the growth of any of the variants in either cell line. Overall, this work suggested that exploring the biology of SARS-CoV-2 is complicated by population dynamics and that these need to be considered with new variants. In the context of variation seen in other coronaviruses, the variants currently observed for SARS-CoV-2 are very similar in terms of their clinical spectrum of disease. IMPORTANCE SARS-CoV-2 is the causative agent of COVID-19. The virus has spread across the planet, causing a global pandemic. In common with other coronaviruses, SARS-CoV-2 genomes can become quite diverse as a consequence of replicating inside cells. This has given rise to multiple variants from the original virus that infected humans. These variants may have different properties and in the context of a widespread vaccination program may render vaccines less effective. Our research confirms the degree of genetic diversity of SARS-CoV-2 in patients. By comparing the growth of previous variants to the pattern seen with four variants of concern (VOCs) (Alpha, Beta, Delta, and Omicron), we show that, at least in cells, Beta variant growth exceeds that of Alpha, Delta, and Omicron VOCs at 24 to 48 h in both Vero E6 and hACE2-A549 cells, but by 72 h postinfection, the amount of virus is not different from that of the other VOCs.
Collapse
Affiliation(s)
- Tessa Prince
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, United Kingdom
| | - Xiaofeng Dong
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Rebekah Penrice-Randal
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Nadine Randle
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Catherine Hartley
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Hannah Goldswain
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Benjamin Jones
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Malcolm G. Semple
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, United Kingdom
- Department of Respiratory Medicine, Alder Hey Children’s Hospital, Liverpool, United Kingdom
| | - J. Kenneth Baillie
- The Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Peter J. M. Openshaw
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Lance Turtle
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, United Kingdom
| | - Grant L. Hughes
- Departments of Vector Biology and Tropical Disease Biology, Center for Neglected Tropical Diseases, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Enyia R. Anderson
- Departments of Vector Biology and Tropical Disease Biology, Center for Neglected Tropical Diseases, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Edward I. Patterson
- Departments of Vector Biology and Tropical Disease Biology, Center for Neglected Tropical Diseases, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Julian Druce
- Virus Identification Laboratory, Doherty Institute, University of Melbourne, Melbourne, Australia
| | - Gavin Screaton
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Miles W. Carroll
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- Public Health England, Salisbury, United Kingdom
| | - James P. Stewart
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- Department of Infectious Disease, University of Georgia, Georgia, USA
| | - Julian A. Hiscox
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Liverpool, United Kingdom
- A*STAR Infectious Diseases Laboratories (A*STAR ID Labs), Agency for Science, Technology and Research (A*STAR), Singapore
| |
Collapse
|
22
|
Performance of the TaqMan COVID-19 Pooling Kit for detection of SARS-CoV-2 in asymptomatic and symptomatic populations. PLoS One 2022; 17:e0269798. [PMID: 35687578 PMCID: PMC9187085 DOI: 10.1371/journal.pone.0269798] [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: 11/29/2021] [Accepted: 05/30/2022] [Indexed: 11/24/2022] Open
Abstract
Clinical evidence for asymptomatic cases of coronavirus disease (COVID-19) has reinforced the significance of effective surveillance testing programs. Quantitative reverse transcriptase polymerase chain reaction (RT-qPCR) assays are considered the ‘gold standard’ for detection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) RNA. However, the labor and resource requirements can be prohibitive with respect to large testing volumes associated with the pandemic. Pooled testing algorithms may serve to increase testing capacity with more efficient resource utilization. Due to the lack of carefully curated cohorts, there is limited evidence for the applicability of RT-PCR pooling in asymptomatic COVID-19 cases. In this study, we compared the analytical sensitivity of the TaqMan™ SARS-CoV-2 Pooling Assay to detect one positive sample in a pool of five anterior nares swabs in symptomatic and asymptomatic cohorts at an institute of higher education. Positive pools were deconvoluted and each individual sample was retested using the TaqPath™ COVID-19 Combo Kit. Both assays target the open reading frame (ORF) 1ab, nucleocapsid (N), and spike (S) gene of the strain that originated in Wuhan, Hubei, China. Qualitative results demonstrated absolute agreement between pooled and deconvoluted samples in both cohorts. Independent t-test performed on Ct shifts supported an insignificant difference between cohorts with p-values of 0.306 (Orf1ab), 0.147 (N), and 0.052 (S). All negative pools were correctly reported as negative. Pooled PCR testing up to five samples is a valid method for surveillance testing of students and staff in a university setting, especially when the prevalence is expected to be low.
Collapse
|
23
|
SARS-CoV-2 non-structural protein 6 triggers NLRP3-dependent pyroptosis by targeting ATP6AP1. Cell Death Differ 2022; 29:1240-1254. [PMID: 34997207 PMCID: PMC9177730 DOI: 10.1038/s41418-021-00916-7] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 11/25/2021] [Accepted: 11/25/2021] [Indexed: 01/18/2023] Open
Abstract
A recent mutation analysis suggested that Non-Structural Protein 6 (NSP6) of the Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is a key determinant of the viral pathogenicity. Here, by transcriptome analysis, we demonstrated that the inflammasome-related NOD-like receptor signaling was activated in SARS-CoV-2-infected lung epithelial cells and Coronavirus Disease 2019 (COVID-19) patients' lung tissues. The induction of inflammasomes/pyroptosis in patients with severe COVID-19 was confirmed by serological markers. Overexpression of NSP6 triggered NLRP3/ASC-dependent caspase-1 activation, interleukin-1β/18 maturation, and pyroptosis of lung epithelial cells. Upstream, NSP6 impaired lysosome acidification to inhibit autophagic flux, whose restoration by 1α,25-dihydroxyvitamin D3, metformin or polydatin abrogated NSP6-induced pyroptosis. NSP6 directly interacted with ATP6AP1, a vacuolar ATPase proton pump component, and inhibited its cleavage-mediated activation. L37F NSP6 variant, which was associated with asymptomatic COVID-19, exhibited reduced binding to ATP6AP1 and weakened ability to impair lysosome acidification to induce pyroptosis. Consistently, infection of cultured lung epithelial cells with live SARS-CoV-2 resulted in autophagic flux stagnation, inflammasome activation, and pyroptosis. Overall, this work supports that NSP6 of SARS-CoV-2 could induce inflammatory cell death in lung epithelial cells, through which pharmacological rectification of autophagic flux might be therapeutically exploited.
Collapse
|
24
|
Newly Emerged Antiviral Strategies for SARS-CoV-2: From Deciphering Viral Protein Structural Function to the Development of Vaccines, Antibodies, and Small Molecules. Int J Mol Sci 2022; 23:ijms23116083. [PMID: 35682761 PMCID: PMC9181103 DOI: 10.3390/ijms23116083] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 05/20/2022] [Accepted: 05/27/2022] [Indexed: 01/09/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) caused by the infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become the most severe health crisis, causing extraordinary economic disruption worldwide. SARS-CoV-2 is a single-stranded RNA-enveloped virus. The process of viral replication and particle packaging is finished in host cells. Viral proteins, including both structural and nonstructural proteins, play important roles in the viral life cycle, which also provides the targets of treatment. Therefore, a better understanding of the structural function of virus proteins is crucial to speed up the development of vaccines and therapeutic strategies. Currently, the structure and function of proteins encoded by the SARS-CoV-2 genome are reviewed by several studies. However, most of them are based on the analysis of SARS-CoV-1 particles, lacking a systematic review update for SARS-CoV-2. Here, we specifically focus on the structure and function of proteins encoded by SARS-CoV-2. Viral proteins that contribute to COVID-19 infection and disease pathogenesis are reviewed according to the most recent research findings. The structure-function correlation of viral proteins provides a fundamental rationale for vaccine development and targeted therapy. Then, current antiviral vaccines are updated, such as inactive viral vaccines and protein-based vaccines and DNA, mRNA, and circular RNA vaccines. A summary of other therapeutic options is also reviewed, including monoclonal antibodies such as a cross-neutralizer antibody, a constructed cobinding antibody, a dual functional monoclonal antibody, an antibody cocktail, and an engineered bispecific antibody, as well as peptide-based inhibitors, chemical compounds, and clustered regularly interspaced short palindromic repeats (CRISPR) exploration. Overall, viral proteins and their functions provide the basis for targeted therapy and vaccine development.
Collapse
|
25
|
Yavarian J, Nejati A, Salimi V, Shafiei Jandaghi NZ, Sadeghi K, Abedi A, Sharifi Zarchi A, Gouya MM, Mokhtari-Azad T. Whole genome sequencing of SARS-CoV2 strains circulating in Iran during five waves of pandemic. PLoS One 2022; 17:e0267847. [PMID: 35499994 PMCID: PMC9060343 DOI: 10.1371/journal.pone.0267847] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 04/14/2022] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Whole genome sequencing of SARS-CoV2 is important to find useful information about the viral lineages, variants of interests and variants of concern. As there are not enough data about the circulating SARS-CoV2 variants in Iran, we sequenced 54 SARS-CoV2 genomes during the 5 waves of pandemic in Iran. METHODS After viral RNA extraction from clinical samples collected during the COVID-19 pandemic, next generation sequencing was performed using the Nextseq platform. The sequencing data were analyzed and compared with reference sequences. RESULTS During the 1st wave, V and L clades were detected. The second wave was recognized by G, GH and GR clades. Circulating clades during the 3rd wave were GH and GR. In the fourth wave GRY (alpha variant), GK (delta variant) and one GH clade (beta variant) were detected. All viruses in the fifth wave were in clade GK (delta variant). There were different mutations in all parts of the genomes but Spike-D614G, NSP12-P323L, N-R203K and N-G204R were the most frequent mutants in these studied viruses. CONCLUSIONS These findings display the significance of SARS-CoV2 monitoring to help on time detection of possible variants for pandemic control and vaccination plans.
Collapse
Affiliation(s)
- Jila Yavarian
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Nejati
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Salimi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Kaveh Sadeghi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Adel Abedi
- Mathematics Department, Shahid Beheshti University, Tehran, Iran
| | - Ali Sharifi Zarchi
- Department of Computer Engineering, Sharif University of Technology, Tehran, Iran
| | | | - Talat Mokhtari-Azad
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Dumache R, Enache A, Macasoi I, Dehelean CA, Dumitrascu V, Mihailescu A, Popescu R, Vlad D, Vlad CS, Muresan C. SARS-CoV-2: An Overview of the Genetic Profile and Vaccine Effectiveness of the Five Variants of Concern. Pathogens 2022; 11:pathogens11050516. [PMID: 35631037 PMCID: PMC9144800 DOI: 10.3390/pathogens11050516] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/21/2022] [Accepted: 04/25/2022] [Indexed: 12/13/2022] Open
Abstract
With the onset of the COVID-19 pandemic, enormous efforts have been made to understand the genus SARS-CoV-2. Due to the high rate of global transmission, mutations in the viral genome were inevitable. A full understanding of the viral genome and its possible changes represents one of the crucial aspects of pandemic management. Structural protein S plays an important role in the pathogenicity of SARS-CoV-2, mutations occurring at this level leading to viral forms with increased affinity for ACE2 receptors, higher transmissibility and infectivity, resistance to neutralizing antibodies and immune escape, increasing the risk of infection and disease severity. Thus, five variants of concern are currently being discussed, Alpha, Beta, Gamma, Delta and Omicron. In the present review, a comprehensive summary of the following critical aspects regarding SARS-CoV-2 has been made: (i) the genomic characteristics of SARS-CoV-2; (ii) the pathological mechanism of transmission, penetration into the cell and action on specific receptors; (iii) mutations in the SARS-CoV-2 genome; and (iv) possible implications of mutations in diagnosis, treatment, and vaccination.
Collapse
Affiliation(s)
- Raluca Dumache
- Ethics and Human Identification Research Center, Department of Neurosciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.D.); (A.E.); (A.M.); (C.M.)
| | - Alexandra Enache
- Ethics and Human Identification Research Center, Department of Neurosciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.D.); (A.E.); (A.M.); (C.M.)
| | - Ioana Macasoi
- Departament of Toxicology and Drug Industry, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Correspondence: (I.M.); (C.A.D.)
| | - Cristina Adriana Dehelean
- Departament of Toxicology and Drug Industry, Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timişoara, Romania
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
- Correspondence: (I.M.); (C.A.D.)
| | - Victor Dumitrascu
- Department of Pharmacology and Biochemistry, Discipline of Pharmacology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (V.D.); (D.V.); (C.S.V.)
| | - Alexandra Mihailescu
- Ethics and Human Identification Research Center, Department of Neurosciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.D.); (A.E.); (A.M.); (C.M.)
- Genetics, Genomic Medicine Research Center, Department of Microscopic Morphology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Roxana Popescu
- Department of Microscopic Morphology, Discipline of Molecular and Cell Biology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania;
| | - Daliborca Vlad
- Department of Pharmacology and Biochemistry, Discipline of Pharmacology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (V.D.); (D.V.); (C.S.V.)
| | - Cristian Sebastian Vlad
- Department of Pharmacology and Biochemistry, Discipline of Pharmacology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (V.D.); (D.V.); (C.S.V.)
| | - Camelia Muresan
- Ethics and Human Identification Research Center, Department of Neurosciences, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania; (R.D.); (A.E.); (A.M.); (C.M.)
| |
Collapse
|
27
|
Anwar MZ, Lodhi MS, Khan MT, Khan MI, Sharif S. Coronavirus Genomes and Unique Mutations in Structural and Non-Structural Proteins in Pakistani SARS-CoV-2 Delta Variants during the Fourth Wave of the Pandemic. Genes (Basel) 2022; 13:552. [PMID: 35328105 PMCID: PMC8951394 DOI: 10.3390/genes13030552] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/10/2022] [Accepted: 03/18/2022] [Indexed: 12/15/2022] Open
Abstract
Genomic epidemiology of SARS-CoV-2 is imperative to explore the transmission, evolution, and also pathogenicity of viruses. The emergence of SARS-CoV-2 variants of concern posed a severe threat to the global public health efforts. To assess the potential consequence of these emerging variants on public health, continuous molecular epidemiology is of vital importance. The current study has been designed to investigate the major SARS-CoV-2 variants and emerging mutations in virus structural and non-structural proteins (NSP) during the fourth wave in September 2021 from the Punjab province of Pakistan. Twenty SARS-CoV-2 positive samples have been collected from major cities were subjected to next-generation sequencing. Among the 20 whole genomes (GenBank Accession SRR16294858-SRR16294877), 2 samples failed to be completely sequenced. These genome sequences harbored 207 non-synonymous mutations, among which 19 were unique to GISAID. The genome sequences were detected: Delta 21I, 21J variants (B.1.617.2). Mutation's spike_F157del, spike_P681R, spike_T478K, spike_T19R, spike_L452R, spike_D614G, spike_G142D, spike_E156G, and spike_R158del have been detected in all samples where K1086Q, E554K, and C1250W were unique in spike protein. These genomic sequences also harbored 129 non-synonymous mutations in NSP. The most common were NSP3_P1469S (N = 17), NSP3_A488S (N = 17), NSP3_P1228L (N = 17), NSP4_V167L (N = 17), NSP4_T492I (N = 17), NSP6_T77A (N = 17), NSP14_A394V (N = 17), NSP12_G671S (N = 18), and NSP13_P77L (N = 18). The mutation, F313Y in NSP12, detected in the current study, was found in a single isolate from Belgium. Numerous other unique mutations have been detected in the virus papain-like protease (NSP3), main protease (NSP5), and RNA-dependent RNA polymerase (NSP12). The most common non-synonymous mutations in the spike protein were subjected to stability analysis, exhibiting a stabilizing effect on structures. The presence of Delta variants may affect therapeutic efforts and vaccine efficacy. Continuous genomic epidemiology of SARS-CoV-2 in Pakistan may be useful for better management of SARS-CoV-2 infections.
Collapse
Affiliation(s)
| | - Madeeha Shahzad Lodhi
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, 1 KM Defence Road, Lahore 58 810, Pakistan; (M.Z.A.); (M.T.K.); (M.I.K.); (S.S.)
| | | | | | | |
Collapse
|
28
|
Lo Presti A, Di Martino A, Faggioni G, Giordani F, Fillo S, Anselmo A, Fain VV, Fortunato A, Petralito G, Molinari F, Palomba S, De Santis R, Fiore S, Fabiani C, Di Mario G, Facchini M, Calzoletti L, Lista F, Rezza G, Stefanelli P. Analysis of Genomic Characteristics of SARS-CoV-2 in Italy, 29 January to 27 March 2020. Viruses 2022; 14:472. [PMID: 35336879 PMCID: PMC8951147 DOI: 10.3390/v14030472] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 02/01/2022] [Accepted: 02/09/2022] [Indexed: 12/14/2022] Open
Abstract
We performed next-generation sequencing (NGS), phylogenetic analysis, gene flows, and N- and O-glycosylation prediction on SARS-CoV-2 genomes collected from lab-confirmed cases from different Italian regions. To this end, a total of 111 SARS-CoV-2 genomes collected in Italy between 29 January and 27 March 2020 were investigated. The majority of the genomes belonged to lineage B.1, with some descendant lineages. The gene flow analysis showed that the spread occurred mainly from the north to the center and to the south of Italy, as confirmed by epidemiological data. The mean evolutionary rate estimated here was 8.731 × 10-4 (95% highest posterior density, HPD intervals 5.809 × 10-4 to 1.19 × 10-3), in line with values reported by other authors. The dated phylogeny suggested that SARS-CoV-2 lineage B.1 probably entered Italy between the end of January and early February 2020. Continuous molecular surveillance is needed to trace virus circulation and evolution.
Collapse
Affiliation(s)
- Alessandra Lo Presti
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.D.M.); (S.F.); (C.F.); (G.D.M.); (M.F.); (L.C.); (P.S.)
| | - Angela Di Martino
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.D.M.); (S.F.); (C.F.); (G.D.M.); (M.F.); (L.C.); (P.S.)
| | - Giovanni Faggioni
- Scientific Department, Army Medical Center, 00184 Rome, Italy; (G.F.); (F.G.); (S.F.); (A.A.); (V.V.F.); (A.F.); (G.P.); (F.M.); (R.D.S.); (F.L.)
| | - Francesco Giordani
- Scientific Department, Army Medical Center, 00184 Rome, Italy; (G.F.); (F.G.); (S.F.); (A.A.); (V.V.F.); (A.F.); (G.P.); (F.M.); (R.D.S.); (F.L.)
| | - Silvia Fillo
- Scientific Department, Army Medical Center, 00184 Rome, Italy; (G.F.); (F.G.); (S.F.); (A.A.); (V.V.F.); (A.F.); (G.P.); (F.M.); (R.D.S.); (F.L.)
| | - Anna Anselmo
- Scientific Department, Army Medical Center, 00184 Rome, Italy; (G.F.); (F.G.); (S.F.); (A.A.); (V.V.F.); (A.F.); (G.P.); (F.M.); (R.D.S.); (F.L.)
| | - Vanessa Vera Fain
- Scientific Department, Army Medical Center, 00184 Rome, Italy; (G.F.); (F.G.); (S.F.); (A.A.); (V.V.F.); (A.F.); (G.P.); (F.M.); (R.D.S.); (F.L.)
- Department of Science, University of Rome “Roma Tre”, 00146 Rome, Italy
| | - Antonella Fortunato
- Scientific Department, Army Medical Center, 00184 Rome, Italy; (G.F.); (F.G.); (S.F.); (A.A.); (V.V.F.); (A.F.); (G.P.); (F.M.); (R.D.S.); (F.L.)
| | - Giancarlo Petralito
- Scientific Department, Army Medical Center, 00184 Rome, Italy; (G.F.); (F.G.); (S.F.); (A.A.); (V.V.F.); (A.F.); (G.P.); (F.M.); (R.D.S.); (F.L.)
| | - Filippo Molinari
- Scientific Department, Army Medical Center, 00184 Rome, Italy; (G.F.); (F.G.); (S.F.); (A.A.); (V.V.F.); (A.F.); (G.P.); (F.M.); (R.D.S.); (F.L.)
| | - Stefano Palomba
- General Directorate of Military Medical Services-Medical Situation Awareness Branch, 00184 Rome, Italy;
| | - Riccardo De Santis
- Scientific Department, Army Medical Center, 00184 Rome, Italy; (G.F.); (F.G.); (S.F.); (A.A.); (V.V.F.); (A.F.); (G.P.); (F.M.); (R.D.S.); (F.L.)
| | - Stefano Fiore
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.D.M.); (S.F.); (C.F.); (G.D.M.); (M.F.); (L.C.); (P.S.)
| | - Concetta Fabiani
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.D.M.); (S.F.); (C.F.); (G.D.M.); (M.F.); (L.C.); (P.S.)
| | - Giuseppina Di Mario
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.D.M.); (S.F.); (C.F.); (G.D.M.); (M.F.); (L.C.); (P.S.)
| | - Marzia Facchini
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.D.M.); (S.F.); (C.F.); (G.D.M.); (M.F.); (L.C.); (P.S.)
| | - Laura Calzoletti
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.D.M.); (S.F.); (C.F.); (G.D.M.); (M.F.); (L.C.); (P.S.)
| | - Florigio Lista
- Scientific Department, Army Medical Center, 00184 Rome, Italy; (G.F.); (F.G.); (S.F.); (A.A.); (V.V.F.); (A.F.); (G.P.); (F.M.); (R.D.S.); (F.L.)
| | - Giovanni Rezza
- Health Prevention Directorate, Ministry of Health, 00144 Rome, Italy;
| | - Paola Stefanelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.D.M.); (S.F.); (C.F.); (G.D.M.); (M.F.); (L.C.); (P.S.)
| |
Collapse
|
29
|
Marçalo R, Neto S, Pinheiro M, Rodrigues AJ, Sousa N, Santos MAS, Simão P, Valente C, Andrade L, Marques A, Moura GR. Evaluation of the genetic risk for COVID-19 outcomes in COPD and differences among worldwide populations. PLoS One 2022; 17:e0264009. [PMID: 35196333 PMCID: PMC8865687 DOI: 10.1371/journal.pone.0264009] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 02/02/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Populations seem to respond differently to the global pandemic of severe acute respiratory syndrome coronavirus 2. Recent studies show individual variability in both susceptibility and clinical response to COVID-19 infection. People with chronic obstructive pulmonary disease (COPD) constitute one of COVID-19 risk groups, being already associated with a poor prognosis upon infection. This study aims contributing to unveil the underlying reasons for such prognosis in people with COPD and the variability in the response observed across worldwide populations, by looking at the genetic background as a possible answer to COVID-19 infection response heterogeneity. METHODS SNPs already associated with susceptibility to COVID-19 infection (rs286914 and rs12329760) and severe COVID-19 with respiratory failure (rs657152 and rs11385942) were assessed and their allelic frequencies used to calculate the probability of having multiple risk alleles. This was performed on a Portuguese case-control COPD cohort, previously clinically characterized and genotyped from saliva samples, and also on worldwide populations (European, Spanish, Italian, African, American and Asian), using publicly available frequencies data. A polygenic risk analysis was also conducted on the Portuguese COPD cohort for the two mentioned phenotypes, and also for hospitalization and survival to COVID-19 infection. FINDINGS No differences in genetic risk for COVID-19 susceptibility, hospitalization, severity or survival were found between people with COPD and the control group (all p-values > 0.01), either considering risk alleles individually, allelic combinations or polygenic risk scores. All populations, even those with European ancestry (Portuguese, Spanish and Italian), showed significant differences from the European population in genetic risk for both COVID-19 susceptibility and severity (all p-values < 0.0001). CONCLUSION Our results indicate a low genetic contribution for COVID-19 infection predisposition or worse outcomes observed in people with COPD. Also, our study unveiled a high genetic heterogeneity across major world populations for the same alleles, even within European sub-populations, demonstrating the need to build a higher resolution European genetic map, so that differences in the distribution of relevant alleles can be easily accessed and used to better manage diseases, ultimately, safeguarding populations with higher genetic predisposition to such diseases.
Collapse
Affiliation(s)
- Rui Marçalo
- Department of Medical Sciences, Genome Medicine Laboratory, Institute of Biomedicine—iBiMED, University of Aveiro, Aveiro, Portugal
- Lab3R-Respiratory Research and Rehabilitation, School for Health Sciences (ESSUA) and Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Sonya Neto
- Department of Medical Sciences, Genome Medicine Laboratory, Institute of Biomedicine—iBiMED, University of Aveiro, Aveiro, Portugal
| | - Miguel Pinheiro
- Department of Medical Sciences, Genome Medicine Laboratory, Institute of Biomedicine—iBiMED, University of Aveiro, Aveiro, Portugal
| | - Ana J. Rodrigues
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho–Braga, Portugal
| | - Nuno Sousa
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho–Braga, Portugal
| | - Manuel A. S. Santos
- Department of Medical Sciences, Genome Medicine Laboratory, Institute of Biomedicine—iBiMED, University of Aveiro, Aveiro, Portugal
| | - Paula Simão
- Pulmonology Department, Unidade Local de Saúde de Matosinhos—Porto, Porto, Portugal
| | - Carla Valente
- Pulmonology Department, Centro Hospitalar do Baixo Vouga–Aveiro, Aveiro, Portugal
| | - Lília Andrade
- Pulmonology Department, Centro Hospitalar do Baixo Vouga–Aveiro, Aveiro, Portugal
| | - Alda Marques
- Lab3R-Respiratory Research and Rehabilitation, School for Health Sciences (ESSUA) and Institute of Biomedicine (iBiMED), University of Aveiro, Aveiro, Portugal
| | - Gabriela R. Moura
- Department of Medical Sciences, Genome Medicine Laboratory, Institute of Biomedicine—iBiMED, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
30
|
Chakraborty C, Sharma AR, Bhattacharya M, Lee SS. A Detailed Overview of Immune Escape, Antibody Escape, Partial Vaccine Escape of SARS-CoV-2 and Their Emerging Variants With Escape Mutations. Front Immunol 2022; 13:801522. [PMID: 35222380 PMCID: PMC8863680 DOI: 10.3389/fimmu.2022.801522] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/05/2022] [Indexed: 01/08/2023] Open
Abstract
The infective SARS-CoV-2 is more prone to immune escape. Presently, the significant variants of SARS-CoV-2 are emerging in due course of time with substantial mutations, having the immune escape property. Simultaneously, the vaccination drive against this virus is in progress worldwide. However, vaccine evasion has been noted by some of the newly emerging variants. Our review provides an overview of the emerging variants' immune escape and vaccine escape ability. We have illustrated a broad view related to viral evolution, variants, and immune escape ability. Subsequently, different immune escape approaches of SARS-CoV-2 have been discussed. Different innate immune escape strategies adopted by the SARS-CoV-2 has been discussed like, IFN-I production dysregulation, cytokines related immune escape, immune escape associated with dendritic cell function and macrophages, natural killer cells and neutrophils related immune escape, PRRs associated immune evasion, and NLRP3 inflammasome associated immune evasion. Simultaneously we have discussed the significant mutations related to emerging variants and immune escape, such as mutations in the RBD region (N439K, L452R, E484K, N501Y, K444R) and other parts (D614G, P681R) of the S-glycoprotein. Mutations in other locations such as NSP1, NSP3, NSP6, ORF3, and ORF8 have also been discussed. Finally, we have illustrated the emerging variants' partial vaccine (BioNTech/Pfizer mRNA/Oxford-AstraZeneca/BBIBP-CorV/ZF2001/Moderna mRNA/Johnson & Johnson vaccine) escape ability. This review will help gain in-depth knowledge related to immune escape, antibody escape, and partial vaccine escape ability of the virus and assist in controlling the current pandemic and prepare for the next.
Collapse
Affiliation(s)
- Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Kolkata, India
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging and Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| | | | - Sang-Soo Lee
- Institute for Skeletal Aging and Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, South Korea
| |
Collapse
|
31
|
Guo X, Gupta A, Sampat A, Zhai C. A stochastic contact network model for assessing outbreak risk of COVID-19 in workplaces. PLoS One 2022; 17:e0262316. [PMID: 35030206 PMCID: PMC8759694 DOI: 10.1371/journal.pone.0262316] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/20/2021] [Indexed: 12/19/2022] Open
Abstract
The COVID-19 pandemic has drastically shifted the way people work. While many businesses can operate remotely, a large number of jobs can only be performed on-site. Moreover as businesses create plans for bringing workers back on-site, they are in need of tools to assess the risk of COVID-19 for their employees in the workplaces. This study aims to fill the gap in risk modeling of COVID-19 outbreaks in facilities like offices and warehouses. We propose a simulation-based stochastic contact network model to assess the cumulative incidence in workplaces. First-generation cases are introduced as a Bernoulli random variable using the local daily new case rate as the success rate. Contact networks are established through randomly sampled daily contacts for each of the first-generation cases and successful transmissions are established based on a randomized secondary attack rate (SAR). Modification factors are provided for SAR based on changes in airflow, speaking volume, and speaking activity within a facility. Control measures such as mask wearing are incorporated through modifications in SAR. We validated the model by comparing the distribution of cumulative incidence in model simulations against real-world outbreaks in workplaces and nursing homes. The comparisons support the model's validity for estimating cumulative incidences for short forecasting periods of up to 15 days. We believe that the current study presents an effective tool for providing short-term forecasts of COVID-19 cases for workplaces and for quantifying the effectiveness of various control measures. The open source model code is made available at github.com/abhineetgupta/covid-workplace-risk.
Collapse
Affiliation(s)
- Xi Guo
- One Concern, Inc., Menlo Park, CA, United States of America
| | - Abhineet Gupta
- One Concern, Inc., Menlo Park, CA, United States of America
| | - Anand Sampat
- One Concern, Inc., Menlo Park, CA, United States of America
| | - Chengwei Zhai
- One Concern, Inc., Menlo Park, CA, United States of America
| |
Collapse
|
32
|
Aiewsakun P, Nilplub P, Wongtrakoongate P, Hongeng S, Thitithanyanont A. SARS-CoV-2 genetic variations associated with COVID-19 pathogenicity. Microb Genom 2021; 7. [PMID: 34870573 PMCID: PMC8767342 DOI: 10.1099/mgen.0.000734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
In this study, we performed genome-wide association analyses on SARS-CoV-2 genomes to identify genetic mutations associated with pre-symptomatic/asymptomatic COVID-19 cases. Various potential covariates and confounding factors of COVID-19 severity, including patient age, gender and country, as well as virus phylogenetic relatedness were adjusted for. In total, 3021 full-length genomes of SARS-CoV-2 generated from original clinical samples and whose patient status could be determined conclusively as either ‘pre-symptomatic/asymptomatic’ or ‘symptomatic’ were retrieved from the GISAID database. We found that the mutation 11 083G>T, located in the coding region of non-structural protein 6, is significantly associated with asymptomatic COVID-19. Patient age is positively correlated with symptomatic infection, while gender is not significantly correlated with the development of the disease. We also found that the effects of the mutation, patient age and gender do not vary significantly among countries, although each country appears to have varying baseline chances of COVID-19 symptom development.
Collapse
Affiliation(s)
- Pakorn Aiewsakun
- Department of Microbiology, Faculty of Science, Mahidol University, 272, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand.,Pornchai Matangkasombut Center for Microbial Genomics, Department of Microbiology, Faculty of Science, Mahidol University, 272, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Patrawee Nilplub
- Pornchai Matangkasombut Center for Microbial Genomics, Department of Microbiology, Faculty of Science, Mahidol University, 272, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Patompon Wongtrakoongate
- Department of Biochemistry, Faculty of Science, Mahidol University, 272, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand.,Center for Neuroscience, Faculty of Science, Mahidol University, 272, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Suradej Hongeng
- Division of Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Ramathibodi Hospital, Mahidol University, 270, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| | - Arunee Thitithanyanont
- Department of Microbiology, Faculty of Science, Mahidol University, 272, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand.,Pornchai Matangkasombut Center for Microbial Genomics, Department of Microbiology, Faculty of Science, Mahidol University, 272, Rama VI Road, Ratchathewi, Bangkok, 10400, Thailand
| |
Collapse
|
33
|
Wang S, Hao M, Pan Z, Lei J, Zou X. Data-driven multi-scale mathematical modeling of SARS-CoV-2 infection reveals heterogeneity among COVID-19 patients. PLoS Comput Biol 2021; 17:e1009587. [PMID: 34818337 PMCID: PMC8654229 DOI: 10.1371/journal.pcbi.1009587] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 12/08/2021] [Accepted: 10/25/2021] [Indexed: 12/17/2022] Open
Abstract
Patients with coronavirus disease 2019 (COVID-19) often exhibit diverse disease progressions associated with various infectious ability, symptoms, and clinical treatments. To systematically and thoroughly understand the heterogeneous progression of COVID-19, we developed a multi-scale computational model to quantitatively understand the heterogeneous progression of COVID-19 patients infected with severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2). The model consists of intracellular viral dynamics, multicellular infection process, and immune responses, and was formulated using a combination of differential equations and stochastic modeling. By integrating multi-source clinical data with model analysis, we quantified individual heterogeneity using two indexes, i.e., the ratio of infected cells and incubation period. Specifically, our simulations revealed that increasing the host antiviral state or virus induced type I interferon (IFN) production rate can prolong the incubation period and postpone the transition from asymptomatic to symptomatic outcomes. We further identified the threshold dynamics of T cell exhaustion in the transition between mild-moderate and severe symptoms, and that patients with severe symptoms exhibited a lack of naïve T cells at a late stage. In addition, we quantified the efficacy of treating COVID-19 patients and investigated the effects of various therapeutic strategies. Simulations results suggested that single antiviral therapy is sufficient for moderate patients, while combination therapies and prevention of T cell exhaustion are needed for severe patients. These results highlight the critical roles of IFN and T cell responses in regulating the stage transition during COVID-19 progression. Our study reveals a quantitative relationship underpinning the heterogeneity of transition stage during COVID-19 progression and can provide a potential guidance for personalized therapy in COVID-19 patients. Coronavirus disease 2019 (COVID-19) is currently destroying both lives and economies. However, patients infected with severe acute respiratory syndrome (SARS)-like coronavirus (SARS-CoV-2) usually present heterogeneous and complicated progressions, such as different incubation periods (short and long), symptoms (asymptomatic and symptomatic) and severity (mild-moderate and severe). Currently, various clinical data and experimental data are available from different countries, which has great significance for integrating different types of data to comprehensively understand the diverse disease progression in COVID-19 patients and guide individual treatment strategies. Here, we developed a multi-scale computational model to describe the dynamical process of patients infected with SARS-CoV-2, including intracellular viral dynamics, multicellular infection process, and immune responses. By combining data integration, stochastic simulation and quantitative analysis based on the multi-scale mathematical model, we addressed an important question regarding how IFN response and T cell exhaustion quantitatively affect heterogeneous progression in patients with respect to incubation periods, symptoms and severity. Furthermore, the efficacy of various therapeutic strategies for treating COVID-19 patients with different severity degrees was evaluated and validated. The computational framework in this study can also be extended to explore the dynamical process of other coronavirus infections.
Collapse
Affiliation(s)
- Shun Wang
- School of Mathematics and Statistics, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China
| | - Mengqian Hao
- School of Mathematics and Statistics, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China
| | - Zishu Pan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, China
| | - Jinzhi Lei
- School of Mathematical Sciences, Center for Applied Mathematics, Tiangong University, Tianjin, China
- * E-mail: (JL); (XZ)
| | - Xiufen Zou
- School of Mathematics and Statistics, Wuhan University, Wuhan, China
- Hubei Key Laboratory of Computational Science, Wuhan University, Wuhan, China
- * E-mail: (JL); (XZ)
| |
Collapse
|
34
|
A Global Mutational Profile of SARS-CoV-2: A Systematic Review and Meta-Analysis of 368,316 COVID-19 Patients. Life (Basel) 2021; 11:life11111224. [PMID: 34833100 PMCID: PMC8620851 DOI: 10.3390/life11111224] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 11/03/2021] [Accepted: 11/08/2021] [Indexed: 12/20/2022] Open
Abstract
Since its first detection in December 2019, more than 232 million cases of COVID-19, including 4.7 million deaths, have been reported by the WHO. The SARS-CoV-2 viral genomes have evolved rapidly worldwide, causing the emergence of new variants. This systematic review and meta-analysis was conducted to provide a global mutational profile of SARS-CoV-2 from December 2019 to October 2020. The review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA), and a study protocol was lodged with PROSPERO. Data from 62 eligible studies involving 368,316 SARS-CoV-2 genomes were analyzed. The mutational data analyzed showed most studies detected mutations in the Spike protein (n = 50), Nucleocapsid phosphoprotein (n = 34), ORF1ab gene (n = 29), 5′-UTR (n = 28) and ORF3a (n = 25). Under the random-effects model, pooled prevalence of SARS-CoV-2 variants was estimated at 95.1% (95% CI; 93.3–96.4%; I2 = 98.952%; p = 0.000) while subgroup meta-analysis by country showed majority of the studies were conducted ‘Worldwide’ (n = 10), followed by ‘Multiple countries’ (n = 6) and the USA (n = 5). The estimated prevalence indicated a need to continuously monitor the prevalence of new mutations due to their potential influence on disease severity, transmissibility and vaccine effectiveness.
Collapse
|
35
|
SARS-CoV-2 evolution in animals suggests mechanisms for rapid variant selection. Proc Natl Acad Sci U S A 2021; 118:2105253118. [PMID: 34716263 PMCID: PMC8612357 DOI: 10.1073/pnas.2105253118] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 09/15/2021] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 spillback from humans into domestic and wild animals has been well documented, and an accumulating number of studies illustrate that human-to-animal transmission is widespread in cats, mink, deer, and other species. Experimental inoculations of cats, mink, and ferrets have perpetuated transmission cycles. We sequenced full genomes of Vero cell-expanded SARS-CoV-2 inoculum and viruses recovered from cats (n = 6), dogs (n = 3), hamsters (n = 3), and a ferret (n = 1) following experimental exposure. Five nonsynonymous changes relative to the USA-WA1/2020 prototype strain were near fixation in the stock used for inoculation but had reverted to wild-type sequences at these sites in dogs, cats, and hamsters within 1- to 3-d postexposure. A total of 14 emergent variants (six in nonstructural genes, six in spike, and one each in orf8 and nucleocapsid) were detected in viruses recovered from animals. This included substitutions in spike residues H69, N501, and D614, which also vary in human lineages of concern. Even though a live virus was not cultured from dogs, substitutions in replicase genes were detected in amplified sequences. The rapid selection of SARS-CoV-2 variants in vitro and in vivo reveals residues with functional significance during host switching. These observations also illustrate the potential for spillback from animal hosts to accelerate the evolution of new viral lineages, findings of particular concern for dogs and cats living in households with COVID-19 patients. More generally, this glimpse into viral host switching reveals the unrealized rapidity and plasticity of viral evolution in experimental animal model systems.
Collapse
|
36
|
Jeewandara C, Jayathilaka D, Ranasinghe D, Hsu NS, Ariyaratne D, Jayadas TT, Panambara Arachchige DM, Lindsey BB, Gomes L, Parker MD, Wijewickrama A, Karunaratne M, Ogg GS, de Silva TI, Malavige GN. Genomic and Epidemiological Analysis of SARS-CoV-2 Viruses in Sri Lanka. Front Microbiol 2021; 12:722838. [PMID: 34603246 PMCID: PMC8483294 DOI: 10.3389/fmicb.2021.722838] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 08/24/2021] [Indexed: 11/13/2022] Open
Abstract
Background: In order to understand the molecular epidemiology of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) in Sri Lanka, since March 2020, we carried out genomic sequencing overlaid on available epidemiological data until April 2021. Methods: Whole genome sequencing was carried out on diagnostic sputum or nasopharyngeal swabs from 373 patients with COVID-19. Molecular clock phylogenetic analysis was undertaken to further explore dominant lineages. Results: The B.1.411 lineage was most prevalent, which was established in Sri Lanka and caused outbreaks throughout the country until March 2021. The estimated time of the most recent common ancestor (tMRCA) of this lineage was June 1, 2020 (with 95% lower and upper bounds March 30 to July 27) suggesting cryptic transmission may have occurred, prior to a large epidemic starting in October 2020. Returning travellers were identified with infections caused by lineage B.1.258, as well as the more transmissible B.1.1.7 lineage, which has replaced B.1.411 to fuel the ongoing large outbreak in the country. Conclusions: The large outbreak that started in early October, is due to spread of a single virus lineage, B.1.411 until the end of March 2021, when B.1.1.7 emerged and became the dominant lineage.
Collapse
Affiliation(s)
- Chandima Jeewandara
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Deshni Jayathilaka
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Diyanath Ranasinghe
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Nienyun Sharon Hsu
- Department of Infection, Immunity and Cardiovascular Diseases, Medical School, University of Sheffield, Sheffield, United Kingdom
- Sheffield Bioinformatics Core, The University of Sheffield, Sheffield, United Kingdom
| | - Dinuka Ariyaratne
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Tibutius Thanesh Jayadas
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | | | - Benjamin B. Lindsey
- Department of Infection, Immunity and Cardiovascular Diseases, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Laksiri Gomes
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
| | - Matthew D. Parker
- Sheffield Bioinformatics Core, The University of Sheffield, Sheffield, United Kingdom
| | | | | | - Graham S. Ogg
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| | - Thushan I. de Silva
- Department of Infection, Immunity and Cardiovascular Diseases, Medical School, University of Sheffield, Sheffield, United Kingdom
| | - Gathsaurie Neelika Malavige
- Allergy, Immunology and Cell Biology Unit, Department of Immunology and Molecular Medicine, University of Sri Jayewardenepura, Nugegoda, Sri Lanka
- MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
37
|
Rahman MM, Kader SB, Rizvi SS. Molecular characterization of SARS-CoV-2 from Bangladesh: implications in genetic diversity, possible origin of the virus, and functional significance of the mutations. Heliyon 2021; 7:e07866. [PMID: 34458642 PMCID: PMC8380069 DOI: 10.1016/j.heliyon.2021.e07866] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 01/16/2021] [Accepted: 08/19/2021] [Indexed: 12/23/2022] Open
Abstract
In a try to understand the pathogenesis, evolution and epidemiology of the SARS-CoV-2 virus, scientists from all over the world are tracking its genomic changes in real-time. Genomic studies can be helpful in understanding the disease dynamics. We have downloaded 324 complete and near complete SARS-CoV-2 genomes submitted in GISAID database from Bangladesh which were isolated between 30 March to 7 September, 2020. We then compared these genomes with Wuhan reference sequence and found 4160 mutation events including 2253 missense single nucleotide variations, 38 deletions and 10 insertions. The C>T nucleotide change was most prevalent (41% of all mutations) possibly due to selective mutation pressure to reduce CpG sites to evade CpG targeted host immune response. The most frequent mutation that occurred in 98% isolates was 3037C>T which is a synonymous change that usually accompanied 3 other mutations that include 241C>T, 14408C>T (P323L in RdRp) and 23403A>G (D614G in spike protein). The P323L was reported to increase mutation rate and D614G is associated with increased viral replication and currently most prevalent variant circulating all over the world. We identified multiple missense mutations in B-cell and T-cell predicted epitope regions and/or PCR target regions (including R203K and G204R that occurred in 86% of the isolates) that may impact immunogenicity and/or RT-PCR based diagnosis. Our analysis revealed 5 large deletion events in ORF7a and ORF8 gene products that may be associated with less severity of the disease and increased viral clearance. Our phylogeny analysis identified most of the isolates belonged to the Nextstrain clade 20B (86%) and GISAID clade GR (88%). Most of our isolates shared common ancestors either directly with European countries or jointly with middle eastern countries as well as Australia and India. Interestingly, the 19B clade (GISAID S clade) was unique to Chittagong, which was originally prevalent in China. This reveals possible multiple introductions of the virus in Bangladesh via different routes. Hence, more genome sequencing and analysis with related clinical data is needed to interpret functional significance and better predict the disease dynamics that may be helpful for policy makers to control the COVID-19 pandemic.
Collapse
Affiliation(s)
- Md. Marufur Rahman
- Centre for Medical Biotechnology, Management Information System, Directorate General of Health Services, Mohakhali, Dhaka, 1212, Bangladesh
| | | | - S.M. Shahriar Rizvi
- Communicable Disease Control, Directorate General of Health Services, Mohakhali, Dhaka, 1212, Bangladesh
| |
Collapse
|
38
|
COVID-19 vaccines and decreased transmission of SARS-CoV-2. Inflammopharmacology 2021; 29:1357-1360. [PMID: 34279767 PMCID: PMC8287551 DOI: 10.1007/s10787-021-00847-2] [Citation(s) in RCA: 77] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/08/2021] [Indexed: 12/23/2022]
Abstract
A massive COVID-19 vaccination campaign is underway worldwide. Epidemiological data from studies indicate excellent efficacy and safety profile for COVID-19 vaccines. However, there are few data from studies on the effect of decreasing the probability of infection of vaccinated subjects compared to unvaccinated subjects. In this short communication, we describe some evidence on this important and current topic providing useful personal reflections.
Collapse
|
39
|
Wang R, Chen J, Gao K, Wei GW. Vaccine-escape and fast-growing mutations in the United Kingdom, the United States, Singapore, Spain, India, and other COVID-19-devastated countries. Genomics 2021; 113:2158-2170. [PMID: 34004284 PMCID: PMC8123493 DOI: 10.1016/j.ygeno.2021.05.006] [Citation(s) in RCA: 134] [Impact Index Per Article: 44.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 04/30/2021] [Accepted: 05/10/2021] [Indexed: 01/04/2023]
Abstract
Recently, the SARS-CoV-2 variants from the United Kingdom (UK), South Africa, and Brazil have received much attention for their increased infectivity, potentially high virulence, and possible threats to existing vaccines and antibody therapies. The question remains if there are other more infectious variants transmitted around the world. We carry out a large-scale study of 506,768 SARS-CoV-2 genome isolates from patients to identify many other rapidly growing mutations on the spike (S) protein receptor-binding domain (RBD). We reveal that essentially all 100 most observed mutations strengthen the binding between the RBD and the host angiotensin-converting enzyme 2 (ACE2), indicating the virus evolves toward more infectious variants. In particular, we discover new fast-growing RBD mutations N439K, S477N, S477R, and N501T that also enhance the RBD and ACE2 binding. We further unveil that mutation N501Y involved in United Kingdom (UK), South Africa, and Brazil variants may moderately weaken the binding between the RBD and many known antibodies, while mutations E484K and K417N found in South Africa and Brazilian variants, L452R and E484Q found in India variants, can potentially disrupt the binding between the RBD and many known antibodies. Among these RBD mutations, L452R is also now known as part of the California variant B.1.427. Finally, we hypothesize that RBD mutations that can simultaneously make SARS-CoV-2 more infectious and disrupt the existing antibodies, called vaccine escape mutations, will pose an imminent threat to the current crop of vaccines. A list of most likely vaccine escape mutations is given, including S494P, Q493L, K417N, F490S, F486L, R403K, E484K, L452R, K417T, F490L, E484Q, and A475S. Mutation T478K appears to make the Mexico variant B.1.1.222 the most infectious one. Our comprehensive genetic analysis and protein-protein binding study show that the genetic evolution of SARS-CoV-2 on the RBD, which may be regulated by host gene editing, viral proofreading, random genetic drift, and natural selection, gives rise to more infectious variants that will potentially compromise existing vaccines and antibody therapies.
Collapse
Affiliation(s)
- Rui Wang
- Department of Mathematics, Michigan State University, MI 48824, USA
| | - Jiahui Chen
- Department of Mathematics, Michigan State University, MI 48824, USA
| | - Kaifu Gao
- Department of Mathematics, Michigan State University, MI 48824, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, MI 48824, USA; Department of Electrical and Computer Engineering, Michigan State University, MI 48824, USA; Department of Biochemistry and Molecular Biology, Michigan State University, MI 48824, USA.
| |
Collapse
|
40
|
Yeh TY, Contreras GP. Viral transmission and evolution dynamics of SARS-CoV-2 in shipboard quarantine. Bull World Health Organ 2021; 99:486-495. [PMID: 34248221 PMCID: PMC8243027 DOI: 10.2471/blt.20.255752] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 12/20/2020] [Accepted: 01/21/2021] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE To examine transmission and evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in shipboard quarantine of the Diamond Princess cruise ship. METHODS We obtained the full SARS-CoV-2 genome sequences of 28 samples from the Global Initiative on Sharing All Influenza Data database. The samples were collected between 10 and 25 February 2020 and came for individuals who had been tested for SARS-CoV-2 during the quarantine on the cruise ship. These samples were later sequenced in either Japan or the United States of America. We analysed evolution dynamics of SARS-CoV-2 using computational tools of phylogenetics, natural selection pressure and genetic linkage. FINDINGS The SARS-CoV-2 outbreak in the cruise most likely originated from either a single person infected with a virus variant identical to the WIV04 isolates, or simultaneously with another primary case infected with a virus containing the 11083G > T mutation. We identified a total of 24 new viral mutations across 64.2% (18/28) of samples, and the virus evolved into at least five subgroups. Increased positive selection of SARS-CoV-2 were statistically significant during the quarantine (Tajima's D: -2.03, P < 0.01; Fu and Li's D: -2.66, P < 0.01; and Zeng's E: -2.37, P < 0.01). Linkage disequilibrium analysis confirmed that ribonucleic acid (RNA) recombination with the11083G > T mutation also contributed to the increase of mutations among the viral progeny. CONCLUSION The findings indicate that the 11083G > T mutation of SARS-CoV-2 spread during shipboard quarantine and arose through de novo RNA recombination under positive selection pressure.
Collapse
Affiliation(s)
- Ting-Yu Yeh
- Auxergen Inc., Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, United States of America
| | - Gregory P Contreras
- Auxergen Inc., Columbus Center, 701 East Pratt Street, Baltimore, MD 21202, United States of America
| |
Collapse
|
41
|
Dao TL, Hoang VT, Colson P, Lagier JC, Million M, Raoult D, Levasseur A, Gautret P. SARS-CoV-2 Infectivity and Severity of COVID-19 According to SARS-CoV-2 Variants: Current Evidence. J Clin Med 2021; 10:2635. [PMID: 34203844 PMCID: PMC8232800 DOI: 10.3390/jcm10122635] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/04/2021] [Accepted: 06/14/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND We conducted this review to summarize the relation between viral mutation and infectivity of SARS-CoV-2 and also the severity of COVID-19 in vivo and in vitro. METHOD Articles were identified through a literature search until 31 May 2021, in PubMed, Web of Science and Google Scholar. RESULTS Sixty-three studies were included. To date, most studies showed that the viral mutations, especially the D614G variant, correlate with a higher infectivity than the wild-type virus. However, the evidence of the association between viral mutation and severity of the disease is scant. A SARS-CoV-2 variant with a 382-nucleotide deletion was associated with less severe infection in patients. The 11,083G > U mutation was significantly associated with asymptomatic patients. By contrast, ORF1ab 4715L and S protein 614G variants were significantly more frequent in patients from countries where high fatality rates were also reported. The current evidence showed that variants of concern have led to increased infectivity and deteriorating epidemiological situations. However, the relation between this variant and severity of COVID-19 infection was contradictory. CONCLUSION The COVID-19 pandemic continues to spread worldwide. It is necessary to anticipate large clinical cohorts to evaluate the virulence and transmissibility of SARS-CoV-2 mutants.
Collapse
Affiliation(s)
- Thi Loi Dao
- IRD, AP-HM, SSA, VITROME, Aix Marseille University, 13005 Marseille, France; (T.L.D.); (V.T.H.)
- IHU—Méditerranée Infection, Aix Marseille University, 13005 Marseille, France; (P.C.); (J.C.L.); (M.M.); (D.R.); (A.L.)
- Thai Binh University of Medicine and Pharmacy, Thai Binh 410000, Vietnam
| | - Van Thuan Hoang
- IRD, AP-HM, SSA, VITROME, Aix Marseille University, 13005 Marseille, France; (T.L.D.); (V.T.H.)
- IHU—Méditerranée Infection, Aix Marseille University, 13005 Marseille, France; (P.C.); (J.C.L.); (M.M.); (D.R.); (A.L.)
- Thai Binh University of Medicine and Pharmacy, Thai Binh 410000, Vietnam
| | - Philippe Colson
- IHU—Méditerranée Infection, Aix Marseille University, 13005 Marseille, France; (P.C.); (J.C.L.); (M.M.); (D.R.); (A.L.)
- IRD, AP-HM, MEPHI, Aix Marseille University, 13005 Marseille, France
| | - Jean Christophe Lagier
- IHU—Méditerranée Infection, Aix Marseille University, 13005 Marseille, France; (P.C.); (J.C.L.); (M.M.); (D.R.); (A.L.)
- IRD, AP-HM, MEPHI, Aix Marseille University, 13005 Marseille, France
| | - Matthieu Million
- IHU—Méditerranée Infection, Aix Marseille University, 13005 Marseille, France; (P.C.); (J.C.L.); (M.M.); (D.R.); (A.L.)
- IRD, AP-HM, MEPHI, Aix Marseille University, 13005 Marseille, France
| | - Didier Raoult
- IHU—Méditerranée Infection, Aix Marseille University, 13005 Marseille, France; (P.C.); (J.C.L.); (M.M.); (D.R.); (A.L.)
- IRD, AP-HM, MEPHI, Aix Marseille University, 13005 Marseille, France
| | - Anthony Levasseur
- IHU—Méditerranée Infection, Aix Marseille University, 13005 Marseille, France; (P.C.); (J.C.L.); (M.M.); (D.R.); (A.L.)
- IRD, AP-HM, MEPHI, Aix Marseille University, 13005 Marseille, France
| | - Philippe Gautret
- IRD, AP-HM, SSA, VITROME, Aix Marseille University, 13005 Marseille, France; (T.L.D.); (V.T.H.)
- IHU—Méditerranée Infection, Aix Marseille University, 13005 Marseille, France; (P.C.); (J.C.L.); (M.M.); (D.R.); (A.L.)
| |
Collapse
|
42
|
Patro LPP, Sathyaseelan C, Uttamrao PP, Rathinavelan T. The evolving proteome of SARS-CoV-2 predominantly uses mutation combination strategy for survival. Comput Struct Biotechnol J 2021; 19:3864-3875. [PMID: 34109017 PMCID: PMC8178965 DOI: 10.1016/j.csbj.2021.05.054] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/29/2021] [Accepted: 05/30/2021] [Indexed: 12/31/2022] Open
Abstract
The knowledge about SARS-CoV-2 proteome variations is important to understand its evolutionary tactics and in drug/vaccine design. An extensive analysis of 125,747 whole proteome reveals 7915 recurring mutations (involving 5146 positions) during December2019-November 2020. Among these, 10 and 51 are highly and moderately recurring mutations respectively. Ever since the pandemic outbreak, ∼50% new proteome variants evolve every month, resulting in 5 major clades. Intriguingly, ∼70% of the variants reported in January 2020 are due to the emergence of new mutations, which sharply declines to ∼ 40% in April 2020 and thenceforth, declines steadily till November 2020(∼10%). An exactly opposite trend is seen for variants evolved with cocktail of existing mutations: the lowest in January 2020(∼20%) and the highest in November 2020(80%). This leads to a steady increase in the average number of mutations per sequence. This indicates that the virus has reached the slow pace to accept new mutations. Instead, it uses a mutation combination strategy for survival.
Collapse
Affiliation(s)
| | | | | | - Thenmalarchelvi Rathinavelan
- Department of Biotechnology, Indian Institute of Technology Hyderabad, Kandi Campus, Telangana State 502285, India
| |
Collapse
|
43
|
López-Mendoza H, Montañés A, Moliner-Lahoz FJ. Disparities in the Evolution of the COVID-19 Pandemic between Spanish Provinces. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:5085. [PMID: 34064938 PMCID: PMC8151898 DOI: 10.3390/ijerph18105085] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 05/04/2021] [Accepted: 05/09/2021] [Indexed: 12/11/2022]
Abstract
Spain experienced a second wave of the COVID-19 pandemic in autumn 2020, which has been approached with different measures by regional authorities. We analyze the presence of convergence in the cumulative incidence for 14 days (CI14) in provinces and self-governing cities. The Phillips-Sul methodology was used to study the grouping of behavior between provinces, and an ordered logit model was estimated to understand the forces that drive creating the different convergence clubs. We reject the presence of a single pattern of behavior in the evolution of the CI14 across territories. Four statistically different convergence clubs and an additional province (Madrid) with divergent behavior are observed. Provinces with developed agricultural and industrial economic sectors, high mobility, and a high proportion of Central and South American immigrants had the highest level of CI14. We show that the transmission of the virus is not homogeneous in the Spanish national territory. Our results are helpful for identifying differences in determinants that could explain the pandemic's evolution and for formulating hypotheses about the effectiveness of implemented measures.
Collapse
Affiliation(s)
- Héctor López-Mendoza
- Directorate-General of Public Health, Aragon Department of Health, 50017 Zaragoza, Spain;
- Preventive Medicine and Public Health Department, Lozano Blesa University Hospital, 50009 Zaragoza, Spain;
| | - Antonio Montañés
- Economic Analysis Department, University of Zaragoza, 50005 Zaragoza, Spain
| | - F. Javier Moliner-Lahoz
- Preventive Medicine and Public Health Department, Lozano Blesa University Hospital, 50009 Zaragoza, Spain;
| |
Collapse
|
44
|
Majumdar P, Niyogi S. SARS-CoV-2 mutations: the biological trackway towards viral fitness. Epidemiol Infect 2021; 149:e110. [PMID: 33928885 PMCID: PMC8134885 DOI: 10.1017/s0950268821001060] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/26/2021] [Accepted: 04/27/2021] [Indexed: 01/10/2023] Open
Abstract
The outbreak of pneumonia-like respiratory disorder at China and its rapid transmission world-wide resulted in public health emergency, which brought lineage B betacoronaviridae SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2) into spotlight. The fairly high mutation rate, frequent recombination and interspecies transmission in betacoronaviridae are largely responsible for their temporal changes in infectivity and virulence. Investigation of global SARS-CoV-2 genotypes revealed considerable mutations in structural, non-structural, accessory proteins as well as untranslated regions. Among the various types of mutations, single-nucleotide substitutions are the predominant ones. In addition, insertion, deletion and frame-shift mutations are also reported, albeit at a lower frequency. Among the structural proteins, spike glycoprotein and nucleocapsid phosphoprotein accumulated a larger number of mutations whereas envelope and membrane proteins are mostly conserved. Spike protein and RNA-dependent RNA polymerase variants, D614G and P323L in combination became dominant world-wide. Divergent genetic variants created serious challenge towards the development of therapeutics and vaccines. This review will consolidate mutations in different SARS-CoV-2 proteins and their implications on viral fitness.
Collapse
Affiliation(s)
| | - Sougata Niyogi
- Dinabandhu Andrews Institute of Technology and Management, Block-S, 1/406A, Patuli, Kolkata, West Bengal700094, India
| |
Collapse
|
45
|
Mourmouris P, Tzelves L, Roidi C, Fotsali A. COVID-19 transmission: a rapid systematic review of current knowledge. Osong Public Health Res Perspect 2021; 12:54-63. [PMID: 33979995 PMCID: PMC8102883 DOI: 10.24171/j.phrp.2021.12.2.02] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 03/21/2021] [Indexed: 12/12/2022] Open
Abstract
Objective The objective of this study was to identify the potential and definite sources of transmission of coronavirus disease 2019 (COVID-19). Methods Due to time constraints and the acute nature of the pandemic, we searched only PubMed/Medline from inception until January 28, 2021. We analyzed the level of evidence and risk of bias in each category and made suggestions accordingly. Results The virus was traced from its potential origin via possible ways of transmission to the last host. Symptomatic human-to-human transmission remains the driver of the epidemic, but asymptomatic transmission can potentially contribute in a substantial manner. Feces and fomites have both been found to contain viable virus; even though transmission through these routes has not been documented, their contribution cannot be ruled out. Finally, transmission from pregnant women to their children has been found to be low (up to 3%). Conclusion Even though robust outcomes cannot be easily assessed, medical personnel must maintain awareness of the main routes of transmission (via droplets and aerosols from even asymptomatic patients). This is the first attempt to systematically review the existing knowledge to produce a paper with a potentially significant clinical impact.
Collapse
Affiliation(s)
- Panagiotis Mourmouris
- Second Department of Urology, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Lazaros Tzelves
- Second Department of Urology, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Christiana Roidi
- Second Department of Urology, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasia Fotsali
- Second Department of Urology, Athens Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
46
|
Banoun H. Evolution of SARS-CoV-2: Review of Mutations, Role of the Host Immune System. Nephron Clin Pract 2021; 145:392-403. [PMID: 33910211 PMCID: PMC8247830 DOI: 10.1159/000515417] [Citation(s) in RCA: 39] [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: 10/06/2020] [Accepted: 02/17/2021] [Indexed: 01/08/2023] Open
Abstract
Since the reporting of the first cases of coronavirus in China and the publication of the first sequence of SARS-CoV-2 in December 2019, the virus has undergone numerous mutations. In Europe, the spring outbreak (March-April) was followed by a drop in the number of cases and deaths. The disease may have evolved into a milder form. The increase in PCR-positive cases in late summer 2020 did not lead to the expected increase in hospitalizations, ICU admissions, and deaths, based on the severity of the disease in the spring. This difference in disease severity could be due to factors independent of the virus or to the evolution of the virus. This review attempts to identify the mutations that have appeared since the beginning of the pandemic and their role in the temporal evolution of the pandemic. There are a cell and humoral type cross-reactivity in a large part of the population to common cold coronaviruses (HCoVs) and SARS-CoV-2. Evolutionarily important mutations and deletions have emerged in the SARS-CoV-2 genes encoding proteins that interact with the host immune system. In addition, one of the major mutations (in viral polymerase) is logically associated with a higher frequency of mutations throughout the genome. This frequency fluctuates over time and shows a peak at the time when the epidemic was most active. The rate of mutations in proteins involved in the relationship to the immune system continues to increase after the first outbreak. The cross-reactivity on the 1 hand and the viral mutations observed on the other hand could explain the evolution of the pandemic until the summer of 2020, partly due to the evolution of the virus in relation to the host immune system. The immunization campaign began in December 2020: concerns are emerging about a possible escape of the circulating variants vaccines in early 2021. These variants could also escape immunity acquired through infection with the 2020 strains.
Collapse
Affiliation(s)
- Helene Banoun
- Independent researcher, Former research fellow at INSERM (French Institute for Health and Medical Research), Marseille, France
| |
Collapse
|
47
|
Esper FP, Cheng YW, Adhikari TM, Tu ZJ, Li D, Li EA, Farkas DH, Procop GW, Ko JS, Chan TA, Jehi L, Rubin BP, Li J. Genomic Epidemiology of SARS-CoV-2 Infection During the Initial Pandemic Wave and Association With Disease Severity. JAMA Netw Open 2021; 4:e217746. [PMID: 33900399 PMCID: PMC8076962 DOI: 10.1001/jamanetworkopen.2021.7746] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 03/07/2021] [Indexed: 12/17/2022] Open
Abstract
Importance Understanding of SARS-CoV-2 variants that alter disease outcomes are important for clinical risk stratification and may provide important clues to the complex virus-host relationship. Objective To examine the association of identified SARS-CoV-2 variants, virus clades, and clade groups with disease severity and patient outcomes. Design, Setting, and Participants In this cross-sectional study, viral genome analysis of clinical specimens obtained from patients at the Cleveland Clinic infected with SARS-CoV-2 during the initial wave of infection (March 11 to April 22, 2020) was performed. Identified variants were matched with clinical outcomes. Data analysis was performed from April to July 2020. Main Outcomes and Measures Hospitalization, intensive care unit (ICU) admission, mortality, and laboratory outcomes were matched with SARS-CoV-2 variants. Results Specimens sent for viral genome sequencing originated from 302 patients with SARS-CoV-2 infection (median [interquartile range] age, 52.6 [22.8 to 82.5] years), of whom 126 (41.7%) were male, 195 (64.6%) were White, 91 (30.1%) required hospitalization, 35 (11.6%) needed ICU admission, and 17 (5.6%) died. From these specimens, 2531 variants (484 of which were unique) were identified. Six different SARS-CoV-2 clades initially circulated followed by a rapid reduction in clade diversity. Several variants were associated with lower hospitalization rate, and those containing 23403A>G (D614G Spike) were associated with increased survival when the patient was hospitalized (64 of 74 patients [86.5%] vs 10 of 17 patients [58.8%]; χ21 = 6.907; P = .009). Hospitalization and ICU admission were similar regardless of clade. Infection with Clade V variants demonstrated higher creatinine levels (median [interquartile range], 2.6 [-0.4 to 5.5] mg/dL vs 1.0 [0.2 to 2.2] mg/dL; mean creatinine difference, 2.9 mg/dL [95% CI, 0.8 to 5.0 mg/dL]; Kruskal-Wallis P = .005) and higher overall mortality rates (3 of 14 patients [21.4%] vs 17 of 302 patients [5.6%]; χ21 = 5.640; P = .02) compared with other variants. Infection by strains lacking the 23403A>G variant showed higher mortality in multivariable analysis (odds ratio [OR], 22.4; 95% CI, 0.6 to 5.6; P = .01). Increased variants of open reading frame (ORF) 3a were associated with decreased hospitalization frequency (OR, 0.4; 95% CI, 0.2 to 0.96; P = .04), whereas increased variants of Spike (OR, 0.01; 95% CI, <0.01 to 0.3; P = .01) and ORF8 (OR, 0.03; 95% CI, <0.01 to 0.6; P = .03) were associated with increased survival. Conclusions and Relevance Within weeks of SARS-CoV-2 circulation, a profound shift toward 23403A>G (D614G) specific genotypes occurred. Replaced clades were associated with worse clinical outcomes, including mortality. These findings help explain persistent hospitalization yet decreasing mortality as the pandemic progresses. SARS-CoV-2 clade assignment is an important factor that may aid in estimating patient outcomes.
Collapse
Affiliation(s)
- Frank P. Esper
- Center for Pediatric Infectious Disease, Cleveland Clinic Children’s, Cleveland, Ohio
| | - Yu-Wei Cheng
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Thamali M. Adhikari
- Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Zheng Jin Tu
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Derek Li
- Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Erik A. Li
- Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, Ohio
| | - Daniel H. Farkas
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Gary W. Procop
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jennifer S. Ko
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Timothy A. Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Center, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Lara Jehi
- Neurological Institute, Chief Research Information Office, Cleveland Clinic, Cleveland, Ohio
| | - Brian P. Rubin
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jing Li
- Department of Computer and Data Sciences, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
48
|
Hozumi Y, Wang R, Yin C, Wei GW. UMAP-assisted K-means clustering of large-scale SARS-CoV-2 mutation datasets. Comput Biol Med 2021; 131:104264. [PMID: 33647832 PMCID: PMC7897976 DOI: 10.1016/j.compbiomed.2021.104264] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/05/2021] [Accepted: 02/06/2021] [Indexed: 12/16/2022]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has a worldwide devastating effect. Understanding the evolution and transmission of SARS-CoV-2 is of paramount importance for controlling, combating and preventing COVID-19. Due to the rapid growth in both the number of SARS-CoV-2 genome sequences and the number of unique mutations, the phylogenetic analysis of SARS-CoV-2 genome isolates faces an emergent large-data challenge. We introduce a dimension-reduced K-means clustering strategy to tackle this challenge. We examine the performance and effectiveness of three dimension-reduction algorithms: principal component analysis (PCA), t-distributed stochastic neighbor embedding (t-SNE), and uniform manifold approximation and projection (UMAP). By using four benchmark datasets, we found that UMAP is the best-suited technique due to its stable, reliable, and efficient performance, its ability to improve clustering accuracy, especially for large Jaccard distanced-based datasets, and its superior clustering visualization. The UMAP-assisted K-means clustering enables us to shed light on increasingly large datasets from SARS-CoV-2 genome isolates.
Collapse
Affiliation(s)
- Yuta Hozumi
- Department of Mathematics, Michigan State University, MI, 48824, USA
| | - Rui Wang
- Department of Mathematics, Michigan State University, MI, 48824, USA
| | - Changchuan Yin
- Department of Mathematics, Statistics, and Computer Science, University of Illinois at Chicago, Chicago, IL, 60607, USA
| | - Guo-Wei Wei
- Department of Mathematics, Michigan State University, MI, 48824, USA; Department of Electrical and Computer Engineering, Michigan State University, MI, 48824, USA; Department of Biochemistry and Molecular Biology, Michigan State University, MI, 48824, USA.
| |
Collapse
|
49
|
Khan MT, Ali S, Khan AS, Muhammad N, Khalil F, Ishfaq M, Irfan M, Al-Sehemi AG, Muhammad S, Malik A, Khan TA, Wei DQ. SARS-CoV-2 Genome from the Khyber Pakhtunkhwa Province of Pakistan. ACS OMEGA 2021; 6:6588-6599. [PMID: 33748571 PMCID: PMC7944396 DOI: 10.1021/acsomega.0c05163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 01/05/2021] [Indexed: 05/08/2023]
Abstract
Among viral outbreaks, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is one of the deadliest ones, and it has triggered the global COVID-19 pandemic. In Pakistan, until 5th September 2020, a total of 6342 deaths have been reported, of which 1255 were from the Khyber Pakhtunkhwa (KPK) province. To understand the disease progression and control and also to produce vaccines and therapeutic efforts, whole genome sequence analysis is important. In the current investigation, we sequenced a single sample of SARS-CoV-2 genomes (accession no. MT879619) from a male suspect from Peshawar, the KPK capital city, during the first wave of infection. The local SARS-CoV-2 strain shows some unique characteristics compared to neighboring Iranian and Chinese isolates in phylogenetic tree and mutations. The circulating strains of SARS-CoV-2 represent an intermediate evolution from China and Iran. Furthermore, eight complete whole genome sequences, including the current Pakistani isolates which have been submitted to Global Initiative on Sharing All Influenza Data (GSAID), were also investigated for specific mutations and characters. Some novel mutations [NSP2 (D268del), NSP5 (N228K), and NS3 (F105S)] and specific characters have been detected in the coding regions, which may affect viral transmission, epidemiology, and disease severity. The computational modeling revealed that a majority of these mutations may have a stabilizing effect on the viral protein structure. In conclusion, the genome sequencing of local strains is important for better understanding the pathogenicity, immunogenicity, and epidemiology of causative agents.
Collapse
Affiliation(s)
- Muhammad Tahir Khan
- Institute of Molecular
Biology and Biotechnology (IMBB), The University
of Lahore, KM Defence Road, Lahore 58810, Pakistan
- State Key Laboratory of Microbial Metabolism,
Shanghai−Islamabad−Belgrade Joint Innovation Center
on Antibacterial Resistances, Joint International Research Laboratory
of Metabolic & Developmental Sciences and School of Life Sciences
and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Peng Cheng Laboratory, Vanke Cloud City Phase I Building 8, Xili Street, Nashan District, Shenzhen, Guangdong 518055, P. R. China
| | - Sajid Ali
- Department of Microbiology, Quaid-i-Azam University Islamabad, Islamabad 45320, Pakistan
| | - Anwar Sheed Khan
- Department of Microbiology, Kohat University of Science and Technology, Bannu Road, Near Jarma Bridge, Kohat 26000, Pakistan
| | - Noor Muhammad
- Department of Microbiology, Kohat University of Science and Technology, Bannu Road, Near Jarma Bridge, Kohat 26000, Pakistan
| | - Faiza Khalil
- Department of Biochemistry, Khyber Medical
College, Peshawar 25160, Pakistan
- University
of Peshawar, Road No.
2, Rahat Abad, Peshawar 25120, Khyber Pakhtunkhwa, Pakistan
| | - Muhammad Ishfaq
- Centre for Omic Sciences, Islamia
College Peshawar. Grand Trunk Road, Rahat Abad, Peshawar 25120, Pakistan
| | - Muhammad Irfan
- Department
of Oral Biology, College of Dentistry, University
of Florida, Gainesville, Florida 32611, United States
| | - Abdullah G. Al-Sehemi
- Research Center for Advanced Materials
Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
- Department of Chemistry, College of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Shabbir Muhammad
- Research Center for Advanced Materials
Science (RCAMS), King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
- Department of Chemistry, College of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
- State Key Laboratory of Microbial Metabolism,
Shanghai−Islamabad−Belgrade Joint Innovation Center
on Antibacterial Resistances, Joint International Research Laboratory
of Metabolic & Developmental Sciences and School of Life Sciences
and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Peng Cheng Laboratory, Vanke Cloud City Phase I Building 8, Xili Street, Nashan District, Shenzhen, Guangdong 518055, P. R. China
| | - Arif Malik
- Institute of Molecular
Biology and Biotechnology (IMBB), The University
of Lahore, KM Defence Road, Lahore 58810, Pakistan
| | - Taj Ali Khan
- Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Phase V, Hayatabad, Peshawar, Khyber Pakhtunkhwa 25000, Pakistan
| | - Dong Qing Wei
- State Key Laboratory of Microbial Metabolism,
Shanghai−Islamabad−Belgrade Joint Innovation Center
on Antibacterial Resistances, Joint International Research Laboratory
of Metabolic & Developmental Sciences and School of Life Sciences
and Biotechnology, Shanghai Jiao Tong University, Shanghai 200030, P. R. China
- Peng Cheng Laboratory, Vanke Cloud City Phase I Building 8, Xili Street, Nashan District, Shenzhen, Guangdong 518055, P. R. China
| |
Collapse
|
50
|
Hong LX, Liu L, Lin A, Yan WH. Risk factors for SARS-CoV-2 re-positivity in COVID-19 patients after discharge. Int Immunopharmacol 2021; 95:107579. [PMID: 33756229 PMCID: PMC7953440 DOI: 10.1016/j.intimp.2021.107579] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 02/24/2021] [Accepted: 03/08/2021] [Indexed: 12/19/2022]
Abstract
Objective Re-positivity of SARS-CoV-2 in discharged COVID-19 patients have been reported; however, early risk factors for SARS-CoV-2 re-positivity evaluation are limited. Methods This is a prospective study, a total of 145 COVID-19 patients were treated and all discharged according to the guideline criteria by Mar 11th 2020. After discharge, clinical visits and viral RT-PCR tests by the second and fourth week follow-up were carried-out. Patient demographic and clinical characteristics and laboratory data on admission and discharge were retrieved, and predictive factors for SARS-CoV-2 re-positivity were analyzed. Results 13 out of 145 (9.0%) COVID-19 patients were confirmed re-positivity of SARS-CoV-2 by RT-PCR test. The median interval between disease onset to recurrence was 38 days. SARS-CoV-2 re-positive cases were of significantly longer virus shedding duration, notably higher body temperature, heart rate and lower TNF-α and IgG levels on admission. Covariate logistic regression analysis revealed virus shedding duration and IgG levels are independent risk factors for SARS-CoV-2 return positive after discharge. Conclusion Longer viral shedding duration and lower IgG levels are risk factors for re-positivity of SARS-CoV-2 for discharged COVID-19 patients.
Collapse
Affiliation(s)
- Lu-Xiao Hong
- Department of Infectious Diseases, Taizhou Hospital of Zhejiang Province, Luqiao, Zhejiang 318050, China
| | - Lian Liu
- Department of Pediatrics, Taizhou Hospital of Zhejiang Province, Luqiao, Zhejiang 318050, China
| | - Aifen Lin
- Biological Resource Center, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, China; Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang, China
| | - Wei-Hua Yan
- Medical Research Center, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang 317000, China; Key Laboratory of Minimally Invasive Techniques & Rapid Rehabilitation of Digestive System Tumor of Zhejiang Province, Taizhou Hospital of Zhejiang Province, Linhai, Zhejiang, China.
| |
Collapse
|