1
|
Khachab Y, Saab A, El Morr C, El-Lahib Y, Sokhn ES. Identifying the panorama of potential pandemic pathogens and their key characteristics: a systematic scoping review. Crit Rev Microbiol 2025; 51:348-368. [PMID: 38900695 DOI: 10.1080/1040841x.2024.2360407] [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: 11/24/2023] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 06/22/2024]
Abstract
The globe has recently seen several terrifying pandemics and outbreaks, underlining the ongoing danger presented by infectious microorganisms. This literature review aims to explore the wide range of infections that have the potential to lead to pandemics in the present and the future and pave the way to the conception of epidemic early warning systems. A systematic review was carried out to identify and compile data on infectious agents known to cause pandemics and those that pose future concerns. One hundred and fifteen articles were included in the review. They provided insights on 25 pathogens that could start or contribute to creating pandemic situations. Diagnostic procedures, clinical symptoms, and infection transmission routes were analyzed for each of these pathogens. Each infectious agent's potential is discussed, shedding light on the crucial aspects that render them potential threats to the future. This literature review provides insights for policymakers, healthcare professionals, and researchers in their quest to identify potential pandemic pathogens, and in their efforts to enhance pandemic preparedness through building early warning systems for continuous epidemiological monitoring.
Collapse
Affiliation(s)
- Yara Khachab
- Laboratory Department, Lebanese Hospital Geitaoui-University Medical Center, Beirut, Lebanon
| | - Antoine Saab
- Quality and Safety Department, Lebanese Hospital Geitaoui-UMC, Beirut, Lebanon
| | - Christo El Morr
- School of Health Policy and Management, York University, Toronto, Canada
| | - Yahya El-Lahib
- Faculty of Social Work, University of Calgary, Calgary, Canada
| | - Elie Salem Sokhn
- Laboratory Department, Lebanese Hospital Geitaoui-University Medical Center, Beirut, Lebanon
- Molecular Testing Laboratory, Medical Laboratory Department, Faculty of Health Sciences, Beirut Arab University, Beirut, Lebanon
| |
Collapse
|
2
|
Mahmood J, Ul Haque MI, Gul M, Ayub A, Ansari FA, Ahmad W. Early Identification of Severe COVID-19 Cases and the Need for ICU Care Based on Clinical and Laboratory Risk Factors. Cureus 2025; 17:e80611. [PMID: 40230780 PMCID: PMC11995811 DOI: 10.7759/cureus.80611] [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] [Accepted: 03/12/2025] [Indexed: 04/16/2025] Open
Abstract
Background and objective Treatment in ICUs became extremely difficult due to the growing number of coronavirus disease 2019 (COVID-19) patients at the height of the pandemic. Consequently, prompt patient triage depends on the early categorization of severe cases in such scenarios. This study aimed to provide an evidence-based strategy to ensure the best use of resources by triaging patients based on objective risk factors. Methods This retrospective observational study comprised 500 inpatients (>age 18 years) who were hospitalized between March 20 and April 19, 2020, at the Khyber Teaching Hospital (KTH) and Hayatabad Medical Complex (HMC) in Peshawar, Pakistan. The clinical, laboratory, and radiological parameters were assessed. Real-time polymerase chain reaction (RT-PCR) findings were used to confirm the diagnosis of COVID-19. Results A total of 19 potential clinical and laboratory risk factors associated with ICU admissions were identified. At least one comorbidity among chronic lung disease, cardiovascular disease (CVD), and diabetes was the factor with the strongest association with ICU admission with a univariable odds ratio (OR) of over 27, followed by renal disease and other COVID-19 sequelae such as diarrhea, respiratory rate (>24 breaths/minute), and positive RT-PCR (vs. negative) with an univariable OR between 9 and 15. Furthermore, a multivariate logistic regression model was further developed with five risk factors, including comorbidity, presence of chronic lung disease, presence of diabetes, and RT-PCR (positive vs. negative), male sex (vs. female), and older age (65.0-80.5 years), suggesting a good fit of the model to the data shown by the area under the receiver operator characteristic curve (AUC) of 0.943 (95% CI: 0.917, 0.969). Additionally, a chest CT scan showed the typical COVID-19 pneumonia with pulmonary involvement of 30-40%, which was further evaluated by the COVID-19 Reporting and Data System (CO-RADS). The typical COVID-19 pneumonia was on a scale of four (15/25) or five (19/25) lung lesions. Conclusions Based on our findings, this approach could be used to screen the severe cases of COVID-19 patients and help them to be treated in ICUs on time while preventing others from unnecessarily using ICUs in the setting of limited medical resources, such as the outbreak of a pandemic.
Collapse
Affiliation(s)
- Jawad Mahmood
- Gastroenterology and Hepatology, Hayatabad Medical Complex, Peshawar, PAK
| | - Muhammad Izhar Ul Haque
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, University of Georgia, Athens, USA
| | - Maria Gul
- Gynecology, Ayub Teaching Hospital, Abbottabad, PAK
| | - Aliya Ayub
- Epidemiology and Public Health, Institute of Public Health and Social Sciences, Khyber Medical University, Peshawar, PAK
| | - Fawwad A Ansari
- Internal Medicine, Piedmont Athens Regional Medical Center, Athens, USA
| | - Wiqas Ahmad
- Gastroenterology and Hepatology, Hayatabad Medical Complex, Peshawar, PAK
| |
Collapse
|
3
|
Gharaibeh A, Gharaibeh MA, Bataineh S, Kecerová AM. Exploring the Spatial and Temporal Patterns of Children and Adolescents with COVID-19 Infections in Slovakia during March 2020 to July 2022. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:931. [PMID: 38929548 PMCID: PMC11205471 DOI: 10.3390/medicina60060931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/11/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024]
Abstract
Background and Objectives: The COVID-19 pandemic has had a significant global impact, necessitating a comprehensive understanding of its spatiotemporal patterns. The objective of this study is to explore the spatial and temporal patterns of COVID-19 infections among five age groups (<1, 1-4, 5-9, 10-14, and 15-19 years) in 72 districts of Slovakia on a quarterly basis from March 2020 to July 2022. Material and Methods: During the study period, a total of 393,429 confirmed PCR cases of COVID-19 or positive antigen tests were recorded across all studied age groups. The analysis examined the spatiotemporal spread of COVID infections per quarter, from September 2021 to May 2022. Additionally, data on hospitalizations, intensive care unit (ICU) admissions, pulmonary ventilation (PV), and death cases were analyzed. Results: The highest number of COVID-19 infections occurred between September 2021 and May 2022, particularly in the 10-14-year-old group (68,695 cases), followed by the 15-19-year-old group (62,232 cases), while the lowest incidence was observed in the <1-year-old group (1235 cases). Out of the total confirmed PCR cases, 18,886 individuals required hospitalization, 456 needed ICU admission, 402 received pulmonary ventilation, and only 16 died. The analysis of total daily confirmed PCR cases for all regions showed two major peaks on 12 December 2021 (6114 cases) and 1 February 2022 (3889 cases). Spatial mapping revealed that during December 2021 to February 2022, the highest number of infections in all age groups were concentrated mainly in Bratislava. Moreover, temporal trends of infections within each age group, considering monthly and yearly variations, exhibited distinct spatial patterns, indicating localized outbreaks in specific regions. Conclusions: The spatial and temporal patterns of COVID-19 infections among different age groups in Slovakia showed a higher number of infections in the 10-14-year-old age group, mainly occurring in urban districts. The temporal pattern of the spread of the virus to neighboring urban and rural districts reflected the movement of infected individuals. Hospitalizations, ICU and PV admissions, and deaths were relatively low. The study highlights the need for more proactive measures to contain outbreaks promptly and ensure the resilience of healthcare systems against future pandemics.
Collapse
Affiliation(s)
- Ahmad Gharaibeh
- Teaching Department of Orthopaedics Musculoskeletal Trauma, Faculty of Medicine, University Hospital of Louise Pasteur, Pavel Jozef Safarik University, 040 11 Košice, Slovakia
| | - Mamoun A. Gharaibeh
- Department of Natural Resources and the Environment, Faculty of Agriculture, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Siham Bataineh
- Department of Civil Engineering, Faculty of Engineering, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan;
| | | |
Collapse
|
4
|
Nagasawa N, Kimura R, Akagawa M, Shirai T, Sada M, Okayama K, Sato-Fujimoto Y, Saito M, Kondo M, Katayama K, Ryo A, Kuroda M, Kimura H. Molecular Evolutionary Analyses of the Spike Protein Gene and Spike Protein in the SARS-CoV-2 Omicron Subvariants. Microorganisms 2023; 11:2336. [PMID: 37764181 PMCID: PMC10537508 DOI: 10.3390/microorganisms11092336] [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: 07/28/2023] [Revised: 09/15/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023] Open
Abstract
To better understand the evolution of the SARS-CoV-2 Omicron subvariants, we performed molecular evolutionary analyses of the spike (S) protein gene/S protein using advanced bioinformatics technologies. First, time-scaled phylogenetic analysis estimated that a common ancestor of the Wuhan, Alpha, Beta, Delta variants, and Omicron variants/subvariants diverged in May 2020. After that, a common ancestor of the Omicron variant generated various Omicron subvariants over one year. Furthermore, a chimeric virus between the BM.1.1.1 and BJ.1 subvariants, known as XBB, diverged in July 2021, leading to the emergence of the prevalent subvariants XBB.1.5 and XBB.1.16. Next, similarity plot (SimPlot) data estimated that the recombination point (breakpoint) corresponded to nucleotide position 1373. As a result, XBB.1.5 subvariants had the 5' nucleotide side from the breakpoint as a strain with a BJ.1 sequence and the 3' nucleotide side as a strain with a BM.1.1.1 sequence. Genome network data showed that Omicron subvariants were genetically linked with the common ancestors of the Wuhan and Delta variants, resulting in many amino acid mutations. Selective pressure analysis estimated that the prevalent subvariants, XBB.1.5 and XBB.1.16, had specific amino acid mutations, such as V445P, G446S, N460K, and F486P, located in the RBD when compared with the BA.4 and BA.5 subvariants. Moreover, some representative immunogenicity-associated amino acid mutations, including L452R, F486V, R493Q, and V490S, were also found in these subvariants. These substitutions were involved in the conformational epitopes, implying that these mutations affect immunogenicity and vaccine evasion. Furthermore, these mutations were identified as positive selection sites. These results suggest that the S gene/S protein Omicron subvariants rapidly evolved, and mutations observed in the conformational epitopes may reduce the effectiveness of the current vaccine, including bivalent vaccines such as mRNA vaccines containing the BA.4/BA.5 subvariants.
Collapse
Affiliation(s)
- Norika Nagasawa
- Department of Health Science, Gunma Paz University Graduate School of Health Sciences, 1-7-1, Tonya-machi, Takasaki-shi 370-0006, Gunma, Japan; (N.N.); (K.O.)
- Department of Medical Technology, Gunma Paz University School of Medical Science and Technology, 1-7-1, Tonya-machi, Takasaki-shi 370-0006, Gunma, Japan;
| | - Ryusuke Kimura
- Advanced Medical Science Research Center, Gunma Paz University Research Institute, 1338-4, Shibukawa, Shibukawa-shi 377-0008, Gunma, Japan; (R.K.); (T.S.)
- Department of Bacteriology, Gunma University Graduate School of Medicine, Maebashi-shi 371-8514, Gunma, Japan
| | - Mao Akagawa
- Department of Clinical Laboratory, Juntendo University Hospital, Bunkyo-ku, Tokyo 113-8431, Japan;
| | - Tatsuya Shirai
- Advanced Medical Science Research Center, Gunma Paz University Research Institute, 1338-4, Shibukawa, Shibukawa-shi 377-0008, Gunma, Japan; (R.K.); (T.S.)
| | - Mitsuru Sada
- Department of Respiratory Medicine, Kyourin University School of Medicine, 6-20-2, Shinkawa, Mitaka-shi 181-8611, Tokyo, Japan;
| | - Kaori Okayama
- Department of Health Science, Gunma Paz University Graduate School of Health Sciences, 1-7-1, Tonya-machi, Takasaki-shi 370-0006, Gunma, Japan; (N.N.); (K.O.)
| | - Yuka Sato-Fujimoto
- Department of Medical Technology, Gunma Paz University School of Medical Science and Technology, 1-7-1, Tonya-machi, Takasaki-shi 370-0006, Gunma, Japan;
| | - Makoto Saito
- Department of Clinical Engineering, Gunma Paz University School of Medical Science and Technology, Takasaki-shi 370-0006, Gunma, Japan; (M.S.); (M.K.)
| | - Mayumi Kondo
- Department of Clinical Engineering, Gunma Paz University School of Medical Science and Technology, Takasaki-shi 370-0006, Gunma, Japan; (M.S.); (M.K.)
| | - Kazuhiko Katayama
- Laboratory of Viral Infection Control, Ōmura Satoshi Memorial Institute, Graduate School of Infection Control Sciences, Kitasato University, 5-9-1, Shirogane, Minato-ku, Tokyo 108-8641, Japan;
| | - Akihide Ryo
- Department of Virology III, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashimurayama-shi 208-0011, Tokyo, Japan;
| | - Makoto Kuroda
- Pathogen Genomics Center, National Institute of Infectious Diseases, 1-23-1, Toyama, Shinjuku-ku, Tokyo 162-8640, Japan;
| | - Hirokazu Kimura
- Department of Health Science, Gunma Paz University Graduate School of Health Sciences, 1-7-1, Tonya-machi, Takasaki-shi 370-0006, Gunma, Japan; (N.N.); (K.O.)
- Advanced Medical Science Research Center, Gunma Paz University Research Institute, 1338-4, Shibukawa, Shibukawa-shi 377-0008, Gunma, Japan; (R.K.); (T.S.)
- Department of Clinical Engineering, Gunma Paz University School of Medical Science and Technology, Takasaki-shi 370-0006, Gunma, Japan; (M.S.); (M.K.)
| |
Collapse
|
5
|
Bukhari AR, Ashraf J, Kanji A, Rahman YA, Trovão NS, Thielen PM, Yameen M, Kanwar S, Khan W, Kabir F, Nisar MI, Merritt B, Hasan R, Spiro D, Rasmussen Z, Aamir UB, Hasan Z. Sequential viral introductions and spread of BA.1 across Pakistan provinces during the Omicron wave. BMC Genomics 2023; 24:432. [PMID: 37532989 PMCID: PMC10399012 DOI: 10.1186/s12864-023-09539-3] [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: 04/30/2023] [Accepted: 07/27/2023] [Indexed: 08/04/2023] Open
Abstract
BACKGROUND COVID-19 waves caused by specific SARS-CoV-2 variants have occurred globally at different times. We focused on Omicron variants to understand the genomic diversity and phylogenetic relatedness of SARS-CoV-2 strains in various regions of Pakistan. METHODS We studied 276,525 COVID-19 cases and 1,031 genomes sequenced from December 2021 to August 2022. Sequences were analyzed and visualized using phylogenetic trees. RESULTS The highest case numbers and deaths were recorded in Sindh and Punjab, the most populous provinces in Pakistan. Omicron variants comprised 93% of all genomes, with BA.2 (32.6%) and BA.5 (38.4%) predominating. The first Omicron wave was associated with the sequential identification of BA.1 in Sindh, then Islamabad Capital Territory, Punjab, Khyber Pakhtunkhwa (KP), Azad Jammu Kashmir (AJK), Gilgit-Baltistan (GB) and Balochistan. Phylogenetic analysis revealed Sindh to be the source of BA.1 and BA.2 introductions into Punjab and Balochistan during early 2022. BA.4 was first introduced in AJK and BA.5 in Punjab. Most recent common ancestor (MRCA) analysis revealed relatedness between the earliest BA.1 genome from Sindh with Balochistan, AJK, Punjab and ICT, and that of first BA.1 from Punjab with strains from KPK and GB. CONCLUSIONS Phylogenetic analysis provides insights into the introduction and transmission dynamics of the Omicron variant in Pakistan, identifying Sindh as a hotspot for viral dissemination. Such data linked with public health efforts can help limit surges of new infections.
Collapse
Affiliation(s)
- Ali Raza Bukhari
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, 74800, Pakistan
| | - Javaria Ashraf
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, 74800, Pakistan
| | - Akbar Kanji
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, 74800, Pakistan
| | - Yusra Abdul Rahman
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, 74800, Pakistan
| | - Nídia S Trovão
- Fogarty International Center, U.S. National Institutes of Health, 16 Center Drive, Bethesda, MD, 20892, USA
| | - Peter M Thielen
- Johns Hopkins University Applied Physics Laboratory, 11100 Johns Hopkins Road, Laurel, MD, 20723, USA
| | - Maliha Yameen
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, 74800, Pakistan
| | - Samiah Kanwar
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, 74800, Pakistan
| | - Waqasuddin Khan
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, 74800, Pakistan
| | - Furqan Kabir
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, 74800, Pakistan
| | - Muhammad Imran Nisar
- Department of Pediatrics and Child Health, Aga Khan University, Karachi, 74800, Pakistan
| | - Brian Merritt
- Johns Hopkins University Applied Physics Laboratory, 11100 Johns Hopkins Road, Laurel, MD, 20723, USA
| | - Rumina Hasan
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, 74800, Pakistan
| | - David Spiro
- Fogarty International Center, U.S. National Institutes of Health, 16 Center Drive, Bethesda, MD, 20892, USA
| | - Zeba Rasmussen
- Fogarty International Center, U.S. National Institutes of Health, 16 Center Drive, Bethesda, MD, 20892, USA
| | - Uzma Bashir Aamir
- World Health Organization Country Office, Park Road, Chak Shahzad, Islamabad, Pakistan
| | - Zahra Hasan
- Department of Pathology and Laboratory Medicine, Aga Khan University, Karachi, 74800, Pakistan.
| |
Collapse
|
6
|
Cárdenas-Fuentes G, Bosch de Basea M, Cobo I, Subirana I, Ceresa M, Famada E, Gimeno-Santos E, Delgado-Ortiz L, Faner R, Molina-Molina M, Agustí À, Muñoz X, Sibila O, Gea J, Garcia-Aymerich J. Validity of prognostic models of critical COVID-19 is variable. A systematic review with external validation. J Clin Epidemiol 2023; 159:274-288. [PMID: 37142168 PMCID: PMC10152752 DOI: 10.1016/j.jclinepi.2023.04.011] [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: 07/28/2022] [Revised: 01/26/2023] [Accepted: 04/25/2023] [Indexed: 05/06/2023]
Abstract
OBJECTIVES To identify prognostic models which estimate the risk of critical COVID-19 in hospitalized patients and to assess their validation properties. STUDY DESIGN AND SETTING We conducted a systematic review in Medline (up to January 2021) of studies developing or updating a model that estimated the risk of critical COVID-19, defined as death, admission to intensive care unit, and/or use of mechanical ventilation during admission. Models were validated in two datasets with different backgrounds (HM [private Spanish hospital network], n = 1,753, and ICS [public Catalan health system], n = 1,104), by assessing discrimination (area under the curve [AUC]) and calibration (plots). RESULTS We validated 18 prognostic models. Discrimination was good in nine of them (AUCs ≥ 80%) and higher in those predicting mortality (AUCs 65%-87%) than those predicting intensive care unit admission or a composite outcome (AUCs 53%-78%). Calibration was poor in all models providing outcome's probabilities and good in four models providing a point-based score. These four models used mortality as outcome and included age, oxygen saturation, and C-reactive protein among their predictors. CONCLUSION The validity of models predicting critical COVID-19 by using only routinely collected predictors is variable. Four models showed good discrimination and calibration when externally validated and are recommended for their use.
Collapse
Affiliation(s)
- Gabriela Cárdenas-Fuentes
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; School of Health Sciences, Blanquerna-Universitat Ramon Llull, Barcelona, Spain.
| | - Magda Bosch de Basea
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Inés Cobo
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Isaac Subirana
- Instituto Hospital del Mar de Investigaciones Médicas (IMIM), Barcelona, Spain; CIBER Enfermedades Cardiovasculares (CIBERCV), ISCIII, Spain
| | - Mario Ceresa
- BCNMedTech, Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | | | - Elena Gimeno-Santos
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain; Respiratory Institute, Hospital Clinic, Barcelona, Spain
| | - Laura Delgado-Ortiz
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| | - Rosa Faner
- Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Universitat de Barcelona, Barcelona, Spain; CIBER Enfermedades Respiratorias (CIBERES), ISCIII, Spain
| | - María Molina-Molina
- CIBER Enfermedades Respiratorias (CIBERES), ISCIII, Spain; Servicio de Neumología, Hospital Universitario de Bellvitge, L'Hospitalet de Llobregat, Spain; Instituto de Investigación Biomédica de Bellvitge (IDIBELL), L'Hospitalet de Llobregat, Spain
| | - Àlvar Agustí
- Respiratory Institute, Hospital Clinic, Barcelona, Spain; Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; Universitat de Barcelona, Barcelona, Spain; CIBER Enfermedades Respiratorias (CIBERES), ISCIII, Spain
| | - Xavier Muñoz
- CIBER Enfermedades Respiratorias (CIBERES), ISCIII, Spain; Servicio de Neumología, Hospital Universitario Vall d'Hebron, Barcelona, Spain; Departamento de Biología celular, fisiología e inmunología, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Oriol Sibila
- Respiratory Institute, Hospital Clinic, Barcelona, Spain; Instituto de Investigaciones Biomédicas August Pi i Sunyer (IDIBAPS), Barcelona, Spain; CIBER Enfermedades Respiratorias (CIBERES), ISCIII, Spain
| | - Joaquim Gea
- Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Enfermedades Respiratorias (CIBERES), ISCIII, Spain; Servicio de Neumología, Hospital del Mar-IMIM, Barcelona, Spain; Fundació Barcelona Respiratory Network (BRN), Barcelona, Spain
| | - Judith Garcia-Aymerich
- ISGlobal, Barcelona, Spain; Universitat Pompeu Fabra (UPF), Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Barcelona, Spain
| |
Collapse
|
7
|
Maschietto C, Otto G, Rouzé P, Debortoli N, Bihin B, Nyinkeu L, Denis O, Huang TD, Mullier F, Bogaerts P, Degosserie J. Minimal requirements for ISO15189 validation and accreditation of three next generation sequencing procedures for SARS-CoV-2 surveillance in clinical setting. Sci Rep 2023; 13:6934. [PMID: 37117393 PMCID: PMC10140720 DOI: 10.1038/s41598-023-34088-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 04/24/2023] [Indexed: 04/30/2023] Open
Abstract
Rapid and recurrent breakthroughs of new SARS-CoV-2 strains (variants) have prompted public health authorities worldwide to set up surveillance networks to monitor the circulation of variants of concern. The use of next-generation sequencing technologies has raised the need for quality control assessment as required in clinical laboratories. The present study is the first to propose a validation guide for SARS-CoV-2 typing using three different NGS methods fulfilling ISO15189 standards. These include the assessment of the risk, specificity, accuracy, reproducibility, and repeatability of the methods. Among the three methods used, two are amplicon-based involving reverse transcription polymerase chain reaction (Artic v3 and Midnight v1) on Oxford Nanopore Technologies while the third one is amplicon-based using reverse complement polymerase chain reaction (Nimagen) on Illumina technology. We found that all methods met the quality requirement (e.g., 100% concordant typing results for accuracy, reproducibility, and repeatability) for SARS-CoV-2 typing in clinical setting. Additionally, the typing results emerging from each of the three sequencing methods were compared using three widely known nomenclatures (WHO, Pangolineage, and Nextclade). They were also compared regarding single nucleotide variations. The outcomes showed that Artic v3 and Nimagen should be privileged for outbreak investigation as they provide higher quality results for samples that do not meet inclusion criteria for analysis in a clinical setting. This study is a first step towards validation of laboratory developed NGS tests in the context of the new European regulation for medical devices and in vitro diagnostics.
Collapse
Affiliation(s)
- Céline Maschietto
- Department of Laboratory Medicine, UCLouvain, CHU UCL Namur, 5530, Yvoir, Belgium
- COVID-19 Federal Testing Platform Bis, CHU UCL Namur & UNamur, 5530, Yvoir, Belgium
| | - Gaëtan Otto
- Department of Laboratory Medicine, UCLouvain, CHU UCL Namur, 5530, Yvoir, Belgium
- COVID-19 Federal Testing Platform Bis, CHU UCL Namur & UNamur, 5530, Yvoir, Belgium
| | - Pauline Rouzé
- Department of Laboratory Medicine, UCLouvain, CHU UCL Namur, 5530, Yvoir, Belgium
- Laboratory of Microbiology, CHU UCL Namur, 5530, Yvoir, Belgium
| | - Nicolas Debortoli
- Department of Laboratory Medicine, UCLouvain, CHU UCL Namur, 5530, Yvoir, Belgium
- Namur Molecular Tech, CHU UCL Namur, 5530, Yvoir, Belgium
| | - Benoît Bihin
- Scientific Support Unit, CHU UCL Namur, 5530, Yvoir, Belgium
| | - Lesly Nyinkeu
- Department of Laboratory Medicine, UCLouvain, CHU UCL Namur, 5530, Yvoir, Belgium
- COVID-19 Federal Testing Platform Bis, CHU UCL Namur & UNamur, 5530, Yvoir, Belgium
- Namur Molecular Tech, CHU UCL Namur, 5530, Yvoir, Belgium
| | - Olivier Denis
- Department of Laboratory Medicine, UCLouvain, CHU UCL Namur, 5530, Yvoir, Belgium
- COVID-19 Federal Testing Platform Bis, CHU UCL Namur & UNamur, 5530, Yvoir, Belgium
- Laboratory of Microbiology, CHU UCL Namur, 5530, Yvoir, Belgium
| | - Te-Din Huang
- Department of Laboratory Medicine, UCLouvain, CHU UCL Namur, 5530, Yvoir, Belgium
- COVID-19 Federal Testing Platform Bis, CHU UCL Namur & UNamur, 5530, Yvoir, Belgium
- Laboratory of Microbiology, CHU UCL Namur, 5530, Yvoir, Belgium
| | - François Mullier
- Department of Laboratory Medicine, UCLouvain, CHU UCL Namur, 5530, Yvoir, Belgium
- COVID-19 Federal Testing Platform Bis, CHU UCL Namur & UNamur, 5530, Yvoir, Belgium
| | - Pierre Bogaerts
- Department of Laboratory Medicine, UCLouvain, CHU UCL Namur, 5530, Yvoir, Belgium
- Laboratory of Microbiology, CHU UCL Namur, 5530, Yvoir, Belgium
| | - Jonathan Degosserie
- Department of Laboratory Medicine, UCLouvain, CHU UCL Namur, 5530, Yvoir, Belgium.
- COVID-19 Federal Testing Platform Bis, CHU UCL Namur & UNamur, 5530, Yvoir, Belgium.
- Namur Molecular Tech, CHU UCL Namur, 5530, Yvoir, Belgium.
| |
Collapse
|
8
|
de Souza Andrade A, Freitas EF, de Castro Barbosa E, Guimarães N, de Melo Iani FC, da Costa AVB, Bernardes AFL, Adelino TER, Ataide ACZ, Gregianini TS, Nunes JD, Stringari LL, Riediger IN, Fernandes SB, de Jesus R, Fonseca V, Caldas S. Potential use of high-resolution melting analyses for SARS-CoV-2 genomic surveillance. J Virol Methods 2023; 317:114742. [PMID: 37116586 PMCID: PMC10132831 DOI: 10.1016/j.jviromet.2023.114742] [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: 02/22/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 04/30/2023]
Abstract
The pandemic caused by COVID-19 and the emergence of new variants of SARS-CoV-2 have generated clinical and epidemiological impacts on a global scale. The use of strategies for monitoring viral circulation and identifying mutations in genomic regions involved in host interaction are important measures to mitigate viral dissemination and reduce its likely complications on population health. In this context, the objective of this work was to explore the potential of high-resolution melting (HRM) analysis combined with one-step real-time reverse transcription PCR in a closed-tube system, as a fast and convenient method of screening for SARS-CoV-2 mutations with possible implications on host-pathogen interactions. The HRM analyses allowed the distinction of the Gamma, Zeta, Alpha, Delta, and Omicron variants against the predecessors (B.1.1.28, B.1.1.33) of occurrence in Brazil. It is concluded that the molecular tool standardized here has the potential to optimize the genomic surveillance of SARS-CoV-2, and could be adapted for genomic surveillance of other pathogens, due to its ability to detect, prior to sequencing, samples suggestive of new variants, selecting them more assertively and earlier for whole genome sequencing when compared to random screening.
Collapse
Affiliation(s)
- Adriana de Souza Andrade
- Serviço de Pesquisa em Doenças Infecciosas, Divisão de Ciência e Inovação, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil
| | - Eduarda Fernandes Freitas
- Serviço de Pesquisa em Doenças Infecciosas, Divisão de Ciência e Inovação, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil; Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Emerson de Castro Barbosa
- Serviço de Pesquisa em Doenças Infecciosas, Divisão de Ciência e Inovação, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil; Serviço de Virologia e Riquetsioses, Divisão de Epidemiologia e Controle de Doenças, Laboratório Central do Estado de Minas Gerais, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil
| | - Natália Guimarães
- Serviço de Virologia e Riquetsioses, Divisão de Epidemiologia e Controle de Doenças, Laboratório Central do Estado de Minas Gerais, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil
| | - Felipe Campos de Melo Iani
- Serviço de Virologia e Riquetsioses, Divisão de Epidemiologia e Controle de Doenças, Laboratório Central do Estado de Minas Gerais, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil; Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alana Vitor Barbosa da Costa
- Serviço de Virologia e Riquetsioses, Divisão de Epidemiologia e Controle de Doenças, Laboratório Central do Estado de Minas Gerais, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil
| | - André Felipe Leal Bernardes
- Serviço de Virologia e Riquetsioses, Divisão de Epidemiologia e Controle de Doenças, Laboratório Central do Estado de Minas Gerais, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil
| | - Talita Emile Ribeiro Adelino
- Serviço de Virologia e Riquetsioses, Divisão de Epidemiologia e Controle de Doenças, Laboratório Central do Estado de Minas Gerais, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil
| | - Ana Caroline Zampiroli Ataide
- Serviço de Pesquisa em Doenças Infecciosas, Divisão de Ciência e Inovação, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil
| | - Tatiana Schäffer Gregianini
- Laboratório de Influenza e outros Vírus Respiratórios, Laboratório de Biologia Molecular de Dengue, Febre Amarela, Zika e Chikungunya, Seção de Virologia. Laboratório Central do Estado do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Jônathas Dias Nunes
- Laboratório Central Noel Nutels. Laboratório Central do Estado do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Lorenzzo L Stringari
- Laboratório Central de Saúde Pública do Estado do Espírito Santo, Secretaria de Estado de Saúde do Espírito Santo, Vitória, ES, Brazil; Núcleo de Doenças Infecciosas, Universidade Federal do Espírito Santo, Vitória, ES, Brazil
| | - Irina Nastassja Riediger
- Divisão dos Laboratórios de Epidemiologia e Controle de Doenças, Laboratório Central do Estado do Paraná, São José dos Pinhais, PR, Brazil
| | | | - Ronaldo de Jesus
- Coordenação-Geral de Laboratórios de Saúde Pública, Ministério da Saúde, Brasília, Distrito Federal, Brazil; Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil
| | - Vagner Fonseca
- Organização Pan-Americana da Saúde/Organização Mundial da Saúde, Brasília, Distrito Federal, Brazil
| | - Sérgio Caldas
- Serviço de Pesquisa em Doenças Infecciosas, Divisão de Ciência e Inovação, Fundação Ezequiel Dias, Belo Horizonte, MG, Brazil.
| |
Collapse
|
9
|
Kwak HW, Park HJ, Jung SY, Oh EY, Park SI, Kim Y, Park HJ, Park S, Kim YJ, Ko HL, Lee JA, Won H, Hwang YH, Kim SY, Kim SE, Bae SE, Yoon M, Kim JO, Song M, Lee SJ, Seo KW, Lee K, Kim D, Kim H, Lee SM, Hong SH, Nam JH. Recombinant measles virus encoding the spike protein of SARS-CoV-2 efficiently induces Th1 responses and neutralizing antibodies that block SARS-CoV-2 variants. Vaccine 2023; 41:1892-1901. [PMID: 36792434 PMCID: PMC9902292 DOI: 10.1016/j.vaccine.2023.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 02/03/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023]
Abstract
Owing to the rapid spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants, the development of effective and safe vaccines has become a priority. The measles virus (MeV) vaccine is an attractive vaccine platform as it has been administered to children for more than 40 years in over 100 countries. In this study, we developed a recombinant MeV expressing the full-length SARS-CoV-2 spike protein (rMeV-S) and tested its efficacy using mouse and hamster models. In hCD46Tg mice, two-dose rMeV-S vaccination induced higher Th1 secretion and humoral responses than one-dose vaccination. Interestingly, neutralizing antibodies induced by one-dose and two-dose rMeV-S immunization effectively blocked the entry of the α, β, γ, and δ variants of SARS-CoV-2. Furthermore, two-dose rMeV-S immunization provided complete protection against SARS-CoV-2 in the hamster model. These results suggest the potential of rMeV-S as a vaccine candidate for targeting SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Hye Won Kwak
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; SML biopharm, Gyeonggi-do, Bucheon, Republic of Korea
| | - Hyo-Jung Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea
| | - Seo-Yeon Jung
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Eun Young Oh
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Sang-In Park
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, Kangwon-do, Republic of Korea
| | - Yeonhwa Kim
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - Hyeong-Jun Park
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; SML biopharm, Gyeonggi-do, Bucheon, Republic of Korea
| | - Sohyun Park
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea
| | - You-Jin Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Hae Li Ko
- Division of Research Program, Scripps Korea Antibody Institute, Chuncheon, Kangwon-do, Republic of Korea
| | - Jung-Ah Lee
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Hyeran Won
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Yun-Ho Hwang
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Seo Yeon Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Se Eun Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Seoung Eun Bae
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Minhyuk Yoon
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Jae-Ouk Kim
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Manki Song
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Su Jeen Lee
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Ki-Weon Seo
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Kunse Lee
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Dokeun Kim
- Division of Infectious Disease Vaccine Research, Center for Vaccine Research, National Institute of Infectious Diseases, National Institute of Health, Republic of Korea
| | - Hun Kim
- Department of R&D, SK bioscience, Pangyoro, Bundang-gu, Republic of Korea
| | - Sang-Myeong Lee
- College of Veterinary Medicine, Chungbuk National University, Cheongju 28644, Republic of Korea.
| | - So-Hee Hong
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul 07804, Republic of Korea.
| | - Jae-Hwan Nam
- Department of Medical and Biological Sciences, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea; BK Plus Department of Biotechnology, The Catholic University of Korea, Gyeonggi-do, Bucheon, Republic of Korea.
| |
Collapse
|
10
|
Nazmunnahar, Ahmed I, Islam MR. Risk evaluation and mitigation strategies for newly detected SARS-CoV-2 Omicron BF.7 subvariant: A brief report. Health Sci Rep 2023; 6:e1127. [PMID: 36875932 PMCID: PMC9981880 DOI: 10.1002/hsr2.1127] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 02/12/2023] [Accepted: 02/13/2023] [Indexed: 03/06/2023] Open
Abstract
Mutations of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) are always going on. The pathogenic characteristics of a virus are influenced by mutations in the viral genome. Therefore, the recently identified Omicron BF.7 subvariant might harm humans. Here we aimed to evaluate the potential risks of this newly detected variant and identify possible mitigation strategies. The frequent mutation associated with SARS-CoV-2 makes it more concerning compared to other viruses. The Omicron variant of SARS-CoV-2 has unique changes in the structural amino acid. Thus, Omicron subvariants are different from other coronavirus variants in terms of viral spread, disease severity, vaccine neutralization capacity, and immunity evade. Moreover, Omicron subvariant BF.7 is an offspring of BA.4 and BA.5. Similar S glycoprotein sequences are present among BF.7, BA.4, and BA.5. There is a change in the R346T gene in the receptor binding site of Omicron BF.7 than other Omicron subvariants. This BF.7 subvariant has created a limitation in current monoclonal antibody therapy. Omicron has mutated since it emerged, and the subvariants are improving in terms of transmission as well as antibody evasion. Therefore, the healthcare authorities should pay attention to the BF.7 subvariant of Omicron. The recent upsurge may create havoc all of a sudden. Scientists and researchers across the world should monitor the nature and mutations of SARS-CoV-2 variants. Also, they should find ways to fight the current circulatory variants and any future mutations.
Collapse
Affiliation(s)
- Nazmunnahar
- Department of Sociology, Eden Women's CollegeNational University BangladeshGazipurBangladesh
| | - Iftekhar Ahmed
- Department of PharmacyUniversity of Asia PacificDhakaBangladesh
| | | |
Collapse
|
11
|
Paulino-Ramírez R, Pham K, Breban MI, Peguero A, Jabier M, Sánchez N, Eustate I, Ruiz I, Grubaugh ND, Hahn AM. Genome Sequence of a Recombinant SARS-CoV-2 Lineage XAM (BA.1.1/BA.2.9) Strain from a Clinical Sample in Santo Domingo, Dominican Republic. Microbiol Resour Announc 2023; 12:e0111322. [PMID: 36448812 PMCID: PMC9872639 DOI: 10.1128/mra.01113-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/16/2022] [Indexed: 12/05/2022] Open
Abstract
Here, we report a recombinant severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) lineage XAM (Omicron BA.1.1/BA.2.9) strain that was collected in Santo Domingo, Dominican Republic. This demonstrates how SARS-CoV-2 variants can vary greatly between regions and thus underlines the great importance of regional genomic surveillance efforts.
Collapse
Affiliation(s)
- Robert Paulino-Ramírez
- Instituto de Medicina Tropical y Salud Global, Universidad Iberoamericana, UNIBE Research Hub, Santo Domingo, Dominican Republic
| | - Kien Pham
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Mallery I. Breban
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Armando Peguero
- Instituto de Medicina Tropical y Salud Global, Universidad Iberoamericana, UNIBE Research Hub, Santo Domingo, Dominican Republic
| | - Maridania Jabier
- Instituto de Medicina Tropical y Salud Global, Universidad Iberoamericana, UNIBE Research Hub, Santo Domingo, Dominican Republic
- Servicio Nacional de Salud, Ministry of Health, Santo Domingo, Dominican Republic
| | - Nelissa Sánchez
- Instituto de Medicina Tropical y Salud Global, Universidad Iberoamericana, UNIBE Research Hub, Santo Domingo, Dominican Republic
| | - Iscania Eustate
- Instituto de Medicina Tropical y Salud Global, Universidad Iberoamericana, UNIBE Research Hub, Santo Domingo, Dominican Republic
- Servicio Nacional de Salud, Ministry of Health, Santo Domingo, Dominican Republic
| | - Ingrid Ruiz
- Instituto de Medicina Tropical y Salud Global, Universidad Iberoamericana, UNIBE Research Hub, Santo Domingo, Dominican Republic
| | - Nathan D. Grubaugh
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| | - Anne M. Hahn
- Department of Epidemiology of Microbial Diseases, Yale School of Public Health, New Haven, Connecticut, USA
| |
Collapse
|
12
|
Perico CP, De Pierri CR, Neto GP, Fernandes DR, Pedrosa FO, de Souza EM, Raittz RT. Genomic landscape of the SARS-CoV-2 pandemic in Brazil suggests an external P.1 variant origin. Front Microbiol 2022; 13:1037455. [PMID: 36620039 PMCID: PMC9814972 DOI: 10.3389/fmicb.2022.1037455] [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: 09/05/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
Brazil was the epicenter of worldwide pandemics at the peak of its second wave. The genomic/proteomic perspective of the COVID-19 pandemic in Brazil could provide insights to understand the global pandemics behavior. In this study, we track SARS-CoV-2 molecular information in Brazil using real-time bioinformatics and data science strategies to provide a comparative and evolutive panorama of the lineages in the country. SWeeP vectors represented the Brazilian and worldwide genomic/proteomic data from Global Initiative on Sharing Avian Influenza Data (GISAID) between February 2020 and August 2021. Clusters were analyzed and compared with PANGO lineages. Hierarchical clustering provided phylogenetic and evolutionary analyses of the lineages, and we tracked the P.1 (Gamma) variant origin. The genomic diversity based on Chao's estimation allowed us to compare richness and coverage among Brazilian states and other representative countries. We found that epidemics in Brazil occurred in two moments with different genetic profiles. The P.1 lineages emerged in the second wave, which was more aggressive. We could not trace the origin of P.1 from the variants present in Brazil. Instead, we found evidence pointing to its external source and a possible recombinant event that may relate P.1 to a B.1.1.28 variant subset. We discussed the potential application of the pipeline for emerging variants detection and the PANGO terminology stability over time. The diversity analysis showed that the low coverage and unbalanced sequencing among states in Brazil could have allowed the silent entry and dissemination of P.1 and other dangerous variants. This study may help to understand the development and consequences of variants of concern (VOC) entry.
Collapse
Affiliation(s)
- Camila P Perico
- Laboratory of Artificial Intelligence Applied to Bioinformatics, Professional and Technological Education Sector (SEPT), Federal University of Paraná, Curitiba, Brazil
- Graduate Program in Bioinformatics, Professional and Technological Education Sector (SEPT), Federal University of Paraná, Curitiba, Brazil
| | - Camilla R De Pierri
- Laboratory of Artificial Intelligence Applied to Bioinformatics, Professional and Technological Education Sector (SEPT), Federal University of Paraná, Curitiba, Brazil
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, Brazil
| | - Giuseppe Pasqualato Neto
- Laboratory of Artificial Intelligence Applied to Bioinformatics, Professional and Technological Education Sector (SEPT), Federal University of Paraná, Curitiba, Brazil
| | - Danrley R Fernandes
- Laboratory of Artificial Intelligence Applied to Bioinformatics, Professional and Technological Education Sector (SEPT), Federal University of Paraná, Curitiba, Brazil
- Graduate Program in Bioinformatics, Professional and Technological Education Sector (SEPT), Federal University of Paraná, Curitiba, Brazil
| | - Fabio O Pedrosa
- Graduate Program in Bioinformatics, Professional and Technological Education Sector (SEPT), Federal University of Paraná, Curitiba, Brazil
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, Brazil
| | - Emanuel M de Souza
- Graduate Program in Bioinformatics, Professional and Technological Education Sector (SEPT), Federal University of Paraná, Curitiba, Brazil
- Department of Biochemistry and Molecular Biology, Federal University of Paraná, Curitiba, Brazil
| | - Roberto T Raittz
- Laboratory of Artificial Intelligence Applied to Bioinformatics, Professional and Technological Education Sector (SEPT), Federal University of Paraná, Curitiba, Brazil
- Graduate Program in Bioinformatics, Professional and Technological Education Sector (SEPT), Federal University of Paraná, Curitiba, Brazil
| |
Collapse
|
13
|
Andrés C, Piñana M, Borràs-Bermejo B, González-Sánchez A, García-Cehic D, Esperalba J, Rando A, Zules-Oña RG, Campos C, Codina MG, Blanco-Grau A, Colomer-Castell S, Martín MC, Castillo C, García-Comuñas K, Vásquez-Mercado R, Martins-Martins R, Saubi N, Campins-Martí M, Pumarola T, Quer J, Antón A. A year living with SARS-CoV-2: an epidemiological overview of viral lineage circulation by whole-genome sequencing in Barcelona city (Catalonia, Spain). Emerg Microbes Infect 2022; 11:172-181. [PMID: 34842496 PMCID: PMC8741249 DOI: 10.1080/22221751.2021.2011617] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/23/2021] [Indexed: 12/24/2022]
Abstract
Herein, we describe the genetic diversity of circulating SARS-CoV-2 viruses by whole-genome sequencing (WGS) in Barcelona city (Catalonia, Spain) throughout the first four pandemic waves. From weeks 11/2020-24/2021, SARS-CoV-2-positive respiratory samples were randomly selected per clinical setting (80% from primary care or 20% from the hospital), age group, and week. WGS was performed following the ARTICv3 protocol on MiSeq or NextSeq2000 Illumina platforms. Nearly complete consensus sequences were used for genetic characterization based on GISAID and PANGOLIN nomenclatures. From 2475 samples, 2166 (87%) were fully sequenced (78% from primary care and 22% from hospital settings). Multiple genetic lineages were co-circulating, but four were predominant at different periods. While B.1.5 (50.68%) and B.1.1 (32.88%) were the major lineages during the first pandemic wave, B.1.177 (66.85%) and B.1.1.7 (83.80%) were predominant during the second, third, and fourth waves, respectively. Almost all (96.4%) were carrying D614G mutation in the S protein, with additional mutations that define lineages or variants. But some mutations of concern, such as E484K from B.1.351 and P.1 lineages are currently under monitoring, together with those observed in the receptor-binding domain or N-terminal domain, such as L452R and T478K from B.1.617.2 lineage. The fact that a predominant lineage was observed in each pandemic wave suggests advantageous properties over other contemporary co-circulating variants. This genetic variability should be monitored, especially when a massive vaccination campaign is ongoing because the potential selection and emergence of novel antigenic SARS-CoV-2 strains related to immunological escapement events.
Collapse
Affiliation(s)
- Cristina Andrés
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Maria Piñana
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Blanca Borràs-Bermejo
- Preventive Medicine and Epidemiology Department, Vall d´Hebron Research Institute (VHIR), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Alejandra González-Sánchez
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Damir García-Cehic
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Juliana Esperalba
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Ariadna Rando
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Ricardo-Gabriel Zules-Oña
- Preventive Medicine and Epidemiology Department, Vall d´Hebron Research Institute (VHIR), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Carolina Campos
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Maria Gema Codina
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Albert Blanco-Grau
- Clinical Biochemistry (Clinical Laboratories), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Sergi Colomer-Castell
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Maria Carmen Martín
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Carla Castillo
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Karen García-Comuñas
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Rodrigo Vásquez-Mercado
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Reginaldo Martins-Martins
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Narcís Saubi
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron, Barcelona, Spain
| | - Magda Campins-Martí
- Preventive Medicine and Epidemiology Department, Vall d´Hebron Research Institute (VHIR), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Tomàs Pumarola
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| | - Josep Quer
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital Campus, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Madrid, Spain
| | - Andrés Antón
- Respiratory Viruses Unit, Microbiology Department, Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Barcelona Hospital CampusBarcelona, Spain
| |
Collapse
|
14
|
A comparison of high-throughput SARS-CoV-2 sequencing methods from nasopharyngeal samples. Sci Rep 2022; 12:12561. [PMID: 35869099 PMCID: PMC9306416 DOI: 10.1038/s41598-022-16549-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/12/2022] [Indexed: 12/25/2022] Open
Abstract
The COVID-19 pandemic caused by the new Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to threaten public health and burden healthcare systems worldwide. Whole SARS-CoV-2 genome sequencing has become essential for epidemiological monitoring and identification of new variants, which could represent a risk of increased transmissibility, virulence, or resistance to vaccines or treatment. Different next-generation sequencing approaches are used in SARS-CoV-2 sequencing, although with different ability to provide whole genome coverage without gaps and to reliably detect new variants. In this study, we compared the performance of three target enrichment methods (two multiplex amplification methods and one hybridization capture) using nasopharyngeal swabs from infected individuals. We applied these target enrichment methods to the same set of nasopharyngeal samples (N = 93) in high-throughput mode. SARS-CoV-2 genome was obtained using short-read next-generation sequencing. We observed that each method has some advantages, such as high mapping rate (CleanPlex and COVIDSeq) or absence of systematic variant calling error (SureSelect) as well as their limitations such as suboptimal uniformity of coverage (CleanPlex), high cost (SureSelect) or supply shortages (COVIDSeq). Nevertheless, each of the three target enrichment kits tested in this study yielded acceptable results of whole SARS-CoV-2 genome sequencing and either of them can therefore be used in prospective programs of genomic surveillance of SARS-CoV-2. Genomic surveillance will be crucial to overcoming the ongoing pandemic of COVID-19, despite its successive waves and continually emerging variants.
Collapse
|
15
|
Phan T, Boes S, McCullough M, Gribschaw J, Wells A. Mini-evaluation of the Lyra SARS-CoV-2 assay to detect Omicron BA.1 and BA.2 in nasopharyngeal swabs. JOURNAL OF CLINICAL VIROLOGY PLUS 2022; 2:100118. [PMID: 36277996 PMCID: PMC9578329 DOI: 10.1016/j.jcvp.2022.100118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 10/07/2022] [Accepted: 10/17/2022] [Indexed: 11/30/2022] Open
Affiliation(s)
- Tung Phan
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Stephanie Boes
- Clinical Microbiology Laboratory, UPMC Hospital System, Pittsburgh, PA 15261, United States
| | - Melissa McCullough
- Clinical Microbiology Laboratory, UPMC Hospital System, Pittsburgh, PA 15261, United States
| | - Jamie Gribschaw
- Clinical Microbiology Laboratory, UPMC Hospital System, Pittsburgh, PA 15261, United States
| | - Alan Wells
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, United States
| |
Collapse
|
16
|
Guzmán MG, Pérez L, Tejero Y, Mederos D, Aguado ME, Pintos Y, Valdes O, Álvarez M, de León LDRP, Baldoquín W, Martínez Y, Figueredo C, Naranjo C, Nuñes L, López Almaguer Y, Fernández JRDA, Portal Miranda JA, Kourí V. Emergence and evolution of SARS-CoV-2 genetic variants during the Cuban epidemic. JOURNAL OF CLINICAL VIROLOGY PLUS 2022; 2:100104. [PMID: 36034515 PMCID: PMC9394106 DOI: 10.1016/j.jcvp.2022.100104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Accepted: 08/21/2022] [Indexed: 11/23/2022] Open
Abstract
The SARS CoV-2 D614G variant circulated in Cuba in 2020. New viral variants were detected after the opening of the border in November 2020. We show the results of the genomic surveillance in Cuba from December 28, 2020, to September 28, 2021 and their relationship to the epidemiological situation in the country. A total of 1,406 nasopharyngeal exudates from COVID-19 patients were processed for RNA extraction and the 1836 bp fragment of the spike gene was amplified and sequenced. The mutations present were determined using the GISAID database. Prevalence ratios were estimated by fitting Poisson univariate and multivariate regression models to investigate associations between SARS-CoV-2 variant group (VOC, non-VOC) and disease outcome. Seventeen genetic variants were detected including VOC Alpha, Beta, Gamma and Delta, one variant of interest (VOI) (Lambda) and two previous VOI (A.2.5.1 and Zeta/P.2). Beta (34.77%), Delta (24.89%) and D614G (19%) variants were the most frequently detected. By June, Delta increased in frequency, displacing Beta. Disease severity increased significantly with age and VOC (PR =1.98, IC 95%: 1.33-3.05, p <0.05). Genomic surveillance allowed us to identify the upsurge of novel variants. Coinciding with the higher epidemic period, multiple variants were co-circulating. Although we cannot rule out that failure in the transmission containment measures occurred, the increase in the number of cases associated with the circulation of several variants, particularly the Beta and Delta variants is highly suggestive. A greater association of Beta variant with clinical severity and Delta variant with a greater transmissibility was observed.
Collapse
Affiliation(s)
- María G Guzmán
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| | - Lissette Pérez
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| | - Yahisel Tejero
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| | - Dailyn Mederos
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| | - Mirtha E Aguado
- Center of Protein Studies, Faculty of Biology, University of Havana, Havana, Cuba
| | - Yanet Pintos
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| | - Odalys Valdes
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| | - Mayling Álvarez
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| | | | - Waldemar Baldoquín
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| | | | - Claudia Figueredo
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| | - Celine Naranjo
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| | - Lirialys Nuñes
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| | | | | | | | - Vivian Kourí
- Department of Virology, Pedro Kourí Tropical Medicine Institute, Havana, Cuba
| |
Collapse
|
17
|
Shrestha LB, Foster C, Rawlinson W, Tedla N, Bull RA. Evolution of the SARS-CoV-2 omicron variants BA.1 to BA.5: Implications for immune escape and transmission. Rev Med Virol 2022; 32:e2381. [PMID: 35856385 PMCID: PMC9349777 DOI: 10.1002/rmv.2381] [Citation(s) in RCA: 309] [Impact Index Per Article: 103.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 12/15/2022]
Abstract
The first dominant SARS-CoV-2 Omicron variant BA.1 harbours 35 mutations in its Spike protein from the original SARS-CoV-2 variant that emerged late 2019. Soon after its discovery, BA.1 rapidly emerged to become the dominant variant worldwide and has since evolved into several variants. Omicron is of major public health concern owing to its high infectivity and antibody evasion. This review article examines the theories that have been proposed on the evolution of Omicron including zoonotic spillage, infection in immunocompromised individuals and cryptic spread in the community without being diagnosed. Added to the complexity of Omicron's evolution are the multiple reports of recombination events occurring between co-circulating variants of Omicron with Delta and other variants such as XE. Current literature suggests that the combination of the novel mutations in Omicron has resulted in the variant having higher infectivity than the original Wuhan-Hu-1 and Delta variant. However, severity is believed to be less owing to the reduced syncytia formation and lower multiplication in the human lung tissue. Perhaps most challenging is that several studies indicate that the efficacy of the available vaccines have been reduced against Omicron variant (8-127 times reduction) as compared to the Wuhan-Hu-1 variant. The administration of booster vaccine, however, compensates with the reduction and improves the efficacy by 12-35 fold. Concerningly though, the broadly neutralising monoclonal antibodies, including those approved by FDA for therapeutic use against previous SARS-CoV-2 variants, are mostly ineffective against Omicron with the exception of Sotrovimab and recent reports suggest that the Omicron BA.2 is also resistant to Sotrovimab. Currently two new Omicron variants BA.4 and BA.5 are emerging and are reported to be more transmissible and resistant to immunity generated by previous variants including Omicron BA.1 and most monoclonal antibodies. As new variants of SARS-CoV-2 will likely continue to emerge it is important that the evolution, and biological consequences of new mutations, in existing variants be well understood.
Collapse
Affiliation(s)
- Lok Bahadur Shrestha
- School of Medical SciencesFaculty of MedicineUNSWSydneyNew South WalesAustralia
- The Kirby InstituteUNSWSydneyNew South WalesAustralia
| | - Charles Foster
- School of Medical SciencesFaculty of MedicineUNSWSydneyNew South WalesAustralia
- Serology and Virology DivisionDepartment of MicrobiologyNew South Wales Health PathologySydneyNew South WalesAustralia
| | - William Rawlinson
- School of Medical SciencesFaculty of MedicineUNSWSydneyNew South WalesAustralia
- Serology and Virology DivisionDepartment of MicrobiologyNew South Wales Health PathologySydneyNew South WalesAustralia
| | - Nicodemus Tedla
- School of Medical SciencesFaculty of MedicineUNSWSydneyNew South WalesAustralia
| | - Rowena A. Bull
- School of Medical SciencesFaculty of MedicineUNSWSydneyNew South WalesAustralia
- The Kirby InstituteUNSWSydneyNew South WalesAustralia
| |
Collapse
|
18
|
Neto DFDL, Fonseca V, Jesus R, Dutra LH, Portela LMDO, Freitas C, Fillizola E, Soares B, Abreu ALD, Twiari S, Azevedo V, Goes-Neto A, de Medeiros AC, Lopes NP, Zanotto PMDA, Kato RB. Molecular dynamics simulations of the SARS-CoV-2 Spike protein and variants of concern: structural evidence for convergent adaptive evolution. J Biomol Struct Dyn 2022:1-13. [PMID: 35848330 DOI: 10.1080/07391102.2022.2097955] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The Spike protein's structure of the SARS-CoV-2 provides a unique opportunity to consider perturbations at the atomic level. We used the cryo-electron microscopy structure of the open conformation of the Spike protein to assess the impact of the mutations observed in the variants of concern at the molecular level. Molecular dynamics were subsequently performed with both the wt and the mutated forms to compare the flexibility and variation data for each residue of the three-dimensional fluctuations in the region associated with each alpha carbon. Additionally, protein-protein docking was used to investigate the interaction of each mutated profile with the ACE-2 receptor. After the molecular dynamics, the results show that the mutations increased the stability of the trimeric protein, with greater stability observed in the Gamma variant harboring the 10 characteristic mutations. The results of molecular dynamics, as shown by RMSF demonstrated for the residues that comprise the binding domain receptor (RBD), exhibited a reduction in flexibility, which was more pronounced in the Gamma variant. Finally, protein-protein docking experiments revealed an increase in the number of hydrophobic interactions and hydrogen bonds in the Gamma variant against the ACE-2 receptor, as opposed to the other variants. Taken together, these in silico experiments suggest that the evolution of the mutations favored the increased stability of Spike protein while potentially improving its interaction with the ACE-2 receptor, which in turn may indicate putative structural outcomes of the selection of these mutations in the convergent adaptive evolution as it has been observed for SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Daniel Ferreira de Lima Neto
- Coordenação-Geral de Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, Distrito Federal, Brazil
| | - Vagner Fonseca
- Coordenação-Geral de Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, Distrito Federal, Brazil.,KwaZulu-Natal Research Innovation and Sequencing Platform (KRISP), University of KwaZulu-Natal, Durban, South Africa.,Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ronaldo Jesus
- Coordenação-Geral de Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, Distrito Federal, Brazil.,Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Leonardo Hermes Dutra
- Coordenação-Geral de Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, Distrito Federal, Brazil
| | - Layssa Miranda de Oliveria Portela
- Coordenação-Geral de Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, Distrito Federal, Brazil
| | - Carla Freitas
- Coordenação-Geral de Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, Distrito Federal, Brazil
| | - Eduardo Fillizola
- Coordenação-Geral de Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, Distrito Federal, Brazil
| | - Breno Soares
- Coordenação-Geral de Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, Distrito Federal, Brazil
| | - André Luiz de Abreu
- Coordenação-Geral de Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, Distrito Federal, Brazil
| | - Sandeep Twiari
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Vasco Azevedo
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Aristóteles Goes-Neto
- Laboratório de Genética Celular e Molecular, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Arnaldo Correia de Medeiros
- Coordenação-Geral de Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, Distrito Federal, Brazil
| | - Norberto Peporine Lopes
- Núcleo de Pesquisa em Produtos Naturais e Sintéticos. Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, São Paulo, Brazil
| | - Paolo Marinho de Andrade Zanotto
- Laboratório de Evolução Molecular e Bioinformática (LEMB), Departamento de Microbiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, Brazil
| | - Rodrigo Bentes Kato
- Coordenação-Geral de Laboratórios de Saúde Pública, Secretaria de Vigilância em Saúde, Ministério da Saúde, Brasília, Distrito Federal, Brazil.,Laboratório de Biologia Molecular e Computacional de Fungos, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
19
|
Hufsky F, Abecasis A, Agudelo-Romero P, Bletsa M, Brown K, Claus C, Deinhardt-Emmer S, Deng L, Friedel CC, Gismondi MI, Kostaki EG, Kühnert D, Kulkarni-Kale U, Metzner KJ, Meyer IM, Miozzi L, Nishimura L, Paraskevopoulou S, Pérez-Cataluña A, Rahlff J, Thomson E, Tumescheit C, van der Hoek L, Van Espen L, Vandamme AM, Zaheri M, Zuckerman N, Marz M. Women in the European Virus Bioinformatics Center. Viruses 2022; 14:1522. [PMID: 35891501 PMCID: PMC9319252 DOI: 10.3390/v14071522] [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: 06/16/2022] [Revised: 07/05/2022] [Accepted: 07/07/2022] [Indexed: 02/01/2023] Open
Abstract
Viruses are the cause of a considerable burden to human, animal and plant health, while on the other hand playing an important role in regulating entire ecosystems. The power of new sequencing technologies combined with new tools for processing "Big Data" offers unprecedented opportunities to answer fundamental questions in virology. Virologists have an urgent need for virus-specific bioinformatics tools. These developments have led to the formation of the European Virus Bioinformatics Center, a network of experts in virology and bioinformatics who are joining forces to enable extensive exchange and collaboration between these research areas. The EVBC strives to provide talented researchers with a supportive environment free of gender bias, but the gender gap in science, especially in math-intensive fields such as computer science, persists. To bring more talented women into research and keep them there, we need to highlight role models to spark their interest, and we need to ensure that female scientists are not kept at lower levels but are given the opportunity to lead the field. Here we showcase the work of the EVBC and highlight the achievements of some outstanding women experts in virology and viral bioinformatics.
Collapse
Affiliation(s)
- Franziska Hufsky
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, 07743 Jena, Germany
| | - Ana Abecasis
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Global Health and Tropical Medicine, Institute of Hygiene and Tropical Medicine, New University of Lisbon, 1349-008 Lisbon, Portugal
| | - Patricia Agudelo-Romero
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Wal-Yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Nedlands, WA 6009, Australia
| | - Magda Bletsa
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Katherine Brown
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge CB2 1TN, UK
| | - Claudia Claus
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Institute of Medical Microbiology and Virology, Medical Faculty, Leipzig University, 04103 Leipzig, Germany
| | - Stefanie Deinhardt-Emmer
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Institute of Medical Microbiology, Jena University Hospital, 07747 Jena, Germany
| | - Li Deng
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Institute of Virology, Helmholtz Centre Munich-German Research Center for Environmental Health, 85764 Neuherberg, Germany
- Microbial Disease Prevention, School of Life Sciences, Technical University of Munich, 85354 Freising, Germany
| | - Caroline C. Friedel
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Institute of Informatics, Ludwig-Maximilians-Universität München, 80333 Munich, Germany
| | - María Inés Gismondi
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Institute of Agrobiotechnology and Molecular Biology (IABIMO), National Institute for Agriculture Technology (INTA), National Research Council (CONICET), Hurlingham B1686IGC, Argentina
- Department of Basic Sciences, National University of Luján, Luján B6702MZP, Argentina
| | - Evangelia Georgia Kostaki
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Department of Hygiene, Epidemiology and Medical Statistics, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece
| | - Denise Kühnert
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Transmission, Infection, Diversification and Evolution Group, Max Planck Institute for the Science of Human History, 07745 Jena, Germany
| | - Urmila Kulkarni-Kale
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Bioinformatics Centre, Savitribai Phule Pune University, Pune 411007, India
| | - Karin J. Metzner
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Irmtraud M. Meyer
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, 10115 Berlin, Germany
- Institute of Chemistry and Biochemistry, Department of Biology, Chemistry and Pharmacy, Freie Universität Berlin, 14195 Berlin, Germany
- Faculty of Mathematics and Computer Science, Freie Universität Berlin, 14195 Berlin, Germany
| | - Laura Miozzi
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Institute for Sustainable Plant Protection, National Research Council of Italy, 10135 Torino, Italy
| | - Luca Nishimura
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Department of Genetics, School of Life Science, The Graduate University for Advanced Studies (SOKENDAI), Mishima 411-8540, Japan
- Human Genetics Laboratory, National Institute of Genetics, Mishima 411-8540, Japan
| | - Sofia Paraskevopoulou
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Methods Development and Research Infrastructure, Bioinformatics and Systems Biology, Robert Koch Institute, 13353 Berlin, Germany
| | - Alba Pérez-Cataluña
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- VISAFELab, Department of Preservation and Food Safety Technologies, Institute of Agrochemistry and Food Technology, IATA-CSIC, 46980 Valencia, Spain
| | - Janina Rahlff
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Centre for Ecology and Evolution in Microbial Model Systems (EEMiS), Department of Biology and Environmental Science, Linneaus University, 391 82 Kalmar, Sweden
| | - Emma Thomson
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde, Glasgow G51 4TF, UK
- MRC-University of Glasgow Centre for Virus Research, Glasgow G61 1QH, UK
| | - Charlotte Tumescheit
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Lia van der Hoek
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Laboratory of Experimental Virology, Department of Medical Microbiology and Infection Prevention, Amsterdam UMC, University of Amsterdam, 1012 WX Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, 1100 DD Amsterdam, The Netherlands
| | - Lore Van Espen
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Anne-Mieke Vandamme
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Department of Microbiology, Immunology and Transplantation, Rega Institute, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, 1349-008 Lisbon, Portugal
- Institute for the Future, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium
| | - Maryam Zaheri
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Neta Zuckerman
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- Central Virology Laboratory, Public Health Services, Ministry of Health and Sheba Medical Center, Ramat Gan 52621, Israel
| | - Manja Marz
- European Virus Bioinformatics Center, 07743 Jena, Germany; (A.A.); (P.A.-R.); (M.B.); (K.B.); (C.C.); (S.D.-E.); (L.D.); (C.C.F.); (M.I.G.); (E.G.K.); (D.K.); (U.K.-K.); (K.J.M.); (I.M.M.); (L.M.); (L.N.); (S.P.); (A.P.-C.); (J.R.); (E.T.); (C.T.); (L.v.d.H.); (L.V.E.); (A.-M.V.); (M.Z.); (N.Z.)
- RNA Bioinformatics and High-Throughput Analysis, Friedrich Schiller University Jena, 07743 Jena, Germany
| |
Collapse
|
20
|
Spira B. The Impact of the Highly Virulent SARS-CoV-2 Gamma Variant on Young Adults in the State of São Paulo: Was It Inevitable? Cureus 2022; 14:e26486. [PMID: 35919213 PMCID: PMC9339207 DOI: 10.7759/cureus.26486] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2022] [Indexed: 11/21/2022] Open
Abstract
Background The coronavirus disease 2019 (COVID-19) pandemic had and is still having a tremendous impact on people all over the world, but it has been particularly harsh in South America. Nine out of 13 South American countries are among the 50 countries with the highest COVID-19 death rates. The gamma severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant that emerged by the end of 2020 in the Brazilian Amazon quickly spread throughout the country causing the harsh COVID-19 second wave. This variant displayed high viral loads, high transmissibility, and increased virulence as compared to previous variants. Aims The aim of this retrospective study is to revisit and analyse the epidemiology of the COVID-19 second wave in the state of São Paulo, the most populous Brazilian state. In addition to examining the possible factors that led to the emergence and propagation of the gamma variant, measures that could have prevented its spread and that of other highly virulent variants were also investigated. Materials and methods Data from São Paulo's official sources on morbidity, mortality, age distribution, and testing prior to and during the COVID-19 second wave (February - June 2021) and data regarding the distribution of SARS-CoV-2 variants in the country were parsed, analyzed, and compared to the period that anteceded the eruption of the second COVID-19 wave. Results In the state of São Paulo, the toll of the COVID-19 second wave surpassed that of the first 11 months of the pandemic (from March 2020 to January 2021), as 56% of the deaths occurred in the five months of the second wave between February and June 2021. The mean age of COVID-19 victims, which was already below life expectancy in the state dropped even further in the pandemic's second wave, reaching an average of 60 years of age. The years of life lost per death per month doubled and the case-fatality rate (CFR) of young adults (20-39 years old) more than trebled during this period. A number of hypotheses have been raised that might explain the emergence and spread of the gamma variant and the measures that could have been taken to prevent it and minimise its impact on the population. Conclusions Over 142,000 people died as a result of the SARS-CoV-2 gamma variant sweep in São Paulo in the first semester of 2021. Due to its high viral load, the gamma variant displayed high transmissibility and a high degree of virulence resulting in increased case fatality rates across most age tiers. Notably, this second wave was marked by a very significant increase in deaths among young adults. This increase was at least partially due to a deterioration in general health provoked by non-pharmaceutical interventions. In hindsight, a safer and more effective measure might have been to allow the free spread of the virus among the young and healthy in the first wave, thus conferring immunity against more virulent variants that emerged later on.
Collapse
|
21
|
Soares GDS, Morais LV, Silva KCN, Ferreira EM, Shio MT, Romano CM, Conde CR, Sabino EC, França CN, Nali LH. Low frequency of SARS-CoV2 infection in daycare centers during the reopening of school activities in the Southeast's poor area of Brazil. Rev Inst Med Trop Sao Paulo 2022; 64:e46. [PMID: 35749418 PMCID: PMC9236202 DOI: 10.1590/s1678-9946202264046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 05/20/2022] [Indexed: 11/21/2022] Open
Abstract
SARS-Cov2 has already infected over 482 million people and caused more than 6.1 million deaths. The beginning of the pandemic has led the health authorities of several countries to adopt non-pharmacological preventive measures such as daycare closures. The reopening took place when the country had the highest rates of infection and mortality (mainly due to the gamma variant (P.1) outbreak) and the beginning of the vaccination program. Therefore, we aimed to investigate the prevalence of SARS-CoV2 in daycare after educational activities resumed. The study was conducted in seven childcare facilities. Swab samples from the nasopharynx were collected from children and staff members. The viral RNA was obtained through PureLink RNA extraction kit purification and SARS-CoV2 presence was detected using the All plex SARS-CoV2 kit. The study population included 201 participants, including daycare workers and children. The average age of the workers and children is 40 and 3 years old, respectively. Among the children, 47.5% are female and among the workers, 91.4%. One (0.5%) test came out positive for the presence of SARS-CoV-2, which was from a sample of an asymptomatic childcare worker, and no secondary infections were detected. Considering that the return to daycare activities occurred during a period with a high number of deaths and a lack of vaccines throughout the country, the small number of cases indicates the effectiveness of the several preventive measures used by daycare centers in preventing SARS-CoV2 transmission.
Collapse
Affiliation(s)
| | - Lucas Vinicius Morais
- Universidade Santo Amaro, Programa de Pós-Graduação em Ciências da Saúde, São Paulo, São Paulo, Brazil
| | | | - Elaine Moura Ferreira
- Universidade Santo Amaro, Programa de Pós-Graduação em Ciências da Saúde, São Paulo, São Paulo, Brazil
| | - Marina Tiemi Shio
- Universidade Santo Amaro, Programa de Pós-Graduação em Ciências da Saúde, São Paulo, São Paulo, Brazil
| | - Camila Malta Romano
- Universidade de São Paulo, Faculdade de Medicina, Hospital das Clínicas (LIM 52), São Paulo, São Paulo, Brazil.,Universidade de São Paulo, Instituto de Medicina Tropical de São Paulo, Laboratório de Virologia, São Paulo, São Paulo, Brazil
| | - Carla Regiani Conde
- Universidade Santo Amaro, Escola de Enfermagem, São Paulo, São Paulo, Brazil
| | - Ester Cerdeira Sabino
- Universidade de São Paulo, Instituto de Medicina Tropical de São Paulo, Laboratório de Genética Aplicada às Doenças Infecciosas, São Paulo, São Paulo, Brazil
| | - Carolina Nunes França
- Universidade Santo Amaro, Programa de Pós-Graduação em Ciências da Saúde, São Paulo, São Paulo, Brazil
| | - Luiz Henrique Nali
- Universidade Santo Amaro, Programa de Pós-Graduação em Ciências da Saúde, São Paulo, São Paulo, Brazil
| |
Collapse
|
22
|
Identification of SARS-CoV-2 Variants of Concern Using Amplicon Next-Generation Sequencing. Microbiol Spectr 2022; 10:e0073622. [PMID: 35758686 PMCID: PMC9430688 DOI: 10.1128/spectrum.00736-22] [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] [Indexed: 11/20/2022] Open
Abstract
COVID-19 is caused by SARS-CoV-2, several virulent variants of which have emerged since 2019. More than 529 million people have been infected, and at least 6 million have died. Our aim was to develop a fast, accurate, low-cost method for detecting and identifying newly emerging variants of concern (VOCs) that could pose a global threat. The 341-bp DNA sequence of a specific region of the SARS-CoV-2’s spike protein was amplified by a one-step PCR on RNA samples from 46 patients. The product was sequenced using next-generation sequencing (NGS). DNA sequences from seven genomes, the original Wuhan isolate and six different representative variants obtained from the GISAID website, were used as references. Complete whole-genome sequences from local isolates were also obtained from the GISAID website, and their RNA was used for comparison. We used an amplicon-based NGS method (termed VOC-NGS) for genotyping and successfully identified all 46 samples. Fifteen (32.6%) were like the original isolate. Twenty-seven were VOCs: nine (19.5%) Alpha, eight (19%) Delta, six (14%) Beta, and four (8.7%) Omicron. Two were variants of interest (VOI): one (2%) Kappa and one (2%) Zeta. Two samples were mixtures of two variants, one of Alpha and Beta and one of Alpha and Delta. The Spearman correlation between whole-genome sequencing (WGS) and VOC-NGS was significant (P < 0.001) with perfect agreement (Kappa = 0.916) for 36/38 (94.7%) samples with VOC-NGS detecting all the known VOCs. Genotyping by VOC-NGS enables rapid screening of high-throughput clinical samples that includes the identification of VOCs and mixtures of variants, at lower cost than WGS. IMPORTANCE The manuscript described SARS-Cov-2 genotyping by VOC-NGS, which presents an ideal balance of accuracy, rapidity, and cost for detecting and globally tracking VOCs and some VOI of SARS-CoV-2. A large number of clinical samples can be tested together. Rapid introduction of new mutations at a specific site of the spike protein necessitates efficient strain detection and identification to enable choice of treatment and the application of vaccination, as well as planning public health policy.
Collapse
|
23
|
Mulder L, Carrères B, Muggli F, Zollinger A, Corthésy J, Klijn A, Togni G. A Comparative Study of Nine SARS-CoV-2 IgG Lateral Flow Assays Using Both Post-Infection and Post-Vaccination Samples. J Clin Med 2022; 11:jcm11082100. [PMID: 35456192 PMCID: PMC9032267 DOI: 10.3390/jcm11082100] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/18/2022] [Accepted: 04/05/2022] [Indexed: 12/10/2022] Open
Abstract
Background: Since the SARS-CoV-2 pandemic, lateral flow assays (LFA) detecting specific antibodies have entered the market in abundance. Despite being CE-IVD-labeled, the antigenic compounds of the assays are often unknown, the performance characteristics provided by the manufacturer are often incomplete, and the samples used to obtain the data are not detailed. Objective: To perform a comparative evaluation of nine lateral flow assays to detect IgG responses against SARS-CoV-2. For the evaluation, a carefully designed serum panel containing post-infection samples and post-vaccination (both mRNA vaccine and inactivated virus vaccine) samples was used. Results: The sensitivity of the assays overall ranged from 9 to 90.3% and the specificity ranged from 94.2 to 100%. Spike protein-containing assays performed generally better than the assays with only nucleocapsid protein. The sensitivity of some assays was higher on post-infection samples, while other assays had a higher sensitivity to post-vaccination samples. Conclusion: A comparative approach in the verification of LFAs with an adequately designed serum panel enabled the identification of the antigens used in the assays. Sensitivities differed between post-infection and post-vaccination samples, depending on the assays used. This demonstrates that the verification of assays must be performed with samples representative of the intended use of the assay.
Collapse
Affiliation(s)
- Leontine Mulder
- Clinical Laboratory, Medlon BV, P.O. Box 50000, 7500 KA Enschede, The Netherlands;
- Clinical Chemistry, Ziekenhuis Groep Twente, P.O. Box 7600, 7600 SZ Almelo, The Netherlands
| | - Benoit Carrères
- Société de Produits Nestlé S.A., Nestlé Research, Route du Jorat 57, 1000 Lausanne, Switzerland; (B.C.); (A.Z.); (J.C.); (A.K.)
| | - Franco Muggli
- Faculty of Biomedical Science, Università della Svizzera Italiana, 6900 Lugano, Switzerland;
| | - Alix Zollinger
- Société de Produits Nestlé S.A., Nestlé Research, Route du Jorat 57, 1000 Lausanne, Switzerland; (B.C.); (A.Z.); (J.C.); (A.K.)
| | - John Corthésy
- Société de Produits Nestlé S.A., Nestlé Research, Route du Jorat 57, 1000 Lausanne, Switzerland; (B.C.); (A.Z.); (J.C.); (A.K.)
| | - Adrianne Klijn
- Société de Produits Nestlé S.A., Nestlé Research, Route du Jorat 57, 1000 Lausanne, Switzerland; (B.C.); (A.Z.); (J.C.); (A.K.)
| | - Giuseppe Togni
- Unilabs Central Laboratory, 1296 Coppet, Switzerland
- Correspondence:
| |
Collapse
|
24
|
Ghosh N, Nandi S, Saha I. A review on evolution of emerging SARS-CoV-2 variants based on spike glycoprotein. Int Immunopharmacol 2022; 105:108565. [PMID: 35123183 PMCID: PMC8799522 DOI: 10.1016/j.intimp.2022.108565] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/30/2022]
Abstract
Since the inception of SARS-CoV-2 in December 2019, many variants have emerged over time. Some of these variants have resulted in transmissibility changes of the virus and may also have impact on diagnosis, therapeutics and even vaccines, thereby raising particular concerns in the scientific community. The variants which have mutations in Spike glycoprotein are the primary focus as it is the main target for neutralising antibodies. SARS-CoV-2 is known to infect human through Spike glycoprotein and uses receptor-binding domain (RBD) to bind to the ACE2 receptor in human. Thus, it is of utmost importance to study these variants and their corresponding mutations. Such 12 different important variants identified so far are B.1.1.7 (Alpha), B.1.351 (Beta), B.1.525 (Eta), B.1.427/B.1.429 (Epsilon), B.1.526 (Iota), B.1.617.1 (Kappa), B.1.617.2 (Delta), C.37 (Lambda), P.1 (Gamma), P.2 (Zeta), P.3 (Theta) and the recently discovered B.1.1.529 (Omicron). These variants have 84 unique mutations in Spike glycoprotein. To analyse such mutations, multiple sequence alignment of 77681 SARS-CoV-2 genomes of 98 countries over the period from January 2020 to July 2021 is performed followed by phylogenetic analysis. Also, characteristics of new emerging variants are elaborately discussed. The individual evolution of these mutation points and the respective variants are visualised and their characteristics are also reported. Moreover, to judge the characteristics of the non-synonymous mutation points (substitutions), their biological functions are evaluated by PolyPhen-2 while protein structural stability is evaluated using I-Mutant 2.0.
Collapse
Affiliation(s)
- Nimisha Ghosh
- Faculty of Mathematics, Informatics, and Mechanics, University of Warsaw, Warsaw, Poland; Department of Computer Science and Information Technology, Institute of Technical Education and Research, Siksha `O' Anusandhan (Deemed to be) University, Bhubaneswar, Odisha, India
| | - Suman Nandi
- Department of Computer Science and Engineering, National Institute of Technical Teachers' Training and Research, Kolkata, West Bengal, India
| | - Indrajit Saha
- Department of Computer Science and Engineering, National Institute of Technical Teachers' Training and Research, Kolkata, West Bengal, India.
| |
Collapse
|
25
|
Quer J, Colomer-Castell S, Campos C, Andrés C, Piñana M, Cortese MF, González-Sánchez A, Garcia-Cehic D, Ibáñez M, Pumarola T, Rodríguez-Frías F, Antón A, Tabernero D. Next-Generation Sequencing for Confronting Virus Pandemics. Viruses 2022; 14:600. [PMID: 35337007 PMCID: PMC8950049 DOI: 10.3390/v14030600] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/01/2022] [Accepted: 03/10/2022] [Indexed: 02/06/2023] Open
Abstract
Virus pandemics have happened, are happening and will happen again. In recent decades, the rate of zoonotic viral spillover into humans has accelerated, mirroring the expansion of our global footprint and travel network, including the expansion of viral vectors and the destruction of natural spaces, bringing humans closer to wild animals. Once viral cross-species transmission to humans occurs, transmission cannot be stopped by cement walls but by developing barriers based on knowledge that can prevent or reduce the effects of any pandemic. Controlling a local transmission affecting few individuals is more efficient that confronting a community outbreak in which infections cannot be traced. Genetic detection, identification, and characterization of infectious agents using next-generation sequencing (NGS) has been proven to be a powerful tool allowing for the development of fast PCR-based molecular assays, the rapid development of vaccines based on mRNA and DNA, the identification of outbreaks, transmission dynamics and spill-over events, the detection of new variants and treatment of vaccine resistance mutations, the development of direct-acting antiviral drugs, the discovery of relevant minority variants to improve knowledge of the viral life cycle, strengths and weaknesses, the potential for becoming dominant to take appropriate preventive measures, and the discovery of new routes of viral transmission.
Collapse
Affiliation(s)
- Josep Quer
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (S.C.-C.); (C.C.); (D.G.-C.); (M.I.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain; (M.F.C.); (F.R.-F.); (D.T.)
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), UAB Campus, Plaça Cívica, 08193 Bellaterra, Spain
| | - Sergi Colomer-Castell
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (S.C.-C.); (C.C.); (D.G.-C.); (M.I.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain; (M.F.C.); (F.R.-F.); (D.T.)
| | - Carolina Campos
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (S.C.-C.); (C.C.); (D.G.-C.); (M.I.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain; (M.F.C.); (F.R.-F.); (D.T.)
| | - Cristina Andrés
- Microbiology Department, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (C.A.); (M.P.); (A.G.-S.); (T.P.)
| | - Maria Piñana
- Microbiology Department, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (C.A.); (M.P.); (A.G.-S.); (T.P.)
| | - Maria Francesca Cortese
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain; (M.F.C.); (F.R.-F.); (D.T.)
- Clinical Biochemistry Research Group, Biochemistry Department, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Alejandra González-Sánchez
- Microbiology Department, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (C.A.); (M.P.); (A.G.-S.); (T.P.)
| | - Damir Garcia-Cehic
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (S.C.-C.); (C.C.); (D.G.-C.); (M.I.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain; (M.F.C.); (F.R.-F.); (D.T.)
| | - Marta Ibáñez
- Liver Diseases-Viral Hepatitis, Liver Unit, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (S.C.-C.); (C.C.); (D.G.-C.); (M.I.)
| | - Tomàs Pumarola
- Microbiology Department, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (C.A.); (M.P.); (A.G.-S.); (T.P.)
- Microbiology Department, Universitat Autònoma de Barcelona (UAB), UAB Campus, Plaça Cívica, 08193 Bellaterra, Spain
| | - Francisco Rodríguez-Frías
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain; (M.F.C.); (F.R.-F.); (D.T.)
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona (UAB), UAB Campus, Plaça Cívica, 08193 Bellaterra, Spain
- Clinical Biochemistry Research Group, Biochemistry Department, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| | - Andrés Antón
- Microbiology Department, Vall d’Hebron Institut of Research (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain; (C.A.); (M.P.); (A.G.-S.); (T.P.)
- Microbiology Department, Universitat Autònoma de Barcelona (UAB), UAB Campus, Plaça Cívica, 08193 Bellaterra, Spain
| | - David Tabernero
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Instituto de Salud Carlos III, Av. Monforte de Lemos 3-5, 28029 Madrid, Spain; (M.F.C.); (F.R.-F.); (D.T.)
- Microbiology Departments, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain
| |
Collapse
|
26
|
Cedro-Tanda A, Gómez-Romero L, de Anda-Jauregui G, Garnica-López D, Alfaro-Mora Y, Sánchez-Xochipa S, García-García EF, Mendoza-Vargas A, Frías-Jiménez EJ, Moreno B, Campos-Romero A, Moreno-Camacho JL, Alcantar-Fernández J, Ortíz-Ramírez J, Benitez-González M, Trejo-González R, Aguirre-Chavarría D, Núñez-Martínez ME, Uribe-Figueroa L, Angulo O, Ruiz R, Hidalgo-Miranda A, Herrera LA. Early Genomic, Epidemiological, and Clinical Description of the SARS-CoV-2 Omicron Variant in Mexico City. Viruses 2022; 14:545. [PMID: 35336952 PMCID: PMC8950183 DOI: 10.3390/v14030545] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 02/26/2022] [Accepted: 03/02/2022] [Indexed: 01/27/2023] Open
Abstract
Omicron is the most mutated SARS-CoV-2 variant-a factor that can affect transmissibility, disease severity, and immune evasiveness. Its genomic surveillance is important in cities with millions of inhabitants and an economic center, such as Mexico City. Results. From 16 November to 31 December 2021, we observed an increase of 88% in Omicron prevalence in Mexico City. We explored the R346K substitution, prevalent in 42% of Omicron variants, known to be associated with immune escape by monoclonal antibodies. In a phylogenetic analysis, we found several independent exchanges between Mexico and the world, and there was an event followed by local transmission that gave rise to most of the Omicron diversity in Mexico City. A haplotype analysis revealed that there was no association between haplotype and vaccination status. Among the 66% of patients who have been vaccinated, no reported comorbidities were associated with Omicron; the presence of odynophagia and the absence of dysgeusia were significant predictor symptoms for Omicron, and the RT-qPCR Ct values were lower for Omicron. Conclusions. Genomic surveillance is key to detecting the emergence and spread of SARS-CoV-2 variants in a timely manner, even weeks before the onset of an infection wave, and can inform public health decisions and detect the spread of any mutation that may affect therapeutic efficacy.
Collapse
Affiliation(s)
- Alberto Cedro-Tanda
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
| | - Laura Gómez-Romero
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
| | - Guillermo de Anda-Jauregui
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
- Researchers for Mexico (Previously Cátedras CONACYT para Jóvenes Investigadores), Av. de los Insurgentes Sur 1582, Crédito Constructor, Benito Juárez, Mexico City 03940, Mexico
- Centro de Ciencias de la Complejidad, Universidad Nacional Autónoma de México (UNAM), Circuito Centro Cultural S/N, Cd. Universitaria, Delegación Coyoacán, Mexico City 04510, Mexico
| | - Dora Garnica-López
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
| | - Yair Alfaro-Mora
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
| | - Sonia Sánchez-Xochipa
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
| | - Eulices F. García-García
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
| | - Alfredo Mendoza-Vargas
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
| | - Emmanuel J. Frías-Jiménez
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
| | - Bernardo Moreno
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
| | - Abraham Campos-Romero
- Innovation and Research Department, Salud Digna, Culiacan 80000, Mexico; (A.C.-R.); (J.A.-F.)
| | | | | | - Jesús Ortíz-Ramírez
- Hospital General Ajusco Medio, Secretaría de Salud de la Ciudad de México (SEDESA), Encinos 41, Miguel Hidalgo 4ta Secc, Tlalpan, Mexico City 14250, Mexico; (J.O.-R.); (M.B.-G.)
| | - Mariana Benitez-González
- Hospital General Ajusco Medio, Secretaría de Salud de la Ciudad de México (SEDESA), Encinos 41, Miguel Hidalgo 4ta Secc, Tlalpan, Mexico City 14250, Mexico; (J.O.-R.); (M.B.-G.)
| | - Roxana Trejo-González
- Centro Médico ABC, Av. Carlos Fernández Graef 154, Santa Fe, Contadero, Cuajimalpa de Morelos, Mexico City 05330, Mexico; (R.T.-G.); (D.A.-C.); (M.E.N.-M.)
| | - Daniel Aguirre-Chavarría
- Centro Médico ABC, Av. Carlos Fernández Graef 154, Santa Fe, Contadero, Cuajimalpa de Morelos, Mexico City 05330, Mexico; (R.T.-G.); (D.A.-C.); (M.E.N.-M.)
| | - Marcela E. Núñez-Martínez
- Centro Médico ABC, Av. Carlos Fernández Graef 154, Santa Fe, Contadero, Cuajimalpa de Morelos, Mexico City 05330, Mexico; (R.T.-G.); (D.A.-C.); (M.E.N.-M.)
| | - Laura Uribe-Figueroa
- Laboratorio Arion Genética, Margaritas 440-Bis, Hacienda de Guadalupe Chimalistac, Chimalistac, Álvaro Obregón, Mexico City 01050, Mexico;
| | - Ofelia Angulo
- Secretaría de Educación, Ciencia, Tecnología e Innovación de la Ciudad de México (SECTEI), Av Chapultepec 49, Colonia Centro, Cuauhtémoc, Mexico City 06010, Mexico; (O.A.); (R.R.)
| | - Rosaura Ruiz
- Secretaría de Educación, Ciencia, Tecnología e Innovación de la Ciudad de México (SECTEI), Av Chapultepec 49, Colonia Centro, Cuauhtémoc, Mexico City 06010, Mexico; (O.A.); (R.R.)
| | - Alfredo Hidalgo-Miranda
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
| | - Luis A. Herrera
- Instituto Nacional de Medicina Genómica, Periferico Sur 4809, Arenal Tepepan, Mexico City 14610, Mexico; (A.C.-T.); (L.G.-R.); (G.d.A.-J.); (D.G.-L.); (Y.A.-M.); (S.S.-X.); (E.F.G.-G.); (A.M.-V.); (E.J.F.-J.); (B.M.)
- Unidad de Investigación Biomédica en Cáncer, Instituto Nacional de Cancerología-Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México (UNAM), Av. San Fernando 22, Belisario Domínguez Secc 16, Tlalpan, Mexico City 14080, Mexico
| |
Collapse
|
27
|
Malhotra HS, Dandu H, Garg RK. Expeditious quest for the best vaccine: the game is on. QJM 2022; 115:122-123. [PMID: 34014314 DOI: 10.1093/qjmed/hcab146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- H S Malhotra
- Department of Neurology, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - H Dandu
- Department of Infectious Diseases, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| | - R K Garg
- Department of Neurology, King George's Medical University, Lucknow, Uttar Pradesh 226003, India
| |
Collapse
|
28
|
Efficacy and Safety of a Phytopharmaceutical Drug Derived from Cocculus hirsutus in Adults with Moderate COVID-19: a Phase 2, Open-label, Multicenter, Randomized Controlled Trial. Infect Dis Ther 2022; 11:807-826. [PMID: 35179709 PMCID: PMC8855350 DOI: 10.1007/s40121-022-00604-0] [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] [Received: 11/10/2021] [Accepted: 02/01/2022] [Indexed: 12/12/2022] Open
Abstract
Introduction There is an urgent need for an effective, oral therapy for COVID-19. Purified aqueous extract of Cocculus hirsutus (AQCH) has shown robust antiviral activity in in vitro studies. We aimed to evaluate the efficacy and safety of AQCH plus standard of care in hospitalized patients with moderate COVID-19. Methods In an open-label, multicenter, randomized controlled trial conducted in India, eligible patients (aged 18–75 years) were randomized (1:1) to receive AQCH 400 mg orally three times a day plus standard of care (AQCH group) or standard of care alone (control group) for 10 days. Primary endpoint was the proportion of patients showing clinical improvement by day 14. Time to clinical improvement, time to viral clearance, and duration of hospitalization were secondary endpoints. Results A total of 210 patients were randomized. By day 14 most patients in both groups showed clinical improvement [difference − 0.01 (95% CI − 0.07 to 0.05); p = 1.0]. Median time to clinical improvement was 8 days (IQR 8–11) in the AQCH group versus 11 days (IQR 8–11) in the control group [HR 1.27 (95% CI 0.95–1.71); p = 0.032]. Time to viral clearance and duration of hospitalization were also significantly shorter in the AQCH group (p = 0.0002 and p = 0.016, respectively). AQCH was well tolerated, with no safety concerns identified. Conclusions AQCH significantly reduced time to clinical improvement, time to viral clearance, and duration of hospitalization. In a pandemic, this has significant potential to decrease healthcare resource utilization and increase hospital bed availability. Further investigation of the therapeutic potential of AQCH in patients with COVID-19 is warranted. Trial Registration Clinical Trials Registry – India (CTRI/2020/05/025397). Supplementary Information The online version contains supplementary material available at 10.1007/s40121-022-00604-0.
Collapse
|
29
|
Wang Y, Chen XY, Yang L, Yao Q, Chen KP. Human SARS-CoV-2 has evolved to increase U content and reduce genome size. Int J Biol Macromol 2022; 204:356-363. [PMID: 35149094 PMCID: PMC8824384 DOI: 10.1016/j.ijbiomac.2022.02.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 02/06/2022] [Accepted: 02/07/2022] [Indexed: 12/30/2022]
Abstract
Infections caused by SARS-CoV-2 have brought great harm to human health. After transmission for over two years, SARS-CoV-2 has diverged greatly and formed dozens of different lineages. Understanding the trend of its genome evolution could help foresee difficulties in controlling transmission of the virus. In this study, we conducted an extensive monthly survey and in-depth analysis on variations of nucleotide, amino acid and codon numbers in 311,260 virus samples collected till January 2022. The results demonstrate that the evolution of SARS-CoV-2 is toward increasing U-content and reducing genome-size. C, G and A to U mutations have all contributed to this U-content increase. Mutations of C, G and A at codon position 1, 2 or 3 have no significant difference in most SARS-CoV-2 lineages. Current viruses are more cryptic and more efficient in replication, and are thus less virulent yet more infectious. Delta and Omicron variants have high mutability over other lineages, bringing new threat to human health. This trend of genome evolution may provide a clue for tracing the origin of SARS-CoV-2, because ancestral viruses should have lower U-content and probably bigger genome-size.
Collapse
Affiliation(s)
- Yong Wang
- School of Food and Biological Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China.
| | - Xin-Yu Chen
- School of Food and Biological Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China
| | - Liu Yang
- School of Food and Biological Engineering, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China
| | - Qin Yao
- School of Life Sciences, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China.
| | - Ke-Ping Chen
- School of Life Sciences, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China.
| |
Collapse
|
30
|
Utility of a commercial RT-qPCR assay to detect SARS-CoV-2 gene variations as an indicator of lineages. J Virol Methods 2022; 300:114428. [PMID: 34906667 PMCID: PMC8665652 DOI: 10.1016/j.jviromet.2021.114428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND The World Health Organization (WHO) recommended RT-qPCR tests as the reference technique for SARS-CoV-2 molecular detection, however with the rapid spread of the infection, mutations in specific RT-qPCR target regions have been widely described could allow the presumptive identification. OBJECTIVE In this study, we evaluated the analytical performance of the Allplex™SARS-CoV-2/FluA/FluB/RSV assay for the additional presumptive identification of SARS-CoV-2 variants in a real-life setting. RESULTS We observed gene-specific changes in the cycle threshold (Ct) of the N and RdRp genes compared with the Ct yielded for the S gene when the SARS-CoV-2 testing was performed Allplex™SARS-CoV-2/FluA/FluB/RSV assay. Seventeen samples showed Ct variations in the N and/or RdRp. In 10 cases, the N gene was affected, delayed or negative and in 14 cases, the RdRp gene showed a delay or negative concerning the S gene. A delay in the Ct of both genes (RdRp and N) was observed in six cases. Sequencing determined that all samples identified as B.1.1.7 showed changes in the PCR curves of the N and RdRp. However, samples identified as B.1.177 only showed variations for the RdRp gene. CONCLUSIONS Allplex™SARS-CoV-2/FluA/FluB/RSV assay, the diagnosis could presumably allow the rapid assignment of lineages B.1.1.7 and B.1.177, and emphasizes the importance of exhaustive surveillance for circulating variants of the SARS-CoV-2 virus to reduce community transmission.
Collapse
|
31
|
Wang X, Luo J, Wen Z, Shuai L, Wang C, Zhong G, He X, Cao H, Liu R, Ge J, Hua R, Sun Z, Wang X, Wang J, Bu Z. Diltiazem inhibits SARS-CoV-2 cell attachment and internalization and decreases the viral infection in mouse lung. PLoS Pathog 2022; 18:e1010343. [PMID: 35176124 PMCID: PMC8890723 DOI: 10.1371/journal.ppat.1010343] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/02/2022] [Accepted: 02/05/2022] [Indexed: 12/20/2022] Open
Abstract
The continuous emergence of severe acute respiratory coronavirus 2 (SARS-CoV-2) variants and the increasing number of breakthrough infection cases among vaccinated people support the urgent need for research and development of antiviral drugs. Viral entry is an intriguing target for antiviral drug development. We found that diltiazem, a blocker of the L-type calcium channel Cav1.2 pore-forming subunit (Cav1.2 α1c) and an FDA-approved drug, inhibits the binding and internalization of SARS-CoV-2, and decreases SARS-CoV-2 infection in cells and mouse lung. Cav1.2 α1c interacts with SARS-CoV-2 spike protein and ACE2, and affects the attachment and internalization of SARS-CoV-2. Our finding suggests that diltiazem has potential as a drug against SARS-CoV-2 infection and that Cav1.2 α1c is a promising target for antiviral drug development for COVID-19.
Collapse
Affiliation(s)
- Xinxin Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Jie Luo
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Zhiyuan Wen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Lei Shuai
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Chong Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Gongxun Zhong
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Xijun He
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Huizhen Cao
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Renqiang Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Jinying Ge
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Ronghong Hua
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Ziruo Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Xijun Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Jinliang Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
| | - Zhigao Bu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, People’s Republic of China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, P. R. China
| |
Collapse
|
32
|
Forni D, Cagliani R, Pontremoli C, Clerici M, Sironi M. The substitution spectra of coronavirus genomes. Brief Bioinform 2022; 23:bbab382. [PMID: 34518866 PMCID: PMC8499949 DOI: 10.1093/bib/bbab382] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/23/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has triggered an unprecedented international effort to sequence complete viral genomes. We leveraged this wealth of information to characterize the substitution spectrum of SARS-CoV-2 and to compare it with those of other human and animal coronaviruses. We show that, once nucleotide composition is taken into account, human and most animal coronaviruses display a mutation spectrum dominated by C to U and G to U substitutions, a feature that is not shared by other positive-sense RNA viruses. However, the proportions of C to U and G to U substitutions tend to decrease as divergence increases, suggesting that, whatever their origin, a proportion of these changes is subsequently eliminated by purifying selection. Analysis of the sequence context of C to U substitutions showed little evidence of apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC)-mediated editing and such contexts were similar for SARS-CoV-2 and Middle East respiratory syndrome coronavirus sampled from different hosts, despite different repertoires of APOBEC3 proteins in distinct species. Conversely, we found evidence that C to U and G to U changes affect CpG dinucleotides at a frequency higher than expected. Whereas this suggests ongoing selective reduction of CpGs, this effect alone cannot account for the substitution spectra. Finally, we show that, during the first months of SARS-CoV-2 pandemic spread, the frequency of both G to U and C to U substitutions increased. Our data suggest that the substitution spectrum of SARS-CoV-2 is determined by an interplay of factors, including intrinsic biases of the replication process, avoidance of CpG dinucleotides and other constraints exerted by the new host.
Collapse
Affiliation(s)
- Diego Forni
- Scientific Institute IRCCS E. MEDEA, Bioinformatics, Bosisio Parini, Italy
| | - Rachele Cagliani
- Scientific Institute IRCCS E. MEDEA, Bioinformatics, Bosisio Parini, Italy
| | - Chiara Pontremoli
- Scientific Institute IRCCS E. MEDEA, Bioinformatics, Bosisio Parini, Italy
| | - Mario Clerici
- Department of Physiopathology and Transplantation, University of Milan, Milan, Italy
- Don C. Gnocchi Foundation ONLUS, IRCCS, Milan, Italy
| | - Manuela Sironi
- Scientific Institute IRCCS E. MEDEA, Bioinformatics, Bosisio Parini, Italy
| |
Collapse
|
33
|
Villafañe L, Vaulet LG, Viere FM, Klepp LI, Forrellad MA, Bigi MM, Romano MI, Magistrelli G, Fermepin MR, Bigi F. Development and evaluation of a low cost IgG ELISA test based in RBD protein for COVID-19. J Immunol Methods 2022; 500:113182. [PMID: 34762914 PMCID: PMC8574101 DOI: 10.1016/j.jim.2021.113182] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 09/08/2021] [Accepted: 11/03/2021] [Indexed: 01/24/2023]
Abstract
Serology tests for SARS-CoV-2 have proven to be important tools to fight against the COVID-19 pandemic. These serological tests can be used in low-income and remote areas for patient contact tracing, epidemiologic studies and vaccine efficacy evaluations. In this study, we used a semi-stable mammalian episomal expression system to produce high quantities of the receptor-binding domain-RBD of SARS-CoV-2 in a simple and very economical way. The recombinant antigen was tested in an in-house IgG ELISA for COVID-19 with a panel of human sera. A performance comparison of this serology test with a commercial test based on the full-length spike protein showed 100% of concordance between tests. Thus, this serological test can be an attractive and inexpensive option in scenarios of limited resources to face the COVID-19 pandemic.
Collapse
Affiliation(s)
- Luciana Villafañe
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), UEDD INTA-CONICET, CICVyA, Instituto Nacional de Tecnología Agropecuaria (Institute of Biotechnology, NationalInstitute of AgriculturalTechnology), Argentina.
| | - Lucía Gallo Vaulet
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica, Cátedra de Microbiología Clínica, Argentina; Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina
| | | | - Laura I Klepp
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), UEDD INTA-CONICET, CICVyA, Instituto Nacional de Tecnología Agropecuaria (Institute of Biotechnology, NationalInstitute of AgriculturalTechnology), Argentina.
| | - Marina A Forrellad
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), UEDD INTA-CONICET, CICVyA, Instituto Nacional de Tecnología Agropecuaria (Institute of Biotechnology, NationalInstitute of AgriculturalTechnology), Argentina.
| | - María M Bigi
- Universidad de Buenos Aires, Instituto De Investigaciones Biomédicas (INBIOMED), Argentina.
| | - María I Romano
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), UEDD INTA-CONICET, CICVyA, Instituto Nacional de Tecnología Agropecuaria (Institute of Biotechnology, NationalInstitute of AgriculturalTechnology), Argentina.
| | | | - Marcelo Rodríguez Fermepin
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica, Cátedra de Microbiología Clínica, Argentina; Universidad de Buenos Aires, Instituto de Fisiopatología y Bioquímica Clínica (INFIBIOC), Argentina.
| | - Fabiana Bigi
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO), UEDD INTA-CONICET, CICVyA, Instituto Nacional de Tecnología Agropecuaria (Institute of Biotechnology, NationalInstitute of AgriculturalTechnology), Argentina.
| |
Collapse
|
34
|
Dubey A, Choudhary S, Kumar P, Tomar S. Emerging SARS-CoV-2 Variants: Genetic Variability and Clinical Implications. Curr Microbiol 2021; 79:20. [PMID: 34905108 PMCID: PMC8669229 DOI: 10.1007/s00284-021-02724-1] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/08/2021] [Indexed: 12/12/2022]
Abstract
The sudden rise in COVID-19 cases in 2020 and the incessant emergence of fast-spreading variants have created an alarming situation worldwide. Besides the continuous advancements in the design and development of vaccines to combat this deadly pandemic, new variants are frequently reported, possessing mutations that rapidly outcompeted an existing population of circulating variants. As concerns grow about the effects of mutations on the efficacy of vaccines, increased transmissibility, immune escape, and diagnostic failures are few other apprehensions liable for more deadly waves of COVID-19. Although the phenomenon of antigenic drift in new variants of SARS-CoV-2 is still not validated, it is conceived that the virus is acquiring new mutations as a fitness advantage for rapid transmission or to overcome immunological resistance of the host cell. Considerable evolution of SARS-CoV-2 has been observed since its first appearance in 2019, and despite the progress in sequencing efforts to characterize the mutations, their impacts in many variants have not been analyzed. The present article provides a substantial review of literature explaining the emerging variants of SARS-CoV-2 circulating globally, key mutations in viral genome, and the possible impacts of these new mutations on prevention and therapeutic strategies currently administered to combat this pandemic. Rising infections, mortalities, and hospitalizations can possibly be tackled through mass vaccination, social distancing, better management of available healthcare infrastructure, and by prioritizing genome sequencing for better serosurveillance studies and community tracking.
Collapse
Affiliation(s)
- Aakriti Dubey
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Shweta Choudhary
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, 247667, India.
| |
Collapse
|
35
|
Wang J, Yang G, Wang X, Wen Z, Shuai L, Luo J, Wang C, Sun Z, Liu R, Ge J, He X, Hua R, Wang X, Yang X, Chen W, Zhong G, Bu Z. SARS-CoV-2 uses metabotropic glutamate receptor subtype 2 as an internalization factor to infect cells. Cell Discov 2021; 7:119. [PMID: 34903715 PMCID: PMC8668938 DOI: 10.1038/s41421-021-00357-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 11/18/2021] [Indexed: 12/23/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) uses angiotensin-converting enzyme 2 (ACE2) as a binding receptor to enter cells via clathrin-mediated endocytosis (CME). However, receptors involved in other steps of SARS-CoV-2 infection remain largely unknown. Here, we found that metabotropic glutamate receptor subtype 2 (mGluR2) is an internalization factor for SARS-CoV-2. Our results show that mGluR2 directly interacts with the SARS-CoV-2 spike protein and that knockdown of mGluR2 decreases internalization of SARS-CoV-2 but not cell binding. Further, mGluR2 is uncovered to cooperate with ACE2 to facilitate SARS-CoV-2 internalization through CME and mGluR2 knockout in mice abolished SARS-CoV-2 infection in the nasal turbinates and significantly reduced viral infection in the lungs. Notably, mGluR2 is also important for SARS-CoV spike protein- and Middle East respiratory syndrome coronavirus spike protein-mediated internalization. Thus, our study identifies a novel internalization factor used by SARS-CoV-2 and opens a new door for antiviral development against coronavirus infection.
Collapse
Affiliation(s)
- Jinliang Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Guan Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Xinxin Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Zhiyuan Wen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Lei Shuai
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Jie Luo
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Chong Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Ziruo Sun
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Renqiang Liu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Jinying Ge
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Xijun He
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Ronghong Hua
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Xijun Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Xiao Yang
- State Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing, China
| | - Weiye Chen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Gongxun Zhong
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China
| | - Zhigao Bu
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, Heilongjiang, China.
| |
Collapse
|
36
|
Rodríguez-Frías F, Quer J, Tabernero D, Cortese MF, Garcia-Garcia S, Rando-Segura A, Pumarola T. Microorganisms as Shapers of Human Civilization, from Pandemics to Even Our Genomes: Villains or Friends? A Historical Approach. Microorganisms 2021; 9:2518. [PMID: 34946123 PMCID: PMC8708650 DOI: 10.3390/microorganisms9122518] [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] [Received: 10/25/2021] [Revised: 11/23/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023] Open
Abstract
Universal history is characterized by continuous evolution, in which civilizations are born and die. This evolution is associated with multiple factors, among which the role of microorganisms is often overlooked. Viruses and bacteria have written or decisively contributed to terrible episodes of history, such as the Black Death in 14th century Europe, the annihilation of pre-Columbian American civilizations, and pandemics such as the 1918 Spanish flu or the current COVID-19 pandemic caused by the coronavirus SARS-CoV-2. Nevertheless, it is clear that we could not live in a world without these tiny beings. Endogenous retroviruses have been key to our evolution and for the regulation of gene expression, and the gut microbiota helps us digest compounds that we could not otherwise process. In addition, we have used microorganisms to preserve or prepare food for millennia and more recently to obtain drugs such as antibiotics or to develop recombinant DNA technologies. Due to the enormous importance of microorganisms for our survival, they have significantly influenced the population genetics of different human groups. This paper will review the role of microorganisms as "villains" who have been responsible for tremendous mortality throughout history but also as "friends" who help us survive and evolve.
Collapse
Affiliation(s)
- Francisco Rodríguez-Frías
- Clinical Biochemistry Research Group, Department of Biochemistry, Vall d’Hebron Institut Recerca-Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (M.F.C.); (S.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Liver Pathology Unit, Departments of Biochemistry and Microbiology, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
| | - Josep Quer
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, 28029 Madrid, Spain;
- Liver Unit, Liver Disease Laboratory-Viral Hepatitis, Vall d’Hebron Institut Recerca, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain
| | - David Tabernero
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Maria Francesca Cortese
- Clinical Biochemistry Research Group, Department of Biochemistry, Vall d’Hebron Institut Recerca-Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (M.F.C.); (S.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Selene Garcia-Garcia
- Clinical Biochemistry Research Group, Department of Biochemistry, Vall d’Hebron Institut Recerca-Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain; (M.F.C.); (S.G.-G.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Instituto de Salud Carlos III, 28029 Madrid, Spain;
| | - Ariadna Rando-Segura
- Liver Pathology Unit, Departments of Biochemistry and Microbiology, Hospital Universitari Vall d’Hebron, Universitat Autònoma de Barcelona, 08035 Barcelona, Spain;
- Department of Microbiology, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain;
| | - Tomas Pumarola
- Department of Microbiology, Hospital Universitari Vall d’Hebron, Vall d’Hebron Barcelona Hospital Campus, Passeig Vall d’Hebron 119-129, 08035 Barcelona, Spain;
| |
Collapse
|
37
|
Gupta D, Sharma P, Singh M, Kumar M, Ethayathulla AS, Kaur P. Structural and functional insights into the spike protein mutations of emerging SARS-CoV-2 variants. Cell Mol Life Sci 2021; 78:7967-7989. [PMID: 34731254 PMCID: PMC11073194 DOI: 10.1007/s00018-021-04008-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/20/2021] [Accepted: 10/22/2021] [Indexed: 02/07/2023]
Abstract
Since the emergence of the first case of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus (SARS-CoV-2), the viral genome has constantly undergone rapid mutations for better adaptation in the host system. These newer mutations have given rise to several lineages/ variants of the virus that have resulted in high transmission and virulence rates compared to the previously circulating variants. Owing to this, the overall caseload and related mortality have tremendously increased globally to > 233 million infections and > 4.7 million deaths as of Sept. 28th, 2021. SARS-CoV-2, Spike (S) protein binds to host cells by recognizing human angiotensin-converting enzyme 2 (hACE2) receptor. The viral S protein contains S1 and S2 domains that constitute the binding and fusion machinery, respectively. Structural analysis of viral S protein reveals that the virus undergoes conformational flexibility and dynamicity to interact with the hACE2 receptor. The SARS-CoV-2 variants and mutations might be associated with affecting the conformational plasticity of S protein, potentially linked to its altered affinity, infectivity, and immunogenicity. This review focuses on the current circulating variants of SARS-CoV-2 and the structure-function analysis of key S protein mutations linked with increased affinity, higher infectivity, enhanced transmission rates, and immune escape against this infection.
Collapse
Affiliation(s)
- Deepali Gupta
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi,, Delhi, 110029, India
| | - Priyanka Sharma
- Department of Microbiology, All India Institute of Medical Sciences, New Delhi,, Delhi, 110029, India
| | - Mandeep Singh
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi,, Delhi, 110029, India
| | - Mukesh Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi,, Delhi, 110029, India
| | - A S Ethayathulla
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi,, Delhi, 110029, India
| | - Punit Kaur
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi,, Delhi, 110029, India.
| |
Collapse
|
38
|
Limaye S, Kasibhatla SM, Ramtirthkar M, Kinikar M, Kale MM, Kulkarni-Kale U. Circulation and Evolution of SARS-CoV-2 in India: Let the Data Speak. Viruses 2021; 13:2238. [PMID: 34835044 PMCID: PMC8619538 DOI: 10.3390/v13112238] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/28/2021] [Accepted: 10/28/2021] [Indexed: 01/08/2023] Open
Abstract
The COVID-19 pandemic is a global challenge that impacted 200+ countries. India ranks in the second and third positions in terms of number of reported cases and deaths. Being a populous country with densely packed cities, SARS-CoV-2 spread exponentially. India sequenced ≈0.14% isolates from confirmed cases for pandemic surveillance and contributed ≈1.58% of complete genomes sequenced globally. This study was designed to map the circulating lineage diversity and to understand the evolution of SARS-CoV-2 in India using comparative genomics and population genetics approaches. Despite varied sequencing coverage across Indian States and Union Territories, isolates belonging to variants of concern (VoC) and variants of interest (VoI) circulated, persisted, and diversified during the first seventeen months of the pandemic. Delta and Kappa lineages emerged in India and spread globally. The phylogenetic tree shows lineage-wise monophyletic clusters of VoCs/VoIs and diversified tree topologies for non-VoC/VoI lineages designated as 'Others' in this study. Evolutionary dynamics analyses substantiate a lack of spatio-temporal clustering, which is indicative of multiple global and local introductions. Sites under positive selection and significant variations in spike protein corroborate with the constellation of mutations to be monitored for VoC/VoI as well as substitutions that are characteristic of functions with implications in virus-host interactions, differential glycosylation, immune evasion, and escape from neutralization.
Collapse
Affiliation(s)
- Sanket Limaye
- Bioinformatics Centre, Savitribai Phule Pune University (Formerly University of Pune), Pune 411007, India; (S.L.); (S.M.K.); (M.K.)
| | - Sunitha M. Kasibhatla
- Bioinformatics Centre, Savitribai Phule Pune University (Formerly University of Pune), Pune 411007, India; (S.L.); (S.M.K.); (M.K.)
- HPC-Medical & Bioinformatics Applications Group, Centre for Development of Advanced Computing, Pune 411008, India
| | - Mukund Ramtirthkar
- Department of Statistics, Savitribai Phule Pune University (Formerly University of Pune), Pune 411007, India; (M.R.); (M.M.K.)
| | - Meenal Kinikar
- Bioinformatics Centre, Savitribai Phule Pune University (Formerly University of Pune), Pune 411007, India; (S.L.); (S.M.K.); (M.K.)
| | - Mohan M. Kale
- Department of Statistics, Savitribai Phule Pune University (Formerly University of Pune), Pune 411007, India; (M.R.); (M.M.K.)
| | - Urmila Kulkarni-Kale
- Bioinformatics Centre, Savitribai Phule Pune University (Formerly University of Pune), Pune 411007, India; (S.L.); (S.M.K.); (M.K.)
| |
Collapse
|
39
|
Ghosh N, Saha I, Sharma N. Interactome of human and SARS-CoV-2 proteins to identify human hub proteins associated with comorbidities. Comput Biol Med 2021; 138:104889. [PMID: 34655901 PMCID: PMC8492901 DOI: 10.1016/j.compbiomed.2021.104889] [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] [Received: 05/31/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 02/06/2023]
Abstract
SARS-CoV-2 has a higher chance of progression in adults of any age with certain underlying health conditions or comorbidities like cancer, neurological diseases and in certain cases may even lead to death. Like other viruses, SARS-CoV-2 also interacts with host proteins to pave its entry into host cells. Therefore, to understand the behaviour of SARS-CoV-2 and design of effective antiviral drugs, host-virus protein-protein interactions (PPIs) can be very useful. In this regard, we have initially created a human-SARS-CoV-2 PPI database from existing works in the literature which has resulted in 7085 unique PPIs. Subsequently, we have identified at most 10 proteins with highest degrees viz. hub proteins from interacting human proteins for individual virus protein. The identification of these hub proteins is important as they are connected to most of the other human proteins. Consequently, when they get affected, the potential diseases are triggered in the corresponding pathways, thereby leading to comorbidities. Furthermore, the biological significance of the identified hub proteins is shown using KEGG pathway and GO enrichment analysis. KEGG pathway analysis is also essential for identifying the pathways leading to comorbidities. Among others, SARS-CoV-2 proteins viz. NSP2, NSP5, Envelope and ORF10 interacting with human hub proteins like COX4I1, COX5A, COX5B, NDUFS1, CANX, HSP90AA1 and TP53 lead to comorbidities. Such comorbidities are Alzheimer, Parkinson, Huntington, HTLV-1 infection, prostate cancer and viral carcinogenesis. Subsequently, using Enrichr tool possible repurposable drugs which target the human hub proteins are reported in this paper as well. Therefore, this work provides a consolidated study for human-SARS-CoV-2 protein interactions to understand the relationship between comorbidity and hub proteins so that it may pave the way for the development of anti-viral drugs.
Collapse
Affiliation(s)
- Nimisha Ghosh
- Department of Computer Science and Information Technology, Institute of Technical Education and Research, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, India; Faculty of Mathematics, Informatics and Mechanics, University of Warsaw, Warsaw, Poland
| | - Indrajit Saha
- Department of Computer Science and Engineering, National Institute of Technical Teachers' Training and Research, Kolkata, West Bengal, India.
| | - Nikhil Sharma
- Department of Electronics and Communication Engineering, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, India
| |
Collapse
|
40
|
A Single Dose of a Hybrid hAdV5-Based Anti-COVID-19 Vaccine Induces a Long-Lasting Immune Response and Broad Coverage against VOC. Vaccines (Basel) 2021; 9:vaccines9101106. [PMID: 34696219 PMCID: PMC8537385 DOI: 10.3390/vaccines9101106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/22/2021] [Accepted: 09/24/2021] [Indexed: 12/18/2022] Open
Abstract
Most approved vaccines against COVID-19 have to be administered in a prime/boost regimen. We engineered a novel vaccine based on a chimeric human adenovirus 5 (hAdV5) vector. The vaccine (named CoroVaxG.3) is based on three pillars: (i) high expression of Spike to enhance its immunodominance by using a potent promoter and an mRNA stabilizer; (ii) enhanced infection of muscle and dendritic cells by replacing the fiber knob domain of hAdV5 by hAdV3; (iii) use of Spike stabilized in a prefusion conformation. The transduction with CoroVaxG.3-expressing Spike (D614G) dramatically enhanced the Spike expression in human muscle cells, monocytes and dendritic cells compared to CoroVaxG.5 that expressed the native fiber knob domain. A single dose of CoroVaxG.3 induced a potent humoral immunity with a balanced Th1/Th2 ratio and potent T-cell immunity, both lasting for at least 5 months. Sera from CoroVaxG.3-vaccinated mice was able to neutralize pseudoviruses expressing B.1 (wild type D614G), B.1.117 (alpha), P.1 (gamma) and B.1.617.2 (delta) Spikes, as well as an authentic P.1 SARS-CoV-2 isolate. Neutralizing antibodies did not wane even after 5 months, making this kind of vaccine a likely candidate to enter clinical trials.
Collapse
|
41
|
Basu S. Special Issue on 'Coronavirus: Vaccines and Other Therapeutics' (2020-2021). Vaccines (Basel) 2021; 9:1083. [PMID: 34696191 PMCID: PMC8537649 DOI: 10.3390/vaccines9101083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 09/24/2021] [Indexed: 11/16/2022] Open
Abstract
As is well known, the emergence of SARS-CoV-2 ever since late 2019 [...].
Collapse
Affiliation(s)
- Sankar Basu
- Department of Microbiology, Asutosh College, 92, Shyama Prasad Mukherjee Rd, Bhowanipore, Affiliated to Calcutta University, Kolkata 700026, India;
- 3BIO Group (External Scientific Collaborator), ULB, 1050 Brussels, Belgium
| |
Collapse
|
42
|
Osman IO, Levasseur A, Brechard L, Abdillahi Hassan I, Salah Abdillahi I, Ali Waberi Z, Delerce J, Bedotto M, Houhamdi L, Fournier PE, Colson P, Aboubaker MH, Raoult D, Devaux CA. Whole Genome Sequencing of SARS-CoV-2 Strains in COVID-19 Patients From Djibouti Shows Novel Mutations and Clades Replacing Over Time. Front Med (Lausanne) 2021; 8:737602. [PMID: 34540874 PMCID: PMC8440879 DOI: 10.3389/fmed.2021.737602] [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] [Received: 07/07/2021] [Accepted: 08/05/2021] [Indexed: 01/08/2023] Open
Abstract
Since the start of COVID-19 pandemic the Republic of Djibouti, in the horn of Africa, has experienced two epidemic waves of the virus between April and August 2020 and between February and May 2021. By May 2021, COVID-19 had affected 1.18% of the Djiboutian population and caused 152 deaths. Djibouti hosts several foreign military bases which makes it a potential hot-spot for the introduction of different SARS-CoV-2 strains. We genotyped fifty three viruses that have spread during the two epidemic waves. Next, using spike sequencing of twenty-eight strains and whole genome sequencing of thirteen strains, we found that Nexstrain clades 20A and 20B with a typically European D614G substitution in the spike and a frequent P2633L substitution in nsp16 were the dominant viruses during the first epidemic wave, while the clade 20H South African variants spread during the second wave characterized by an increase in the number of severe forms of COVID-19.
Collapse
Affiliation(s)
- Ikram Omar Osman
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France.,Laboratoire de Diagnostic, Centre de Soins 1, Caisse Nationale de Sécurité Sociale (CNSS), Djibouti, Djibouti
| | - Anthony Levasseur
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Ludivine Brechard
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Iman Abdillahi Hassan
- Laboratoire de Diagnostic, Centre de Soins 1, Caisse Nationale de Sécurité Sociale (CNSS), Djibouti, Djibouti
| | - Idil Salah Abdillahi
- Laboratoire de Diagnostic, Centre de Soins 1, Caisse Nationale de Sécurité Sociale (CNSS), Djibouti, Djibouti
| | - Zeinab Ali Waberi
- Laboratoire de Diagnostic, Centre de Soins 1, Caisse Nationale de Sécurité Sociale (CNSS), Djibouti, Djibouti
| | - Jeremy Delerce
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Marielle Bedotto
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | | | - Pierre-Edouard Fournier
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Philippe Colson
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Mohamed Houmed Aboubaker
- Laboratoire de Diagnostic, Centre de Soins 1, Caisse Nationale de Sécurité Sociale (CNSS), Djibouti, Djibouti
| | - Didier Raoult
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France
| | - Christian A Devaux
- IHU Méditerranée Infection, Marseille, France.,Aix-Marseille University, Institut de Recherche pour le Développement (IRD), Assistance Publique - Hôpitaux de Marseille (AP-HM), MEPHI, Marseille, France.,Centre National de la Recherche Scientifique, Marseille, France
| |
Collapse
|
43
|
Anantharajah A, Helaers R, Defour JP, Olive N, Kabera F, Croonen L, Deldime F, Vaerman JL, Barbée C, Bodéus M, Scohy A, Verroken A, Rodriguez-Villalobos H, Kabamba-Mukadi B. How to choose the right real-time RT-PCR primer sets for the SARS-CoV-2 genome detection? J Virol Methods 2021; 295:114197. [PMID: 34033854 PMCID: PMC8141720 DOI: 10.1016/j.jviromet.2021.114197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 05/12/2021] [Accepted: 05/21/2021] [Indexed: 01/10/2023]
Abstract
OBJECTIVES The SARS-CoV-2 pandemic has created an unprecedented need for rapid large-scale diagnostic testing to prompt clinical and public health interventions. Currently, several quantitative reverse-transcription polymerase chain reaction (RT-qPCR) assays recommended by the World Health Organization are being used by clinical and public health laboratories and typically target regions of the RNA-dependent RNA polymerase (RdRp), envelope (E) and nucleocapsid (N) coding region. However, it is currently unclear if results from different tests are comparable. This study aimed to clarify the clinical performances of the primer/probe sets designed by US CDC and Charité/Berlin to help clinical laboratories in assay selection for SARS-CoV-2 routine detection. METHODS We compared the clinical performances of the recommended primer/probe sets using one hundred nasopharyngeal swab specimens from patients who were clinically diagnosed with COVID-19. An additional 30 "pre-intervention screening" samples from patients who were not suspected of COVID-19 were also included in this study. We also performed sequence alignment between 31064 European SARS-CoV-2 and variants of concern genomes and the recommended primer/probe sets. RESULTS The present study demonstrates substantial differences in SARS-CoV-2 RNA detection sensitivity among the primer/probe sets recommended by the World Health Organization especially for low-level viral loads. The alignment of thousands of SARS-CoV-2 sequences reveals that the genetic diversity remains relatively low at the primer/probe binding sites. However, multiple nucleotide mismatches might contribute to false negatives. CONCLUSION An understanding of the limitations depending on the targeted genes and primer/probe sets may influence the selection of molecular detection assays by clinical laboratories.
Collapse
Affiliation(s)
- Ahalieyah Anantharajah
- Department of Microbiology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium; Department of Molecular Biology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium.
| | - Raphaël Helaers
- Human Molecular Genetics, de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Jean-Philippe Defour
- Department of Hematology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium; Ludwig Institute for Cancer Research & de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Nathalie Olive
- Department of Molecular Biology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Florence Kabera
- Department of Molecular Biology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Luc Croonen
- Department of Molecular Biology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Françoise Deldime
- Department of Molecular Biology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Jean-Luc Vaerman
- Department of Molecular Biology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Cindy Barbée
- Department of Molecular Biology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Monique Bodéus
- Department of Microbiology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Anais Scohy
- Department of Microbiology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Alexia Verroken
- Department of Microbiology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Hector Rodriguez-Villalobos
- Department of Microbiology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| | - Benoît Kabamba-Mukadi
- Department of Microbiology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium; Department of Molecular Biology, Cliniques universitaires Saint-Luc, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|