1
|
Almakrami M, Bazuqamah M, A. Alshehri M, M. S. Alqahtani A, F. Kadasah S, Harthi N, Ali Alyami R, Alqurashi A, A. Al Ruwaithi A. Identification of Significant Mutations in Spike Protein of SARS-CoV-2 Variants of Concern and the Discovery of Potent Inhibitors. Glob Health Epidemiol Genom 2025; 2025:5042190. [PMID: 40330793 PMCID: PMC12052452 DOI: 10.1155/ghe3/5042190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 03/11/2025] [Accepted: 03/21/2025] [Indexed: 05/08/2025] Open
Abstract
Background: SARS-CoV-2 is a positive-sense single-stranded RNA virus that has a propensity for infecting epithelial cells and the respiratory system. The two important proteins, structural and nonstructural proteins, make the architecture of this virus. Aim: This research aimed at studying significant mutations in spike protein of SARS-CoV-2 variants of concern (VoCs) and finding shared mutations among omicron and other four variants (alpha, beta, gamma, and delta). The purpose of this study was to draw structural comparisons between wild type and mutant proteins, followed by identifying potent inhibitors (ligand) that could be used against SARS-CoV-2 spike protein and its latest omicron VoC. Methodology: In this research, we had studied 16 major mutations as well as shared mutations (6) present in spike region of SARS-CoV-2. Subsequently, we determined the structure of the wild-type SARS-CoV-2 protein from the Protein Data Bank (PDB) with the ID 7R4I. Furthermore, the structure of the mutant protein of SARS-CoV-2 omicron variant was modeled in SWISS-MODEL. The ligand dataset for spike protein of SARS-CoV-2 was also collected from literature and different databases. Both wild type and mutant proteins were docked with ligand database in Molecular Operating Environment (MOE). The docking analysis was performed, and two best ligand molecules, AZ_2 and AZ_13, were finalized based on their energy values, interactions, and docking scores to be used against our wild and mutant proteins. Results: AZ_2 demonstrated a docking score of -6.1753 in MOE, with energy values of -4.3889 and -6.1753. It formed key hydrogen bond interactions. AZ_13 showed a docking score of -5.9, with energy values of -9.3 and -5.9, forming hydrogen donor and acceptor interactions with Asp950 (3.06 Å), Ile312 (3.13 Å), and Glu309 (3.27 Å). These interactions suggest strong binding affinity and potential efficacy. Thus, present research work emphasized on identification of significant mutations and finding a potent target-based drug against SARS-CoV-2 and its omicron variant. Outcomes: Based on this computational analysis performed, it is suggested that proposed compound can be used as remedy against SARS-CoV-2 and its omicron variant.
Collapse
Affiliation(s)
- Mohsen Almakrami
- Department of Pathology and Laboratory Medicine, King Khaled Hospital, Najran 66262, Saudi Arabia
| | - Mohammed Bazuqamah
- Department of Pathology and Laboratory Medicine, King Khaled Hospital, Najran 66262, Saudi Arabia
| | - Mohammed A. Alshehri
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Abdulaziz M. S. Alqahtani
- Department of Biology, Faculty of Science, University of Bisha, P.O. Box 551, Bisha 61922, Saudi Arabia
| | - Sultan F. Kadasah
- Department of Biology, Faculty of Science, University of Bisha, P.O. Box 551, Bisha 61922, Saudi Arabia
| | - Naif Harthi
- Emergency Medical Services, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Rami Ali Alyami
- Respiratory Therapy Department, Faculty of Applied Medical Sciences, Jazan University, Jazan, Saudi Arabia
| | - Abdulmajeed Alqurashi
- Department of Biology, College of Science, Taibah University, Madinah 42353, Saudi Arabia
| | | |
Collapse
|
2
|
Usserbayev B, Sultankulova KT, Burashev Y, Melisbek A, Shirinbekov M, Myrzakhmetova BS, Zhunushov A, Smekenov I, Kerimbaev A, Nurabaev S, Chervyakova O, Kozhabergenov N, Kutumbetov LB. Genetic Variations of Three Kazakhstan Strains of the SARS-CoV-2 Virus. Viruses 2025; 17:415. [PMID: 40143342 PMCID: PMC11945512 DOI: 10.3390/v17030415] [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: 01/30/2025] [Revised: 03/06/2025] [Accepted: 03/11/2025] [Indexed: 03/28/2025] Open
Abstract
Prompt determination of the etiological agent is important in an outbreak of pathogens with pandemic potential, particularly for dangerous infectious diseases. Molecular genetic methods allow for arriving at an accurate diagnosis, employing timely preventive measures, and controlling the spread of the disease-causing agent. In this study, whole-genome sequencing of three SARS-CoV-2 strains was performed using the Sanger method, which provides high accuracy in determining nucleotide sequences and avoids errors associated with multiple DNA amplification. Complete nucleotide sequences of samples, KAZ/Britain/2021, KAZ/B1.1/2021, and KAZ/Delta020/2021 were obtained, with sizes of 29.751 bp, 29.815 bp, and 29.840 bp, respectively. According to the COVID-19 Genome Annotator, 127 mutations were detected in the studied samples compared to the reference strain. The strain KAZ/Britain/2021 contained 3 deletions, 7 synonymous mutations, and 27 non-synonymous mutations, the second strain KAZ/B1.1/2021 contained 1 deletion, 5 synonymous mutations, and 31 non-synonymous mutations, and the third strain KAZ/Delta020/2021 contained 1 deletion, 5 synonymous mutations, and 37 non-synonymous mutations, respectively. The variations C241T, F106F, P314L, and D614G found in the 5' UTR, ORF1ab, and S regions were common to all three studied samples, respectively. According to PROVEAN data, the loss-of-function mutations identified in strains KAZ/Britain/2021, KAZ/B1.1/2021, and KAZ/Delta020/2021 include 5 mutations (P218L, T716I, W149L, R52I, and Y73C), 2 mutations (S813I and Q992H), and 8 mutations (P77L, L452R, I82T, P45L, V82A, F120L, F120L, and R203M), respectively. Phylogenetic analysis showed that the strains studied (KAZ/Britain/2021, KAZ/B1.1/2021, and KAZ/Delta020/2021) belong to different SARS-CoV-2 lineages, which are closely related to samples from Germany (OU141323.1 and OU365922.1), Mexico (OK432605.1), and again Germany (OV375251.1 and OU375174.1), respectively. The nucleotide sequences of the studied SARS-CoV-2 virus strains were registered in the Genbank database with the accession numbers: ON692539.1, OP684305, and OQ561548.1.
Collapse
Affiliation(s)
- Bekbolat Usserbayev
- Research Institute for Biological Safety Problems, National Holding QazBioPharm, LLP, Guardeyskiy uts 080409, Kazakhstan; (K.T.S.); (Y.B.); (A.M.); (M.S.); (B.S.M.); (A.K.); (S.N.); (O.C.); (L.B.K.)
- Institute of Biotechnology, National Academy of Science of Kyrgyzstan, Bishkek 720071, Kyrgyzstan
| | - Kulyaisan T. Sultankulova
- Research Institute for Biological Safety Problems, National Holding QazBioPharm, LLP, Guardeyskiy uts 080409, Kazakhstan; (K.T.S.); (Y.B.); (A.M.); (M.S.); (B.S.M.); (A.K.); (S.N.); (O.C.); (L.B.K.)
| | - Yerbol Burashev
- Research Institute for Biological Safety Problems, National Holding QazBioPharm, LLP, Guardeyskiy uts 080409, Kazakhstan; (K.T.S.); (Y.B.); (A.M.); (M.S.); (B.S.M.); (A.K.); (S.N.); (O.C.); (L.B.K.)
- Scientific Research Institute of Biology and Biotechnology Problems, al-Farabi Kazakh National University, Almaty 050040, Kazakhstan;
| | - Aibarys Melisbek
- Research Institute for Biological Safety Problems, National Holding QazBioPharm, LLP, Guardeyskiy uts 080409, Kazakhstan; (K.T.S.); (Y.B.); (A.M.); (M.S.); (B.S.M.); (A.K.); (S.N.); (O.C.); (L.B.K.)
- Scientific Research Institute of Biology and Biotechnology Problems, al-Farabi Kazakh National University, Almaty 050040, Kazakhstan;
| | - Meirzhan Shirinbekov
- Research Institute for Biological Safety Problems, National Holding QazBioPharm, LLP, Guardeyskiy uts 080409, Kazakhstan; (K.T.S.); (Y.B.); (A.M.); (M.S.); (B.S.M.); (A.K.); (S.N.); (O.C.); (L.B.K.)
| | - Balzhan S. Myrzakhmetova
- Research Institute for Biological Safety Problems, National Holding QazBioPharm, LLP, Guardeyskiy uts 080409, Kazakhstan; (K.T.S.); (Y.B.); (A.M.); (M.S.); (B.S.M.); (A.K.); (S.N.); (O.C.); (L.B.K.)
| | - Asankadir Zhunushov
- Institute of Biotechnology, National Academy of Science of Kyrgyzstan, Bishkek 720071, Kyrgyzstan
| | - Izat Smekenov
- Scientific Research Institute of Biology and Biotechnology Problems, al-Farabi Kazakh National University, Almaty 050040, Kazakhstan;
| | - Aslan Kerimbaev
- Research Institute for Biological Safety Problems, National Holding QazBioPharm, LLP, Guardeyskiy uts 080409, Kazakhstan; (K.T.S.); (Y.B.); (A.M.); (M.S.); (B.S.M.); (A.K.); (S.N.); (O.C.); (L.B.K.)
| | - Sergazy Nurabaev
- Research Institute for Biological Safety Problems, National Holding QazBioPharm, LLP, Guardeyskiy uts 080409, Kazakhstan; (K.T.S.); (Y.B.); (A.M.); (M.S.); (B.S.M.); (A.K.); (S.N.); (O.C.); (L.B.K.)
| | - Olga Chervyakova
- Research Institute for Biological Safety Problems, National Holding QazBioPharm, LLP, Guardeyskiy uts 080409, Kazakhstan; (K.T.S.); (Y.B.); (A.M.); (M.S.); (B.S.M.); (A.K.); (S.N.); (O.C.); (L.B.K.)
| | - Nurlan Kozhabergenov
- Research Institute for Biological Safety Problems, National Holding QazBioPharm, LLP, Guardeyskiy uts 080409, Kazakhstan; (K.T.S.); (Y.B.); (A.M.); (M.S.); (B.S.M.); (A.K.); (S.N.); (O.C.); (L.B.K.)
| | - Lesbek B. Kutumbetov
- Research Institute for Biological Safety Problems, National Holding QazBioPharm, LLP, Guardeyskiy uts 080409, Kazakhstan; (K.T.S.); (Y.B.); (A.M.); (M.S.); (B.S.M.); (A.K.); (S.N.); (O.C.); (L.B.K.)
| |
Collapse
|
3
|
Rostamzadeh S, Allafasghari A, Allafasghari A, Abouhossein A. Handgrip strength as a prognostic factor for COVID-19 mortality among older adult patients admitted to the intensive care unit (ICU): a comparison Alpha (B.1.1.7) and Delta (B.1.617.2) variants. Sci Rep 2024; 14:19927. [PMID: 39198687 PMCID: PMC11358457 DOI: 10.1038/s41598-024-71034-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 08/23/2024] [Indexed: 09/01/2024] Open
Abstract
Handgrip strength (HGS) is a non-invasive and reliable biomarker of overall health, physical function, mobility, and mortality. This study aimed to investigate the possible relationship between HGS and mortality in older adult patients hospitalized with COVID-19 in the intensive care unit (ICU) by Alpha (B.1.1.7) and Delta (B.1.617.2) variants. This retrospective cohort study was conducted on 472 COVID-19 patients (222 female and 250 male) aged 60-85 years admitted to the ICU. Demographic data, underlying comorbidities, COVID-19-related symptoms, as well as laboratory and computed tomography (CT) findings were obtained from the patient's medical records. Using a JAMAR® hydraulic dynamometer, the average grip strength value (kg) after three measurements on the dominant side was recorded for subsequent analysis. Low grip strength (LGS) was defined as an arbitrary cut-off of two standard deviations below the gender-specific peak mean value of normative HGS in Iranian healthy population, i.e. < 26 kg in males and < 14 kg in females. The findings showed lower mean grip strength and high frequency of LGS in the non-survivors patients versus survivors group and in the Delta (B.1.617.2) variant vs. Alpha (B.1.1.7) variant, respectively (both p < 0.01). The binary logistic regression analysis showed that chronic obstructive pulmonary disease (COPD) (adjusted odds ratio [OR] 5.125, 95% CI 1.425-25.330), LGS (OR 4.805, 95% CI 1.624-10.776), SaO2 (OR - 3.501, 95% CI 2.452-1.268), C-reactive protein (CRP) level (OR 2.625, 95% CI 1.256-7.356), and age (OR 1.118, 95% CI 1.045-1.092) were found to be independent predictors for mortality of patients with Alpha (B.1.1.7) variant (all p < 0.05). However, only four independent predictors including COPD (OR 6.728, 95% CI 1.683-28.635), LGS (OR 5.405, 95% CI 1.461-11.768), SaO2 (OR - 4.120, 95% CI 2.924-1.428), and CRP level (OR 1.893, 95% CI 1.127-8.692) can be predicted the mortality of patients with Delta (B.1.617.2) variant (p < 0.05). Along with the well-known and common risk factors (i.e. COPD, CRP, and SaO2), handgrip strength can be a quick and low-cost prognostic tool in predicting chances of mortality in older adults who are afflicted with COVID-19 variants.
Collapse
Affiliation(s)
- Sajjad Rostamzadeh
- Department of Ergonomics, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atabak Allafasghari
- Department of Health, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Amin Allafasghari
- Department of Health, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Alireza Abouhossein
- Department of Ergonomics, School of Public Health and Safety, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
4
|
Karim B, Barary M, Fereydouni Z, Sanjari E, Hosseinzadeh R, Salehi-Vaziri M, Maleki A. The nuts and bolts of recombination in the generation of SARS-CoV-2 variants; from XA to XBB. Lett Appl Microbiol 2024; 77:ovae074. [PMID: 39081071 DOI: 10.1093/lambio/ovae074] [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: 04/30/2024] [Revised: 06/23/2024] [Accepted: 07/29/2024] [Indexed: 01/28/2025]
Abstract
Since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), new variants with enhanced transmissibility and pathogenicity have surfaced. The World Health Organization has designated five such variants-Alpha (B.1.1.7), Beta (B.1.351), Gamma (P.1), Delta (B.1.617.2), and Omicron (B.1.1.529)-as variants of concern. Each variant exhibits distinct characteristics, with many displaying a combination of point mutations and insertions/deletions (indels). These genetic alterations, including mutations, recombinations, and rearrangements, contribute to the emergence of new strains that may exhibit modified phenotypes. However, identifying recombinant forms can be challenging due to their resemblance to other lineages. It is critical to monitor the evolution of new recombinant variants, particularly in light of the potential for vaccine-resistant strains and their accelerated propagation. Recombination has played a pivotal role in the development of certain SARS-CoV-2 variants, such as XA, XD, XF, XE, and XBB, among others. This report delves into the significance of recombination in the evolution of SARS-CoV-2 variants, especially Omicron sublineages, underscoring the necessity for continuous surveillance of the SARS-CoV-2 genome to identify newly emerged recombinant variants.
Collapse
Affiliation(s)
- Bardia Karim
- Student Research Committee, Babol University of Medical Sciences, Babol 4717647745, Iran
| | - Mohammad Barary
- Student Research Committee, Virtual School of Medical Education and Management, Shahid Beheshti University of Medical Sciences, Tehran 1983969411, Iran
| | - Zahra Fereydouni
- COVID-19 National Reference Laboratory (CNRL), Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran
| | - Elaheh Sanjari
- Student Research Committee, Faculty of Pharmacy, Islamic Azad University, Ayatollah Amoli Branch, Amol 678, Iran
| | - Rezvan Hosseinzadeh
- Student Research Committee, Babol University of Medical Sciences, Babol 4717647745, Iran
| | - Mostafa Salehi-Vaziri
- Department of Arboviruses and Viral Hemorrhagic Fevers (National Reference Laboratory), Pasteur Institute of Iran, Pasteur Ave., Tehran 01316943551, Iran
| | - Ali Maleki
- COVID-19 National Reference Laboratory (CNRL), Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran
- Department of Influenza and Respiratory Viruses, Pasteur Institute of Iran, Pasteur Ave., Tehran 1316943551, Iran
| |
Collapse
|
5
|
Van Nam L, Dien TC, Bang LVN, Thach PN, Van Duyet L. Genetic features of SARS-CoV-2 Alpha, Delta, and Omicron variants and their association with the clinical severity of COVID-19 in Vietnam. IJID REGIONS 2024; 11:100348. [PMID: 38601946 PMCID: PMC11004080 DOI: 10.1016/j.ijregi.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/12/2024]
Abstract
Objectives We investigated the genetic variations in the Alpha, Delta, and Omicron variants of SARS-CoV-2 and their association with clinical status and treatment outcomes in patients with COVID-19. Methods MiSeq was used to sequence the Alpha, Delta, and Omicron genomes, and MEGA 6.6 was used to define the nucleotide variations. We determined the association between clinical severity and treatment outcomes for the SARS-CoV-2 variants. Results The BA.1.1 and BA.2 lineages of the Omicron variant had 57-59 mutations, which is 2-2.7-fold higher than that of the B.1.1.7 (Alpha), B.1.617.2, and AY.57 (Delta) lineages. We found distinct mutations in SARS-CoV-2: five in Alpha (C26305T, G26558T, G7042T, C14120T, and C27509T); seven in Delta (C26408T, C1403T, C5184T, C9891T, T11418C, C11514T, and C22227T); and three in Omicron (C26408T, C8991T, and C25810T). Patients with the Delta variant had a severe rate of 23.8%, a critical rate of 53.7%, and a mortality rate of 38.9%, which were significantly higher than those with the Omicron and Alpha variants. Conclusions The Alpha, Delta, and Omicron variants in this study had genetic diversity and differed from the strains reported in other countries, with the Delta variant producing significantly more clinical severity and mortality than the Alpha and Omicron variants.
Collapse
Affiliation(s)
- Le Van Nam
- Departments of Infectious Disease, Military Hospital, Hanoi, Vietnam
| | - Trinh Cong Dien
- Departments of Infectious Disease, Military Hospital, Hanoi, Vietnam
| | | | - Pham Ngoc Thach
- Micobiology and Molecular Biology Department, National Hospital for Tropical Diseases, Hanoi, Vietnam
| | - Le Van Duyet
- Micobiology and Molecular Biology Department, National Hospital for Tropical Diseases, Hanoi, Vietnam
| |
Collapse
|
6
|
Amblard F, LeCher JC, De R, Zhou S, Liu P, Goh SL, Tao S, Patel D, Downs-Bowen J, Zandi K, Zhang H, Chaudhry G, McBrayer T, Muczynski M, Al-Homoudi A, Engel J, Lan S, Sarafianos SG, Kovari LC, Schinazi RF. Synthesis and biological evaluation of novel peptidomimetic inhibitors of the coronavirus 3C-like protease. Eur J Med Chem 2024; 268:116263. [PMID: 38432056 PMCID: PMC11188152 DOI: 10.1016/j.ejmech.2024.116263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 03/05/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), and related variants, are responsible for the devastating coronavirus disease 2019 (COVID-19) pandemic. The SARS-CoV-2 main protease (Mpro) plays a central role in the replication of the virus and represents an attractive drug target. Herein, we report the discovery of novel SARS-CoV-2 Mpro covalent inhibitors, including highly effective compound NIP-22c which displays high potency against several key variants and clinically relevant nirmatrelvir Mpro E166V mutants.
Collapse
Affiliation(s)
- Franck Amblard
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA.
| | - Julia C LeCher
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Ramyani De
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Shaoman Zhou
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Peng Liu
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Shu Ling Goh
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Sijia Tao
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Dharmeshkumar Patel
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Jessica Downs-Bowen
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Keivan Zandi
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Huanchun Zhang
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Gitika Chaudhry
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Tamara McBrayer
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Michael Muczynski
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Abdullah Al-Homoudi
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Joseph Engel
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Shuiyun Lan
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Stefan G Sarafianos
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Ladislau C Kovari
- Department of Biochemistry, Microbiology, and Immunology, Wayne State University School of Medicine, Detroit, MI, 48201, USA
| | - Raymond F Schinazi
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine, and Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA.
| |
Collapse
|
7
|
Zhang X, Zhou Z. The Mechanism of bnAb Production and Its Application in Mutable Virus Broad-Spectrum Vaccines: Inspiration from HIV-1 Broad Neutralization Research. Vaccines (Basel) 2023; 11:1143. [PMID: 37514959 PMCID: PMC10384589 DOI: 10.3390/vaccines11071143] [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: 05/18/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
Elite controllers among HIV-1-infected individuals have demonstrated a stronger ability to control the viral load in their bodies. Scientists have isolated antibodies with strong neutralizing ability from these individuals, which can neutralize HIV-1 variations; these are known as broadly neutralizing antibodies. The nucleic acid of some viruses will constantly mutate during replication (such as SARS-CoV-2), which will reduce the protective ability of the corresponding vaccines. The immune escape caused by this mutation is the most severe challenge faced by humans in the battle against the virus. Therefore, developing broad-spectrum vaccines that can induce broadly neutralizing antibodies against various viruses and their mutated strains is the best way to combat virus mutations. Exploring the mechanism by which the human immune system produces broadly neutralizing antibodies and its induction strategies is crucial in the design process of broad-spectrum vaccines.
Collapse
Affiliation(s)
- Xinyu Zhang
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
- Institute for Biological Product Control, National Institutes for Food and Drug Control (NIFDC) and WHO Collaborating Center for Standardization and Evaluation of Biologicals, No. 31 Huatuo Street, Daxing District, Beijing 102629, China
- College of Life Science, Jilin University, Changchun 130012, China
| | - Zehua Zhou
- Research Center for Infectious Diseases, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| |
Collapse
|
8
|
Vilain M, Aris-Brosou S. Machine Learning Algorithms Associate Case Numbers with SARS-CoV-2 Variants Rather Than with Impactful Mutations. Viruses 2023; 15:1226. [PMID: 37376526 DOI: 10.3390/v15061226] [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: 04/03/2023] [Revised: 05/12/2023] [Accepted: 05/13/2023] [Indexed: 06/29/2023] Open
Abstract
During the SARS-CoV-2 pandemic, much effort has been geared towards creating models to predict case numbers. These models typically rely on epidemiological data, and as such overlook viral genomic information, which could be assumed to improve predictions, as different variants show varying levels of virulence. To test this hypothesis, we implemented simple models to predict future case numbers based on the genomic sequences of the Alpha and Delta variants, which were co-circulating in Texas and Minnesota early during the pandemic. Sequences were encoded, matched with case numbers at a future time based on collection date, and used to train two algorithms: one based on random forests and one based on a feed-forward neural network. While prediction accuracies were ≥93%, explainability analyses showed that the models were not associating case numbers with mutations known to have an impact on virulence, but with individual variants. This work highlights the necessity of gaining a better understanding of the data used for training and of conducting explainability analysis to assess whether model predictions are misleading.
Collapse
Affiliation(s)
- Matthieu Vilain
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| | - Stéphane Aris-Brosou
- Department of Biology, University of Ottawa, Ottawa, ON K1N 6N5, Canada
- Department of Mathematics and Statistics, University of Ottawa, Ottawa, ON K1N 6N5, Canada
| |
Collapse
|
9
|
Liang F. Quantitative Mutation Analysis of Genes and Proteins of Major SARS-CoV-2 Variants of Concern and Interest. Viruses 2023; 15:v15051193. [PMID: 37243278 DOI: 10.3390/v15051193] [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: 04/06/2023] [Revised: 05/09/2023] [Accepted: 05/14/2023] [Indexed: 05/28/2023] Open
Abstract
Of various SARS-CoV-2 variants, some have drawn special concern or interest because of their heightened disease threat. The mutability of individual SARS-CoV-2 genes/proteins presumably varies. The present study quantified gene/protein mutations in 13 major SARS-CoV-2 variants of concern/interest, and analyzed viral protein antigenicity using bioinformatics. The results from 187 carefully perused genome clones showed significantly higher mean percent mutations in the spike, ORF8, nucleocapsid, and NSP6 than in other viral proteins. The ORF8 and spike proteins also tolerated higher maximal percent mutations. The omicron variant presented more percent mutations in the NSP6 and structural proteins, whereas the delta featured more in the ORF7a. Omicron subvariant BA.2 exhibited more mutations in ORF6, and omicron BA.4 had more in NSP1, ORF6, and ORF7b, relative to omicron BA.1. Delta subvariants AY.4 and AY.5 bore more mutations in ORF7b and ORF8 than delta B.1.617.2. Predicted antigen ratios of SARS-CoV-2 proteins significantly vary (range: 38-88%). To overcome SARS-CoV-2 immune evasion, the relatively conserved, potentially immunogenic NSP4, NSP13, NSP14, membrane, and ORF3a viral proteins may serve as more suitable targets for molecular vaccines or therapeutics than the mutation-prone NSP6, spike, ORF8, or nucleocapsid protein. Further investigation into distinct mutations of the variants/subvariants may help understand SARS-CoV-2 pathogenesis.
Collapse
Affiliation(s)
- Fengyi Liang
- Department of Anatomy, Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Singapore 117594, Singapore
| |
Collapse
|
10
|
Zafar H, Saier MH. Understanding the Relationship of the Human Bacteriome with COVID-19 Severity and Recovery. Cells 2023; 12:cells12091213. [PMID: 37174613 PMCID: PMC10177376 DOI: 10.3390/cells12091213] [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: 02/23/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 05/15/2023] Open
Abstract
The Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) first emerged in 2019 in China and has resulted in millions of human morbidities and mortalities across the globe. Evidence has been provided that this novel virus originated in animals, mutated, and made the cross-species jump to humans. At the time of this communication, the Coronavirus disease (COVID-19) may be on its way to an endemic form; however, the threat of the virus is more for susceptible (older and immunocompromised) people. The human body has millions of bacterial cells that influence health and disease. As a consequence, the bacteriomes in the human body substantially influence human health and disease. The bacteriomes in the body and the immune system seem to be in constant association during bacterial and viral infections. In this review, we identify various bacterial spp. In major bacteriomes (oral, nasal, lung, and gut) of the body in healthy humans and compare them with dysbiotic bacteriomes of COVID-19 patients. We try to identify key bacterial spp. That have a positive effect on the functionality of the immune system and human health. These select bacterial spp. Could be used as potential probiotics to counter or prevent COVID-19 infections. In addition, we try to identify key metabolites produced by probiotic bacterial spp. That could have potential anti-viral effects against SARS-CoV-2. These metabolites could be subject to future therapeutic trials to determine their anti-viral efficacies.
Collapse
Affiliation(s)
- Hassan Zafar
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, CA 92093-0116, USA
- Central European Institute of Technology, Masaryk University, 625 00 Brno, Czech Republic
| | - Milton H Saier
- Department of Molecular Biology, School of Biological Sciences, University of California, San Diego, CA 92093-0116, USA
| |
Collapse
|
11
|
Peka M, Balatsky V. The impact of mutation sets in receptor-binding domain of SARS-CoV-2 variants on the stability of RBD–ACE2 complex. Future Virol 2023. [PMID: 37064325 PMCID: PMC10089296 DOI: 10.2217/fvl-2022-0152] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 02/01/2023] [Indexed: 04/08/2023]
Abstract
Aim: Bioinformatic analysis of mutation sets in receptor-binding domain (RBD) of currently and previously circulating SARS-CoV-2 variants of concern (VOCs) and interest (VOIs) to assess their ability to bind the ACE2 receptor. Methods: In silico sequence and structure-oriented approaches were used to evaluate the impact of single and multiple mutations. Results: Mutations detected in VOCs and VOIs led to the reduction of binding free energy of the RBD–ACE2 complex, forming additional chemical bonds with ACE2, and to an increase of RBD–ACE2 complex stability. Conclusion: Mutation sets characteristic of SARS-CoV-2 variants have complex effects on the ACE2 receptor-binding affinity associated with amino acid interactions at mutation sites, as well as on the acquisition of other viral adaptive advantages.
Collapse
Affiliation(s)
- Mykyta Peka
- V. N. Karazin Kharkiv National University, Kharkiv, 61022, Ukraine
- Institute of Pig Breeding & Agroindustrial Production, National Academy of Agrarian Sciences of Ukraine, Poltava, 36013, Ukraine
| | - Viktor Balatsky
- V. N. Karazin Kharkiv National University, Kharkiv, 61022, Ukraine
- Institute of Pig Breeding & Agroindustrial Production, National Academy of Agrarian Sciences of Ukraine, Poltava, 36013, Ukraine
| |
Collapse
|
12
|
Petrella C, Zingaropoli MA, Ceci FM, Pasculli P, Latronico T, Liuzzi GM, Ciardi MR, Angeloni A, Ettorre E, Menghi M, Barbato C, Ferraguti G, Minni A, Fiore M. COVID-19 Affects Serum Brain-Derived Neurotrophic Factor and Neurofilament Light Chain in Aged Men: Implications for Morbidity and Mortality. Cells 2023; 12:cells12040655. [PMID: 36831321 PMCID: PMC9954454 DOI: 10.3390/cells12040655] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/14/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023] Open
Abstract
BACKGROUND AND METHODS Severe COVID-19 is known to induce neurological damage (NeuroCOVID), mostly in aged individuals, by affecting brain-derived neurotrophic factor (BDNF), matrix metalloproteinases (MMP) 2 and 9 and the neurofilament light chain (NFL) pathways. Thus, the aim of this pilot study was to investigate BDNF, MMP-2, MMP-9, and NFL in the serum of aged men affected by COVID-19 at the beginning of the hospitalization period and characterized by different outcomes, i.e., attending a hospital ward or an intensive care unit (ICU) or with a fatal outcome. As a control group, we used a novelty of the study, unexposed age-matched men. We also correlated these findings with the routine blood parameters of the recruited individuals. RESULTS We found in COVID-19 individuals with severe or lethal outcomes disrupted serum BDNF, NFL, and MMP-2 presence and gross changes in ALT, GGT, LDH, IL-6, ferritin, and CRP. We also confirmed and extended previous data, using ROC analyses, showing that the ratio MMPs (2 and 9) versus BDNF and NFL might be a useful tool to predict a fatal COVID-19 outcome. CONCLUSIONS Serum BDNF and NFL and/or their ratios with MMP-2 and MMP-9 could represent early predictors of NeuroCOVID in aged men.
Collapse
Affiliation(s)
- Carla Petrella
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: (C.P.); (M.F.)
| | - Maria Antonella Zingaropoli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Flavio Maria Ceci
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Patrizia Pasculli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Tiziana Latronico
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Grazia Maria Liuzzi
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Maria Rosa Ciardi
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Viale del Policlinico 155, 00185 Rome, Italy
| | - Antonio Angeloni
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Evaristo Ettorre
- Department of Clinical, Internal Medicine, Anesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Viale del Policlinico 155, 00161 Rome, Italy
| | - Michela Menghi
- Department of Maternal Infantile and Urological Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Christian Barbato
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
| | - Giampiero Ferraguti
- Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
| | - Antonio Minni
- Department of Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
- Division of Otolaryngology-Head and Neck Surgery, ASL Rieti-Sapienza University, Ospedale San Camillo de Lellis, Viale Kennedy, 02100 Rieti, Italy
| | - Marco Fiore
- Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: (C.P.); (M.F.)
| |
Collapse
|
13
|
Hermens JM, Kesmir C. Role of T cells in severe COVID-19 disease, protection, and long term immunity. Immunogenetics 2023; 75:295-307. [PMID: 36752852 PMCID: PMC9905767 DOI: 10.1007/s00251-023-01294-9] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 01/10/2023] [Indexed: 02/09/2023]
Abstract
Infection with SARS-CoV-2 causes wide range of disease severities from asymptomatic to life-threatening disease. Understanding the contribution of immunological traits in immunity against SARS-CoV-2 and in protection against severe COVID-19 could result in effective measures to prevent development of severe disease. While the role of cytokines and antibodies has been thoroughly studied, this is not the case for T cells. In this review, the association between T cells and COVID-19 disease severity and protection upon reexposure is discussed. While infiltration of overactivated cytotoxic T cells might be harmful in the infected tissue, fast responding T cells are important in the protection against severe COVID-19. This protection could even be viable in the long term as long-living memory T cells seem to be stabilized and mutations do not appear to have a large impact on T cell responses. Thus, after vaccination and infections, memory T cells should be able to help prevent onset of severe disease for most cases. Considering this, it would be useful to add N or M proteins in vaccinations, alongside the S protein which is currently used, as this results in a broader T cell response.
Collapse
Affiliation(s)
- Julia Maret Hermens
- Theoretical Biology and Bioinformatics, Biology Department, Science Faculty, Utrecht University, Utrecht, Netherlands
| | - Can Kesmir
- Theoretical Biology and Bioinformatics, Biology Department, Science Faculty, Utrecht University, Utrecht, Netherlands.
| |
Collapse
|
14
|
Mamurova B, Akan G, Mogol E, Turgay A, Tuncel G, Evren EU, Evren H, Suer K, Sanlidag T, Ergoren MC. Strong Association between Vitamin D Receptor Gene and Severe Acute Respiratory Syndrome coronavirus 2 Infectious Variants. Glob Med Genet 2023; 10:27-33. [PMID: 36819669 PMCID: PMC9935054 DOI: 10.1055/s-0043-1761924] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
Abstract
A coronavirus disease 2019 (COVID-19) disease, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has created significant concern since December 2019 worldwide. The virus is known to be highly transmissible. Heterogenic clinical features even vary more among SARS-CoV-2 variants from asymptomatic forms to severe symptoms. Previous studies revealed an association between COVID-19 and vitamin D deficiency resulting from its low levels in COVID-19 patients. To our knowledge, there is no scientific investigation that evaluates the direct association between SARS-CoV-2 variants of concern and vitamin D receptor ( VDR ) gene markers in Cyprus. Thus, the present study aimed to identify the putative impact of VDR gene polymorphisms on SARS-CoV-2 infection among different variants. The nasopharyngeal swabs were taken from a total number of 600 patients who were admitted to Near East University Hospital COVID-19 Polymerase Chain Reaction (PCR) Diagnosis Laboratory for routine SARS-CoV-2 real-time quantitative reverse transcription PCR (RT-qPCR) test. The RT-qPCR negative resulting samples were taken as control samples ( n = 300). On the contrary, the case group consisted of patients who were SARS-CoV-2 RT-qPCR positive, infected with either SARS-CoV-2 Alpha ( n = 100), Delta ( n = 100), or Omicron ( n = 100) variants. Two VDR gene polymorphisms, Taq I-rs731236 T > C and Fok I-rs10735810 C > T, were genotyped by polymerase chain reaction-restriction fragment length polymorphism. The mean age of the COVID-19 patient's ± standard deviation was 46.12 ± 12.36 and 45.25 ± 12.71 years old for the control group ( p > 0.05). The gender distribution of the patient group was 48.3% female and 51.7% male and for the control group 43% female and 57% male ( p > 0.05). Significant differences were observed in genotype frequencies of FokI and TaqI variants between SARS-CoV-2 patients compared to the control group ( p < 0.005). Furthermore, the risk alleles, FokI T allele and TaqI C, were found to be statistically significant (odds ratio [OR] = 1.80, 95% confidence interval [CI] = 1.42-2.29, OR = 1.62, 95% CI = 1.27-2.05, respectively) in COVID-19 patients. The highest number of patients with wild-type genotype was found in the control group, which is 52.9% compared with 17.5% in the case group. Moreover, most of the COVID-19 patients had heterozygous/homozygous genotypes, reaching 82.5%, while 47.1% of the control group patients had heterozygous/homozygous genotypes. Our results suggested that patients with FokI and TaqI polymorphisms might tend to be more susceptible to getting infected with SARS-CoV-2. Overall, findings from this study provided evidence regarding vitamin D supplements recommendation in individuals with vitamin D deficiency/insufficiency in the peri- or post-COVID-19 pandemic.
Collapse
Affiliation(s)
- Begimai Mamurova
- Department of Molecular Medicine, Institute of Graduate Studies, Near East University, Nicosia, Cyprus
| | - Gokce Akan
- DESAM Research Institute, Near East University, Nicosia, Cyprus
| | - Evren Mogol
- Department of Molecular Medicine, Institute of Graduate Studies, Near East University, Nicosia, Cyprus
| | - Ayla Turgay
- Department of Molecular Medicine, Institute of Graduate Studies, Near East University, Nicosia, Cyprus
- Laboratory of Medical Genetics, Near East University Hospital, Near East University, Nicosia, Cyprus
| | - Gulten Tuncel
- DESAM Research Institute, Near East University, Nicosia, Cyprus
| | - Emine Unal Evren
- Department of Medical Microbiology and Infectious Diseases, Faculty of Medicine, Kyrenia University
| | - Hakan Evren
- Department of Medical Microbiology and Infectious Diseases, Faculty of Medicine, Kyrenia University
| | - Kaya Suer
- Department of Medical Microbiology and Infectious Diseases, Faculty of Medicine, Near East University, Nicosia, Cyprus
| | - Tamer Sanlidag
- DESAM Research Institute, Near East University, Nicosia, Cyprus
| | - Mahmut Cerkez Ergoren
- Department of Molecular Medicine, Institute of Graduate Studies, Near East University, Nicosia, Cyprus
- Laboratory of Medical Genetics, Near East University Hospital, Near East University, Nicosia, Cyprus
- Department of Medical Genetics, Faculty of Medicine, Near East University, Nicosia, Cyprus
| |
Collapse
|
15
|
Rani R, Long S, Pareek A, Dhaka P, Singh A, Kumar P, McInerney G, Tomar S. Multi-target direct-acting SARS-CoV-2 antivirals against the nucleotide-binding pockets of virus-specific proteins. Virology 2022; 577:1-15. [PMID: 36244310 PMCID: PMC9539459 DOI: 10.1016/j.virol.2022.08.008] [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: 06/24/2022] [Revised: 08/20/2022] [Accepted: 08/20/2022] [Indexed: 11/30/2022]
Abstract
The nucleotide-binding pockets (NBPs) in virus-specific proteins have proven to be the most successful antiviral targets for several viral diseases. Functionally important NBPs are found in various structural and non-structural proteins of SARS-CoV-2. In this study, the first successful multi-targeting attempt to identify effective antivirals has been made against NBPs in nsp12, nsp13, nsp14, nsp15, nsp16, and nucleocapsid (N) proteins of SARS-CoV-2. A structure-based drug repurposing in silico screening approach with ADME analysis identified small molecules targeting NBPs in SARS-CoV-2 proteins. Further, isothermal titration calorimetry (ITC) experiments validated the binding of top hit molecules to the purified N-protein. Importantly, cell-based antiviral assays revealed antiviral potency for INCB28060, darglitazone, and columbianadin with EC50 values 15.71 μM, 5.36 μM, and 22.52 μM, respectively. These effective antivirals targeting multiple proteins are envisioned to direct the development of antiviral therapy against SARS-CoV-2 and its emerging variants.
Collapse
Affiliation(s)
- Ruchi Rani
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Siwen Long
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Akshay Pareek
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Preeti Dhaka
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Ankur Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Pravindra Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Gerald McInerney
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, SE-17177 Stockholm, Sweden
| | - Shailly Tomar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India.
| |
Collapse
|
16
|
de la Fuente J. Approaching Challenges Posed by SARS-CoV-2 Genetic Variants. Pathogens 2022; 11:1407. [PMID: 36558741 PMCID: PMC9781686 DOI: 10.3390/pathogens11121407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
In this new collection of the most viewed and cited papers, one of the Editor's chosen articles, published in Pathogens in 2021, addressed the impact and the concerns relating to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants [...].
Collapse
Affiliation(s)
- José de la Fuente
- SaBio, Instituto de Investigación en Recursos Cinegéticos IREC-CSIC-UCLM-JCCM, Ronda de Toledo s/n, 13005 Ciudad Real, Spain
- Department of Veterinary Pathobiology, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
17
|
Khamis F, Al Awaidy S, Ba’Omar M, Osman W, Chhetri S, Ambusaid Z, Al Fahdi Z, Al Lawati J, Al Sulaimi K, Al Bulushi SA, Al Bahrani M, Al-Zakwani I. The Impact of Demographic, Clinical Characteristics and the Various COVID-19 Variant Types on All-Cause Mortality: A Case-Series Retrospective Study. Diseases 2022; 10:100. [PMID: 36412594 PMCID: PMC9680441 DOI: 10.3390/diseases10040100] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 11/01/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022] Open
Abstract
(1) Background: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has rapidly evolved into a pandemic affecting virtually every country in the world. We evaluated the demographic, clinical, laboratory, and all-cause mortality of moderate and severe COVID-19 patients admitted to a tertiary care hospital in Oman during the different COVID-19 waves and variant types. (2) Methods: A case-series retrospective study was carried out between 12 March 2020 and 30 June 2022. All adults over the age of 18 with laboratory-confirmed COVID-19 were enrolled. Analyses were performed using univariate and multivariate statistics. (3) Results: A total of 1462 confirmed cases enrolled with the mean age of the cohort was 55 ± 17 years with significant differences among the groups (p = 0.006). A total of 63% and 80% of the patients were males and citizens of Oman, respectively. Patients infected with the Alpha COVID-19 variant type were more likely to have acute respiratory distress syndrome (ARDS) (p < 0.001), stay longer in the hospital (p < 0.001), and get admitted to the intensive care unit (ICU) (p < 0.001). At the same time, those who had the Omicron COVID-19 type were more likely to have renal impairment (p < 0.001) and less likely to be associated with non-invasive ventilation (NIV) (p = 0.001) compared with other COVID-19 variant types. The Delta (adjusted odds ratio (aOR), 1.8; 95% confidence interval (CI): 1.22−2.66; p = 0.003) and Omicron (aOR, 1.88; 95% CI: 1.09−3.22; p = 0.022) COVID-19 variant types were associated with higher all-cause mortality when compared to the initial COVID-19 variant. Old age (aOR, 1.05; 95% CI: 1.04−1.06; p < 0.001), the presence of respiratory disease (aOR, 1.58; 95% CI: 1.02−2.44; p = 0.04), ICU admission (aOR, 3.41; 95% CI: 2.16−5.39; p < 0.001), lower eGFR (aOR, 1.61; 95% CI: 1.17−2.23; p = 0.004), and ARDS (aOR, 5.75; 95% CI: 3.69−8.98; p < 0.001) were also associated with higher mortality while NIV requirements were associated with lower odds of dying (aOR, 0.65; 95% CI: 0.46−0.91; p = 0.012). (4) Conclusions: Alpha and Delta variants were associated with a longer hospital stay, need for intensive care, mechanical ventilation, and increased mortality. Old age, cardiac renal dysfunction were commonly associated with Omicron variants. Large-scale national studies to further assess the risk factors for mortality related to COVID-19 waves are warranted.
Collapse
Affiliation(s)
- Faryal Khamis
- Department of Infectious Disease, The Royal Hospital, Muscat, PC 111, Oman
| | | | - Muna Ba’Omar
- Department of Infectious Disease, The Royal Hospital, Muscat, PC 111, Oman
| | - Wessam Osman
- Department of Medicine, Royal Hospital, Ministry of Health, Muscat, PC 111, Oman
| | - Shabnam Chhetri
- Department of Infectious Disease, The Royal Hospital, Muscat, PC 111, Oman
| | - Zaiyana Ambusaid
- Department of Medicine, Royal Hospital, Ministry of Health, Muscat, PC 111, Oman
| | - Zakariya Al Fahdi
- Department of Medicine, Nizwa Hospital, Ministry of Health, Nizwa, PC 611, Oman
| | - Jaber Al Lawati
- Department of Medicine, Royal Hospital, Ministry of Health, Muscat, PC 111, Oman
| | - Khalsa Al Sulaimi
- Department of Medicine, Royal Hospital, Ministry of Health, Muscat, PC 111, Oman
| | | | - Maher Al Bahrani
- Department of Anaesthesia, Royal Hospital, Ministry of Health, Muscat, PC 111, Oman
| | - Ibrahim Al-Zakwani
- Department of Pharmacology & Clinical Pharmacy, College of Medicine & Health Sciences, Sultan Qaboos University, Al Khoudh, PC 123, Oman
| |
Collapse
|
18
|
Zhang M, Zhang P, Liang Y, Du B, Li L, Yu Z, Wang H, Wang Q, Zhang X, Zhang W. A systematic review of current status and challenges of vaccinating children against SARS-CoV-2. J Infect Public Health 2022; 15:1212-1224. [PMID: 36257126 PMCID: PMC9557115 DOI: 10.1016/j.jiph.2022.10.006] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 09/22/2022] [Accepted: 10/04/2022] [Indexed: 12/04/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has inflicted immense damage to countries, economies and societies worldwide. Authorized COVID-19 vaccines based on different platforms have been widely inoculated in adults, showing up to 100% immunogenicity with significant efficacy in preventing SARS-CoV-2 infections and the occurrence of severe COVID-19. It has also greatly slowed the evolution of SARS-CoV-2 variants, as shown in clinical trials and real-world evidence. However, the total dosage of COVID-19 vaccines for children is much smaller than that for adults due to limitations from parental concern of vaccine safety, presenting a potential obstacle in ending the COVID-19 pandemic. SARS-CoV-2 not only increases the risk of severe multisystem inflammatory syndrome (MIS-C) in children, but also negatively affects children's psychology and academics, indirectly hindering the maintenance and progress of normal social order. Therefore, this article examines the clinical manifestations of children infected with SARS-CoV-2, the status of vaccination against COVID-19 in children, vaccination-related adverse events, and the unique immune mechanisms of children. In particular, the necessity and challenges of vaccinating children against SARS-CoV-2 were highlighted from the perspectives of society and family. In summary, parental hesitancy is unnecessary as adverse events after COVID-19 vaccination have been proven to be infrequent, comprise of mild symptoms, and have a good prognosis.
Collapse
Affiliation(s)
- Mengxin Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Pin Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Ying Liang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Bang Du
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Lifeng Li
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Zhidan Yu
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Huanmin Wang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China
| | - Qionglin Wang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China.
| | - Xianwei Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China.
| | - Wancun Zhang
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou 450018, China.
| |
Collapse
|
19
|
Kurpas MK, Jaksik R, Kuś P, Kimmel M. Genomic Analysis of SARS-CoV-2 Alpha, Beta and Delta Variants of Concern Uncovers Signatures of Neutral and Non-Neutral Evolution. Viruses 2022; 14:2375. [PMID: 36366473 PMCID: PMC9695218 DOI: 10.3390/v14112375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 01/31/2023] Open
Abstract
Due to the emergence of new variants of the SARS-CoV-2 coronavirus, the question of how the viral genomes evolved, leading to the formation of highly infectious strains, becomes particularly important. Three major emergent strains, Alpha, Beta and Delta, characterized by a significant number of missense mutations, provide a natural test field. We accumulated and aligned 4.7 million SARS-CoV-2 genomes from the GISAID database and carried out a comprehensive set of analyses. This collection covers the period until the end of October 2021, i.e., the beginnings of the Omicron variant. First, we explored combinatorial complexity of the genomic variants emerging and their timing, indicating very strong, albeit hidden, selection forces. Our analyses show that the mutations that define variants of concern did not arise gradually but rather co-evolved rapidly, leading to the emergence of the full variant strain. To explore in more detail the evolutionary forces at work, we developed time trajectories of mutations at all 29,903 sites of the SARS-CoV-2 genome, week by week, and stratified them into trends related to (i) point substitutions, (ii) deletions and (iii) non-sequenceable regions. We focused on classifying the genetic forces active at different ranges of the mutational spectrum. We observed the agreement of the lowest-frequency mutation spectrum with the Griffiths-Tavaré theory, under the Infinite Sites Model and neutrality. If we widen the frequency range, we observe the site frequency spectra much more consistently with the Tung-Durrett model assuming clone competition and selection. The coefficients of the fitting model indicate the possibility of selection acting to promote gradual growth slowdown, as observed in the history of the variants of concern. These results add up to a model of genomic evolution, which partly fits into the classical drift barrier ideas. Certain observations, such as mutation "bands" persistent over the epidemic history, suggest contribution of genetic forces different from mutation, drift and selection, including recombination or other genome transformations. In addition, we show that a "toy" mathematical model can qualitatively reproduce how new variants (clones) stem from rare advantageous driver mutations, and then acquire neutral or disadvantageous passenger mutations which gradually reduce their fitness so they can be then outcompeted by new variants due to other driver mutations.
Collapse
Affiliation(s)
- Monika Klara Kurpas
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland; (M.K.K.); (R.J.); (P.K.)
| | - Roman Jaksik
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland; (M.K.K.); (R.J.); (P.K.)
| | - Pawel Kuś
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland; (M.K.K.); (R.J.); (P.K.)
| | - Marek Kimmel
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Akademicka 16, 44-100 Gliwice, Poland; (M.K.K.); (R.J.); (P.K.)
- Department of Statistics and Bioengineering, Rice University, 6100 Main Street, Houston, TX 77005, USA
| |
Collapse
|
20
|
Zhang Y, Yang Y, Qiao N, Wang X, Ding L, Zhu X, Liang Y, Han Z, Liu F, Zhang X, Yang X. Early assessment of the safety and immunogenicity of a third dose (booster) of COVID-19 immunization in Chinese adults. Front Med 2022; 16:93-101. [PMID: 35122211 PMCID: PMC8815383 DOI: 10.1007/s11684-021-0914-x] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022]
Abstract
Inducing durable and effective immunity against severe acute respiratory syndrome Coronavirus 2 (SARS-CoV-2) via vaccination is essential to combat the current pandemic of coronavirus disease 2019 (COVID-19). It has been noticed that the strength of anti-COVID-19 vaccination-induced immunity fades over time, which calls for an additional vaccination regime, as known as booster immunization, to restore immunity among previously vaccinated populations. Here we report a pilot open-label trial of a third dose of BBIBP-CorV, an inactivated SARS-CoV-2 vaccine (Vero cell), on 136 participants aged between 18 to 63 years. Safety and immunogenicity in terms of neutralizing antibody titers and cytokine/chemokine responses were analyzed as the main endpoint until day 28. While systemic reactogenicity was either absent or mild, SARS-CoV-2-specific neutralizing antibody titers rapidly arose in all participants within 4 weeks, surpassing the peak antibody titers elicited by the initial two-dose immunization regime. Broad increases of cellular immunity-associated cytokines and chemokines were also detected in the majority of participants after the third vaccination. Furthermore, in an exploratory study, a newly developed recombinant protein vaccine, NVSI-06-08 (CHO Cells), was found to be safe and even more effective than BBIBP-CorV in eliciting humoral immune responses in BBIBP-CorV-primed individuals. Together, these results indicate that a third immunization schedule with either homologous or heterologous vaccine showed favorable safety profiles and restored potent SARS-CoV-2-specific immunity, providing support for further trials of booster vaccination in larger populations.
Collapse
Affiliation(s)
- Yuntao Zhang
- China National Biotec Group Company Limited, Beijing, 100024, China
| | - Yunkai Yang
- China National Biotec Group Company Limited, Beijing, 100024, China
| | - Niu Qiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xuewei Wang
- China National Biotec Group Company Limited, Beijing, 100024, China
| | - Ling Ding
- Beijing Institute of Biological Products, China National Biotec Group Company Limited, Beijing, 100176, China
| | - Xiujuan Zhu
- Beijing Institute of Biological Products, China National Biotec Group Company Limited, Beijing, 100176, China
| | - Yu Liang
- National Vaccine and Serum Institute, China National Biotec Group Company Limited, Beijing, 101111, China
| | - Zibo Han
- National Vaccine and Serum Institute, China National Biotec Group Company Limited, Beijing, 101111, China
| | - Feng Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xinxin Zhang
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Xiaoming Yang
- China National Biotec Group Company Limited, Beijing, 100024, China.
| |
Collapse
|
21
|
Zaremba A, Zabolotny Institute of Microbiology and Virology NAS of Ukraine, Zaremba P, Budzanivska I, Zahorodnia S, Zabolotny Institute of Microbiology and Virology NAS of Ukraine, Taras Shevchenko National University of Kyiv, Zabolotny Institute of Microbiology and Virology NAS of Ukraine. PATTERNS OF THE INFLUENCE OF VACCINATION ON THE DYNAMICS OF DIFFERENT SARS-COV-2 VARIANTS SPREAD. TWO-YEAR ANALYSIS. BULLETIN OF TARAS SHEVCHENKO NATIONAL UNIVERSITY OF KYIV. SERIES: BIOLOGY 2022. [DOI: 10.17721/1728.2748.2022.89.39-45] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The new coronavirus, now known as SARS-CoV-2, has attracted the attention of thousands of scientists around the world. This attention is primarily due to the significant impact of this pathogen on the economic and social aspects of the existence of a large number of people. Rapid and powerful anti-epidemic measures in most countries have led to a slowdown in the pandemic. And with the creation of a number of effective vaccines against SARS-CoV-2, we have learned to counteract its spread in the field of resistance of each individual. Which obviously created an additional selection factor. And according to the classical concept of the host-parasite system, human impact on SARS-CoV-2 theoretically requires appropriate adaptive changes of the latter. In this work, by analyzing the statistical data available in open sources, we try to identify and study the existing patterns of the impact of vaccination on the dynamics of the spread of different SARS-CoV-2 variants for the period from the beginning of the pandemic to November 2021.
Collapse
|