1
|
Sharanya CS, Wilbee DS, Sathi SN, Natarajan K. Computational screening combined with well-tempered metadynamics simulations identifies potential TMPRSS2 inhibitors. Sci Rep 2024; 14:16197. [PMID: 39003338 PMCID: PMC11246518 DOI: 10.1038/s41598-024-65296-7] [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: 05/10/2024] [Accepted: 06/19/2024] [Indexed: 07/15/2024] Open
Abstract
Type-II transmembrane serine proteases are effective pharmacological targets for host defence against viral entry and in certain cancer cell progressions. These serine proteases cleave viral spike proteins to expose the fusion peptide for cell entry, which is essential to the life cycle of the virus. TMPRSS2 inhibitors can also fight against respiratory viruses that employ them for cell entry. Our study combining virtual screening, all-atom molecular dynamics, and well-tempered metadynamics simulation identifies vicenin-2, neohesperidin, naringin, and rhoifolin as promising TMPRSS2 antagonists. The binding energies obtained are - 16.3, - 15.4, - 13.6, and - 13.8 kcal/mol for vicenin-2, neohesperidin, naringin, and rhoifolin respectively. The RMSD, RMSF, PCA, DCCM, and binding free energy profiles also correlate with the stable binding of these ligands at the active site of TMPRSS2. The study reveals that these molecules could be promising lead molecules for combating future outbreaks of coronavirus and other respiratory viruses.
Collapse
Affiliation(s)
- C S Sharanya
- Transdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - D Sasikala Wilbee
- Transdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
- College of Pharmaceutical Sciences, Government Medical College, Thiruvananthapuram, Kerala, India
| | - Shijulal Nelson Sathi
- Bioinformatics Laboratory, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India
| | - Kathiresan Natarajan
- Transdisciplinary Biology, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, India.
| |
Collapse
|
2
|
Islam S, Parves MR, Islam MJ, Ali MA, Efaz FM, Hossain MS, Ullah MO, Halim MA. Structural and functional effects of the L84S mutant in the SARS-COV-2 ORF8 dimer based on microsecond molecular dynamics study. J Biomol Struct Dyn 2024; 42:5770-5787. [PMID: 37403295 DOI: 10.1080/07391102.2023.2228919] [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: 01/10/2023] [Accepted: 06/17/2023] [Indexed: 07/06/2023]
Abstract
The L84S mutation has been observed frequently in the ORF8 protein of SARS-CoV-2, which is an accessory protein involved in various important functions such as virus propagation, pathogenesis, and evading the immune response. However, the specific effects of this mutation on the dimeric structure of ORF8 and its impacts on interactions with host components and immune responses are not well understood. In this study, we performed one microsecond molecular dynamics (MD) simulation and analyzed the dimeric behavior of the L84S and L84A mutants in comparison to the native protein. The MD simulations revealed that both mutations caused changes in the conformation of the ORF8 dimer, influenced protein folding mechanisms, and affected the overall structural stability. In particular, the 73YIDI76 motif has found to be significantly affected by the L84S mutation, leading to structural flexibility in the region connecting the C-terminal β4 and β5 strands. This flexibility might be responsible for virus immune modulation. The free energy landscape (FEL) and principle component analysis (PCA) have also supported our investigation. Overall, the L84S and L84A mutations affect the ORF8 dimeric interfaces by reducing the frequency of protein-protein interacting residues (Arg52, Lys53, Arg98, Ile104, Arg115, Val117, Asp119, Phe120, and Ile121) in the ORF8 dimer. Our findings provide detail insights for further research in designing structure-based therapeutics against the SARS-CoV-2.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Shafiqul Islam
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Rimon Parves
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Jahirul Islam
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Ackas Ali
- Department of Chemistry and Biochemistry, Kennesaw State University, Kennesaw, GA, USA
| | - Faiyaz Md Efaz
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Md Shahadat Hossain
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - M Obayed Ullah
- Division of Infectious disease and Division of Computer Aided Drug Design, The Red-Green Research Centre, Dhaka, Bangladesh
| | - Mohammad A Halim
- Department of Chemistry and Biochemistry, Kennesaw State University, Kennesaw, GA, USA
| |
Collapse
|
3
|
Maiti AK. Progressive Evolutionary Dynamics of Gene-Specific ω Led to the Emergence of Novel SARS-CoV-2 Strains Having Super-Infectivity and Virulence with Vaccine Neutralization. Int J Mol Sci 2024; 25:6306. [PMID: 38928018 PMCID: PMC11204377 DOI: 10.3390/ijms25126306] [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: 05/06/2024] [Revised: 05/21/2024] [Accepted: 05/29/2024] [Indexed: 06/28/2024] Open
Abstract
An estimation of the proportion of nonsynonymous to synonymous mutation (dn/ds, ω) of the SARS-CoV-2 genome would indicate the evolutionary dynamics necessary to evolve into novel strains with increased infection, virulence, and vaccine neutralization. A temporal estimation of ω of the whole genome, and all twenty-nine SARS-CoV-2 genes of major virulent strains of alpha, delta and omicron demonstrates that the SARS-CoV-2 genome originally emerged (ω ~ 0.04) with a strong purifying selection (ω < 1) and reached (ω ~ 0.85) in omicron towards diversifying selection (ω > 1). A marked increase in the ω occurred in the spike gene from alpha (ω = 0.2) to omicron (ω = 1.97). The ω of the replication machinery genes including RDRP, NSP3, NSP4, NSP7, NSP8, NSP10, NSP13, NSP14, and ORF9 are markedly increased, indicating that these genes/proteins are yet to be evolutionary stabilized and are contributing to the evolution of novel virulent strains. The delta-specific maximum increase in ω in the immunomodulatory genes of NSP8, NSP10, NSP16, ORF4, ORF5, ORF6, ORF7A, and ORF8 compared to alpha or omicron indicates delta-specific vulnerabilities for severe COVID-19 related hospitalization and death. The maximum values of ω are observed for spike (S), NSP4, ORF8 and NSP15, which indicates that the gene-specific temporal estimation of ω identifies specific genes for its super-infectivity and virulency that could be targeted for drug development.
Collapse
Affiliation(s)
- Amit K Maiti
- Department of Genetics and Genomics, Mydnavar, 28475 Greenfield Rd, Southfield, MI 48076, USA
| |
Collapse
|
4
|
Kumar V, Shefrin S, Sundar D. Molecular insights into the binding interactions and energetics of the omicron spike variant with hACE2 and a neutralizing antibody. J Struct Biol 2024; 216:108087. [PMID: 38494148 DOI: 10.1016/j.jsb.2024.108087] [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: 01/29/2024] [Revised: 03/11/2024] [Accepted: 03/15/2024] [Indexed: 03/19/2024]
Abstract
The global spread of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) since 2019 has led to a continuous evolution of viral variants, with the latest concern being the Omicron (B.1.1.529) variant. In this study, classical molecular dynamics simulations were conducted to elucidate the biophysical aspects of the Omicron spike protein's receptor-binding domain (RBD) in its interaction with human angiotensin-converting enzyme 2 (hACE2) and a neutralizing antibody, comparing it to the wildtype (WT). To model the Omicron variant, 15 in silico mutations were introduced in the RBD region of WT (retrieved from PDB). The simulations of WT spike-hACE2 and Omicron spike-hACE2 complexes revealed comparable binding stability and dynamics. Notably, the Q493R mutation in the Omicron spike increased interactions with hACE2, particularly with ASP38 and ASP355. Additionally, mutations such as N417K, T478K, and Y505H contributed to enhanced structural stability in the Omicron variant. Conversely, when comparing WT with Omicron in complex with a neutralizing antibody, simulation results demonstrated poorer binding dynamics and stability for the Omicron variant. The E484K mutation significantly decreased binding interactions, resulting in an overall decrease in binding energy (∼-57 kcal/mol) compared to WT (∼-84 kcal/mol). This study provides valuable molecular insights into the heightened infectivity of the Omicron variant, shedding light on the specific mutations influencing its interactions with hACE2 and neutralizing antibodies.
Collapse
Affiliation(s)
- Vipul Kumar
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, New Delhi 110016, India.
| | - Seyad Shefrin
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, New Delhi 110016, India.
| | - Durai Sundar
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology (IIT) Delhi, New Delhi 110016, India.
| |
Collapse
|
5
|
Scovino AM, Dahab EC, Diniz-Lima I, de Senna Silveira E, Barroso SPC, Cardoso KM, Nico D, Makhoul GJ, da Silva-Junior EB, Freire-de-Lima CG, Freire-de-Lima L, da Fonseca LM, Valente N, Nacife V, Machado A, Araújo M, Vieira GF, Pauvolid-Corrêa A, Siqueira M, Morrot A. A Comparative Analysis of Innate Immune Responses and the Structural Characterization of Spike from SARS-CoV-2 Gamma Variants and Subvariants. Microorganisms 2024; 12:720. [PMID: 38674664 PMCID: PMC11052025 DOI: 10.3390/microorganisms12040720] [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: 09/05/2023] [Revised: 10/16/2023] [Accepted: 11/28/2023] [Indexed: 04/28/2024] Open
Abstract
The SARS-CoV-2 P.1 variant, responsible for an outbreak in Manaus, Brazil, is distinguished by 12 amino acid differences in the S protein, potentially increasing its ACE-2 affinity and immune evasion capability. We investigated the innate immune response of this variant compared to the original B.1 strain, particularly concerning cytokine production. Blood samples from three severe COVID-19 patients were analyzed post-infection with both strains. Results showed no significant difference in cytokine production of mononuclear cells and neutrophils for either variant. While B.1 had higher cytopathogenicity, neither showed viral replication in mononuclear cells. Structural analyses of the S protein highlighted physicochemical variations, which might be linked to the differences in infectivity between the strains. Our studies point to the increased infectivity of P.1 could stem from altered immunogenicity and receptor-binding affinity.
Collapse
Affiliation(s)
- Aline Miranda Scovino
- Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil (E.C.D.); (D.N.)
- Laboratório de Imunoparasitologia, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-360, Brazil
| | - Elizabeth Chen Dahab
- Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil (E.C.D.); (D.N.)
- Laboratório de Imunoparasitologia, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-360, Brazil
| | - Israel Diniz-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (I.D.-L.); (G.J.M.); (E.B.d.S.-J.); (C.G.F.-d.-L.); (L.F.-d.-L.)
| | - Etiele de Senna Silveira
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 91501-970, Brazil; (E.d.S.S.)
| | - Shana Priscila Coutinho Barroso
- Laboratório de Biologia Molecular, Instituto de Pesquisa Biomédica, Hospital Naval Marcílio Dias, Marinha do Brazil, Rio de Janeiro 20725-090, Brazil; (S.P.C.B.); (K.M.C.)
- Biomanguinhos, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-900, Brazil
| | - Karina Martins Cardoso
- Laboratório de Biologia Molecular, Instituto de Pesquisa Biomédica, Hospital Naval Marcílio Dias, Marinha do Brazil, Rio de Janeiro 20725-090, Brazil; (S.P.C.B.); (K.M.C.)
| | - Dirlei Nico
- Instituto de Microbiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil (E.C.D.); (D.N.)
| | - Gustavo José Makhoul
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (I.D.-L.); (G.J.M.); (E.B.d.S.-J.); (C.G.F.-d.-L.); (L.F.-d.-L.)
| | - Elias Barbosa da Silva-Junior
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (I.D.-L.); (G.J.M.); (E.B.d.S.-J.); (C.G.F.-d.-L.); (L.F.-d.-L.)
| | - Celio Geraldo Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (I.D.-L.); (G.J.M.); (E.B.d.S.-J.); (C.G.F.-d.-L.); (L.F.-d.-L.)
| | - Leonardo Freire-de-Lima
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (I.D.-L.); (G.J.M.); (E.B.d.S.-J.); (C.G.F.-d.-L.); (L.F.-d.-L.)
| | - Leonardo Marques da Fonseca
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-902, Brazil; (I.D.-L.); (G.J.M.); (E.B.d.S.-J.); (C.G.F.-d.-L.); (L.F.-d.-L.)
- Curso de Medicina, Universidade Castelo Branco (UCB), Rio de Janeiro 21710-255, Brazil
| | - Natalia Valente
- Laboratório de Vírus Respiratórios e Sarampo, COVID-19 National Reference Laboratory of Brazil and World Health Organization COVID-19 Reference Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-360, Brazil; (N.V.); (V.N.); (A.M.); (A.P.-C.)
| | - Valeria Nacife
- Laboratório de Vírus Respiratórios e Sarampo, COVID-19 National Reference Laboratory of Brazil and World Health Organization COVID-19 Reference Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-360, Brazil; (N.V.); (V.N.); (A.M.); (A.P.-C.)
| | - Ana Machado
- Laboratório de Vírus Respiratórios e Sarampo, COVID-19 National Reference Laboratory of Brazil and World Health Organization COVID-19 Reference Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-360, Brazil; (N.V.); (V.N.); (A.M.); (A.P.-C.)
| | - Mia Araújo
- Laboratório de Vírus Respiratórios e Sarampo, COVID-19 National Reference Laboratory of Brazil and World Health Organization COVID-19 Reference Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-360, Brazil; (N.V.); (V.N.); (A.M.); (A.P.-C.)
| | - Gustavo Fioravanti Vieira
- Programa de Pós-Graduação em Genética e Biologia Molecular, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre 91501-970, Brazil; (E.d.S.S.)
- PPGSDH—Programa de Pós-Graduação em Saúde e Desenvolvimento Humano, Universidade La Salle, Canoas 92010-000, Brazil
| | - Alex Pauvolid-Corrêa
- Laboratório de Vírus Respiratórios e Sarampo, COVID-19 National Reference Laboratory of Brazil and World Health Organization COVID-19 Reference Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-360, Brazil; (N.V.); (V.N.); (A.M.); (A.P.-C.)
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA
- Laboratório de Virologia Veterinária de Viçosa, Departamento de Veterinária, Universidade Federal de Viçosa, Viçosa 36570-900, Brazil
| | - Marilda Siqueira
- Laboratório de Vírus Respiratórios e Sarampo, COVID-19 National Reference Laboratory of Brazil and World Health Organization COVID-19 Reference Laboratory, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-360, Brazil; (N.V.); (V.N.); (A.M.); (A.P.-C.)
| | - Alexandre Morrot
- Laboratório de Imunoparasitologia, Fundação Oswaldo Cruz (Fiocruz), Rio de Janeiro 21040-360, Brazil
- Escola de Medicina, Universidade Federal do Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| |
Collapse
|
6
|
Seth A, Liu Y, Gupta R, Wang Z, Mittal E, Kolla S, Rathi P, Gupta P, Parikh BA, Genin GM, Gandra S, Storch GA, Philips JA, George IA, Singamaneni S. Plasmon-Enhanced Digital Fluoroimmunoassay for Subfemtomolar Detection of Protein Biomarkers. NANO LETTERS 2024; 24:229-237. [PMID: 38146928 DOI: 10.1021/acs.nanolett.3c03789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2023]
Abstract
Rapid and accurate quantification of low-abundance protein biomarkers in biofluids can transform the diagnosis of a range of pathologies, including infectious diseases. Here, we harness ultrabright plasmonic fluors as "digital nanolabels" and demonstrate the detection and quantification of subfemtomolar concentrations of human IL-6 and SARS-CoV-2 alpha and variant proteins in clinical nasopharyngeal swab and saliva samples from COVID-19 patients. The resulting digital plasmonic fluor-linked immunosorbent assay (digital p-FLISA) enables detection of SARS-CoV-2 nucleocapsid protein, both in solution and in live virions. Digital p-FLISA outperforms the "gold standard" enzyme-linked immunosorbent assay (ELISA), having a nearly 7000-fold lower limit-of-detection, and outperforms a commercial antigen test, having over 5000-fold improvement in analytical sensitivity. Detection and quantification of very low concentrations of target proteins holds potential for early detection of pathological conditions, treatment monitoring, and personalized medicine.
Collapse
Affiliation(s)
- Anushree Seth
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Yuxiong Liu
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Rohit Gupta
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Zheyu Wang
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Ekansh Mittal
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63130, United States
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63130, United States
| | - Samhitha Kolla
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Priya Rathi
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Prashant Gupta
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Bijal A Parikh
- Department of Pathology and Immunology, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| | - Guy M Genin
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
| | - Sumanth Gandra
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63130, United States
| | - Gregory A Storch
- Department of Pediatrics, Washington University School of Medicine, St. Louis, Missouri 63110, United States
| | - Jennifer A Philips
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri 63130, United States
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63130, United States
| | - Ige A George
- Division of Infectious Diseases, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63130, United States
| | - Srikanth Singamaneni
- Department of Mechanical Engineering and Materials Science, Institute of Materials Science and Engineering, Washington University in St. Louis, St. Louis, Missouri 63130, United States
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, Missouri 63110, United States
| |
Collapse
|
7
|
Pillay A, Yeola A, Tea F, Denkova M, Houston S, Burrell R, Merheb V, Lee FXZ, Lopez JA, Moran L, Jadhav A, Sterling K, Lai CL, Vitagliano TL, Aggarwal A, Catchpoole D, Wood N, Phan TG, Nanan R, Hsu P, Turville SG, Britton PN, Brilot F. Infection and Vaccine Induced Spike Antibody Responses Against SARS-CoV-2 Variants of Concern in COVID-19-Naïve Children and Adults. J Clin Immunol 2023; 43:1706-1723. [PMID: 37405544 PMCID: PMC10661752 DOI: 10.1007/s10875-023-01540-5] [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: 03/06/2023] [Accepted: 06/19/2023] [Indexed: 07/06/2023]
Abstract
Although a more efficient adaptive humoral immune response has been proposed to underlie the usually favorable outcome of pediatric COVID-19, the breadth of viral and vaccine cross-reactivity toward the ever-mutating Spike protein among variants of concern (VOCs) has not yet been compared between children and adults. We assessed antibodies to conformational Spike in COVID-19-naïve children and adults vaccinated by BNT162b2 and ChAdOx1, and naturally infected with SARS-CoV-2 Early Clade, Delta, and Omicron. Sera were analyzed against Spike including naturally occurring VOCs Alpha, Beta, Gamma, Delta, and Omicron BA.1, BA.2, BA.5, BQ.1.1, BA2.75.2, and XBB.1, and variants of interest Epsilon, Kappa, Eta, D.2, and artificial mutant Spikes. There was no notable difference between breadth and longevity of antibody against VOCs in children and adults. Vaccinated individuals displayed similar immunoreactivity profiles across variants compared with naturally infected individuals. Delta-infected patients had an enhanced cross-reactivity toward Delta and earlier VOCs compared to patients infected by Early Clade SARS-CoV-2. Although Omicron BA.1, BA.2, BA.5, BQ.1.1, BA2.75.2, and XBB.1 antibody titers were generated after Omicron infection, cross-reactive binding against Omicron subvariants was reduced across all infection, immunization, and age groups. Some mutations, such as 498R and 501Y, epistatically combined to enhance cross-reactive binding, but could not fully compensate for antibody-evasive mutations within the Omicron subvariants tested. Our results reveal important molecular features central to the generation of high antibody titers and broad immunoreactivity that should be considered in future vaccine design and global serosurveillance in the context of limited vaccine boosters available to the pediatric population.
Collapse
Affiliation(s)
- Aleha Pillay
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Avani Yeola
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Fiona Tea
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Martina Denkova
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Samuel Houston
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Rebecca Burrell
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Vera Merheb
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Fiona X Z Lee
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Joseph A Lopez
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Lilly Moran
- Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
- National Center for Immunisation Research and Surveillance, the Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
| | - Ajay Jadhav
- Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
- National Center for Immunisation Research and Surveillance, the Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
| | - Katrina Sterling
- Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
- National Center for Immunisation Research and Surveillance, the Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
| | - Catherine L Lai
- Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Tennille L Vitagliano
- Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Anupriya Aggarwal
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Dan Catchpoole
- Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Nicholas Wood
- Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
- National Center for Immunisation Research and Surveillance, the Sydney Children's Hospitals Network, Sydney, New South Wales, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Healthcare Clinical Campus, School of Clinical Medicine, Faculty of Medicine and Health, The University of New South Wales, Sydney, New South Wales, Australia
| | - Ralph Nanan
- Charles Perkins Center and Sydney Medical School Nepean, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Peter Hsu
- Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Department of Allergy and Immunology, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
| | - Stuart G Turville
- The Kirby Institute, The University of New South Wales, Sydney, New South Wales, Australia
| | - Philip N Britton
- Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Sydney Institute for Infectious Disease, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
| | - Fabienne Brilot
- Brain Autoimmunity Group, Kids Neuroscience Centre, Kids Research at the Children's Hospital at Westmead, Sydney, New South Wales, Australia.
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
- Sydney Institute for Infectious Disease, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
8
|
Song JB, Zhao LQ, Wen HP, Li YP. Herbal combinations against COVID-19: A network pharmacology, molecular docking and dynamics study. JOURNAL OF INTEGRATIVE MEDICINE 2023; 21:593-604. [PMID: 37805293 DOI: 10.1016/j.joim.2023.09.001] [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: 07/05/2022] [Accepted: 05/13/2023] [Indexed: 10/09/2023]
Abstract
OBJECTIVE The aim of this study is to identify molecules from traditional Chinese medicine (TCM) with potential activity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its variants. METHODS We applied the Apriori algorithm to identify important combinations of herbs in the TCM prescriptions for the treatment of coronavirus disease 2019 (COVID-19). Then, we explored the active components and core targets using network pharmacology. In addition, the molecular docking approach was performed to investigate the interaction of these components with the main structural and non-structural proteins, as well as the mutants. Furthermore, their stability in the binding pockets was further evaluated with the molecular dynamics approach. RESULTS A combination of Amygdalus Communis Vas., Ephedra Herba and Scutellaria baicalensis Georgi was selected as the important herbal combination, and 11 main components and 20 core targets against COVID-19 were obtained. These components, including luteolin, naringenin, stigmasterol, baicalein, and so on, were the potentially active compounds against COVID-19. The binding affinity of these compounds with the potential targets was as high as the positive controls. Among them, baicalein could interfere with multiple targets simultaneously, and it also interfered with the interaction between spike protein and angiotensin-converting enzyme 2 receptor. Additionally, almost all the systems reached stability during dynamics simulation. CONCLUSION The combination of A. communis, Ephedra Herba and S. baicalensis was the most important herbal combination for the treatment of COVID-19. Baicalein may be a potential candidate against SARS-CoV-2 and its variants. Please cite this article as: Song JB, Zhao LQ, Wen HP, Li YP. Herbal combinations against COVID-19: A network pharmacology, molecular docking and dynamics study. J Integr Med. 2023;21(6):593-604.
Collapse
Affiliation(s)
- Jian-Bo Song
- Department of Pharmacy, the Fifth Affiliated Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan 030012, Shanxi Province, China
| | - Li-Qin Zhao
- Department of Infectious Disease, the First Affiliated Hospital of Shanxi Medical University, Taiyuan 030012, Shanxi Province, China
| | - Hong-Ping Wen
- Department of Pharmacy, the Fifth Affiliated Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan 030012, Shanxi Province, China
| | - Yuan-Ping Li
- Department of Pharmacy, the Fifth Affiliated Hospital of Shanxi Medical University (Shanxi Provincial People's Hospital), Taiyuan 030012, Shanxi Province, China.
| |
Collapse
|
9
|
Jamalidoust M, Eilami O, Ashkan Z, Ziyaeyan M, Aliabadi N, Habibi M. The rates and symptoms of natural and breakthrough infection pre- and post- Covid-19 non-mRNA vaccination at various peaks amongst Iranian healthcare workers. Virol J 2023; 20:182. [PMID: 37596593 PMCID: PMC10436397 DOI: 10.1186/s12985-023-02156-2] [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: 06/03/2023] [Accepted: 08/09/2023] [Indexed: 08/20/2023] Open
Abstract
BACKGROUND/AIMS The aim of this study was to determine the rate of natural and breakthrough infection and related symptoms of Covid-19 amongst Iranian healthcare workers (HCWs) who were vaccinated by different non-mRNA-based vaccines at peak points. METHODS In this cross-sectional study, the RT-PCR test was performed for a total of 10,581 HCWs suspicious of Covid-19 infection. For each HCW, the frequency of SARS-CoV-2 infection and the time of transmission based on vaccination administration time and schedule were examined during different waves of the pandemic. Based on these findings, the study patients were divided into three groups: natural, natural/breakthrough, and breakthrough. RESULTS In total, 53% of the HCWs were exposed to SARS-CoV-2 infection between 1 and 5 times within two years after the current pandemic, while 20.7% and 32.3% experienced natural and breakthrough SARS-CoV-2 infection, respectively. Only 6% of the breakthrough-infected HCWs had naturally contracted SARS-CoV-2 infection during the initial waves. The highest natural peaks of infection occurred during the interval administration of the first and second dose of the first vaccination series, while the single highest peak of breakthrough infection belonged to the Omicron wave. It occurred simultaneously with the administration of the third vaccination dose. On the other hand, the highest rate of reinfection was observed amongst people who had received the Sinopharm and Bharat vaccines full-doses. CONCLUSION This study compared the clinical differences between the two peaks of Omicron and Delta. This study indicates the rates of natural and breakthrough SARS-CoV-2 infections according to vaccination schedules and different waves of the pandemic.
Collapse
Grants
- Department of Virology, Professor Alborzi Clinical Microbiology Research Center, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz- Iran
- Department of Family Medicine and infectious disease, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Biology, Faculty of Basic Science, Shahrekord University, Shahrekord, Iran.
- Statistics and Information Technology Management, Shiraz University of Medical Sciences, Shiraz, Iran
Collapse
Affiliation(s)
- Marzieh Jamalidoust
- Department of Virology, Professor Alborzi Clinical Microbiology Research Center, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, 71937-11351, Iran.
| | - Owrang Eilami
- Department of Family Medicine and Infectious Disease, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Zahra Ashkan
- Department of Biology, Faculty of Basic Science, Shahrekord University, Shahrekord, Iran
| | - Mazyar Ziyaeyan
- Department of Virology, Professor Alborzi Clinical Microbiology Research Center, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, 71937-11351, Iran
| | - Nasrin Aliabadi
- Department of Virology, Professor Alborzi Clinical Microbiology Research Center, Namazi Hospital, Shiraz University of Medical Sciences, Shiraz, 71937-11351, Iran
| | - Mohammad Habibi
- Statistics and Information Technology Management, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
10
|
Jamir E, Sarma H, Priyadarsinee L, Kiewhuo K, Nagamani S, Sastry GN. Polypharmacology guided drug repositioning approach for SARS-CoV2. PLoS One 2023; 18:e0289890. [PMID: 37556478 PMCID: PMC10411734 DOI: 10.1371/journal.pone.0289890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 07/27/2023] [Indexed: 08/11/2023] Open
Abstract
Drug repurposing has emerged as an important strategy and it has a great potential in identifying therapeutic applications for COVID-19. An extensive virtual screening of 4193 FDA approved drugs has been carried out against 24 proteins of SARS-CoV2 (NSP1-10 and NSP12-16, envelope, membrane, nucleoprotein, spike, ORF3a, ORF6, ORF7a, ORF8, and ORF9b). The drugs were classified into top 10 and bottom 10 drugs based on the docking scores followed by the distribution of their therapeutic indications. As a result, the top 10 drugs were found to have therapeutic indications for cancer, pain, neurological disorders, and viral and bacterial diseases. As drug resistance is one of the major challenges in antiviral drug discovery, polypharmacology and network pharmacology approaches were employed in the study to identify drugs interacting with multiple targets and drugs such as dihydroergotamine, ergotamine, bisdequalinium chloride, midostaurin, temoporfin, tirilazad, and venetoclax were identified among the multi-targeting drugs. Further, a pathway analysis of the genes related to the multi-targeting drugs was carried which provides insight into the mechanism of drugs and identifying targetable genes and biological pathways involved in SARS-CoV2.
Collapse
Affiliation(s)
- Esther Jamir
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Himakshi Sarma
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
| | - Lipsa Priyadarsinee
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kikrusenuo Kiewhuo
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Selvaraman Nagamani
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - G. Narahari Sastry
- Advanced Computation and Data Sciences Division, CSIR–North East Institute of Science and Technology, Jorhat, Assam, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
11
|
Shariq M, Malik AA, Sheikh JA, Hasnain SE, Ehtesham NZ. Regulation of autophagy by SARS-CoV-2: The multifunctional contributions of ORF3a. J Med Virol 2023; 95:e28959. [PMID: 37485696 DOI: 10.1002/jmv.28959] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/25/2023]
Abstract
Severe acute respiratory syndrome-coronavirus-1 (SARS-CoV-2) regulates autophagic flux by blocking the fusion of autophagosomes with lysosomes, causing the accumulation of membranous vesicles for replication. Multiple SARS-CoV-2 proteins regulate autophagy with significant roles attributed to ORF3a. Mechanistically, open reading frame 3a (ORF3a) forms a complex with UV radiation resistance associated, regulating the functions of the PIK3C3-1 and PIK3C3-2 lipid kinase complexes, thereby modulating autophagosome biogenesis. ORF3a sequesters VPS39 onto the late endosome/lysosome, inhibiting assembly of the soluble NSF attachement protein REceptor (SNARE) complex and preventing autolysosome formation. ORF3a promotes the interaction between BECN1 and HMGB1, inducing the assembly of PIK3CA kinases into the ER (endoplasmic reticulum) and activating reticulophagy, proinflammatory responses, and ER stress. ORF3a recruits BORCS6 and ARL8B to lysosomes, initiating the anterograde transport of the virus to the plasma membrane. ORF3a also activates the SNARE complex (STX4-SNAP23-VAMP7), inducing fusion of lysosomes with the plasma membrane for viral egress. These mechanistic details can provide multiple targets for inhibiting SARS-CoV-2 by developing host- or host-pathogen interface-based therapeutics.
Collapse
Affiliation(s)
- Mohd Shariq
- Inflammation Biology and Cell Signalling Laboratory, ICMR-National Institute of Pathology, New Delhi, India
| | - Asrar A Malik
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Javaid A Sheikh
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, India
| | - Seyed E Hasnain
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi, India
| | - Nasreen Z Ehtesham
- Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
12
|
Pandit R, Matthews QL. A SARS-CoV-2: Companion Animal Transmission and Variants Classification. Pathogens 2023; 12:775. [PMID: 37375465 DOI: 10.3390/pathogens12060775] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
The continuous emergence of novel viruses and their diseases are a threat to global public health as there have been three outbreaks of coronaviruses that are highly pathogenic to humans in the span of the last two decades, severe acute respiratory syndrome (SARS)-CoV in 2002, Middle East respiratory syndrome (MERS)-CoV in 2012, and novel SARS-CoV-2 which emerged in 2019. The unprecedented spread of SARS-CoV-2 worldwide has given rise to multiple SARS-CoV-2 variants that have either altered transmissibility, infectivity, or immune escaping ability, causing diseases in a broad range of animals including human and non-human hosts such as companion, farm, zoo, or wild animals. In this review, we have discussed the recent SARS-CoV-2 outbreak, potential animal reservoirs, and natural infections in companion and farm animals, with a particular focus on SARS-CoV-2 variants. The expeditious development of COVID-19 vaccines and the advancements in antiviral therapeutics have contained the COVID-19 pandemic to some extent; however, extensive research and surveillance concerning viral epidemiology, animal transmission, variants, or seroprevalence in diverse hosts are essential for the future eradication of COVID-19.
Collapse
Affiliation(s)
- Rachana Pandit
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA
| | - Qiana L Matthews
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA
- Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA
| |
Collapse
|
13
|
Spicuzza L, Campagna D, Di Maria C, Sciacca E, Mancuso S, Vancheri C, Sambataro G. An update on lateral flow immunoassay for the rapid detection of SARS-CoV-2 antibodies. AIMS Microbiol 2023; 9:375-401. [PMID: 37091823 PMCID: PMC10113162 DOI: 10.3934/microbiol.2023020] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/25/2023] Open
Abstract
Over the last three years, after the outbreak of the COVID-19 pandemic, an unprecedented number of novel diagnostic tests have been developed. Assays to evaluate the immune response to SARS-CoV-2 have been widely considered as part of the control strategy. The lateral flow immunoassay (LFIA), to detect both IgM and IgG against SARS-CoV-2, has been widely studied as a point-of-care (POC) test. Compared to laboratory tests, LFIAs are faster, cheaper and user-friendly, thus available also in areas with low economic resources. Soon after the onset of the pandemic, numerous kits for rapid antibody detection were put on the market with an emergency use authorization. However, since then, scientists have tried to better define the accuracy of these tests and their usefulness in different contexts. In fact, while during the first phase of the pandemic LFIAs for antibody detection were auxiliary to molecular tests for the diagnosis of COVID-19, successively these tests became a tool of seroprevalence surveillance to address infection control policies. When in 2021 a massive vaccination campaign was implemented worldwide, the interest in LFIA reemerged due to the need to establish the extent and the longevity of immunization in the vaccinated population and to establish priorities to guide health policies in low-income countries with limited access to vaccines. Here, we summarize the accuracy, the advantages and limits of LFIAs as POC tests for antibody detection, highlighting the efforts that have been made to improve this technology over the last few years.
Collapse
Affiliation(s)
- Lucia Spicuzza
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | | | | | | | | | | | | |
Collapse
|
14
|
Crnković T, Bokor BJ, Lockwood ME, Cornish VW. Peptide Variant Detection by a Living Yeast Biosensor via an Epitope-Selective Protease. BIODESIGN RESEARCH 2023; 5:0003. [PMID: 37849458 PMCID: PMC10084949 DOI: 10.34133/bdr.0003] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 12/08/2022] [Indexed: 10/19/2023] Open
Abstract
We previously demonstrated that we could hijack the fungal pheromone signaling pathway to provide a living yeast biosensor where peptide biomarkers were recognized by G-protein-coupled receptors and engineered to transcribe a readout. Here, we demonstrated that the protease could be reintroduced to the biosensor to provide a simple mechanism for distinguishing single-amino-acid changes in peptide ligands that, otherwise, would likely be difficult to detect using binding-based assays. We characterized the dose-response curves for five fungal pheromone G-protein-coupled receptors, peptides, and proteases-Saccharomyces cerevisiae, Candida albicans, Schizosaccharomyces pombe, Schizosaccharomyces octosporus, and Schizosaccharomyces japonicus. Alanine scanning was carried out for the most selective of these-S. cerevisiae and C. albicans-with and without the protease. Two peptide variants were discovered, which showed diminished cleavage by the protease (CaPep2A and CaPep2A13A). Those peptides were then distinguished by utilizing the biosensor strains with and without the protease, which selectively cleaved and altered the apparent concentration of peptide required for half-maximal activation for 2 peptides-CaPep and CaPep13A, respectively-by more than one order of magnitude. These results support the hypothesis that the living yeast biosensor with a sequence-specific protease can translate single-amino-acid changes into more than one order of magnitude apparent shift in the concentration of peptide required for half-maximal activation. With further engineering by computational modeling and directed evolution, the biosensor could likely distinguish a wide variety of peptide sequences beyond the alanine scanning carried out here. In the future, we envision incorporating proteases into our living yeast biosensor for use as a point of care diagnostic, a scalable communication language, and other applications.
Collapse
Affiliation(s)
- Tea Crnković
- Department of Chemistry, Columbia University, New York, NY 10027, USA
| | - Benjamin J. Bokor
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Mead E. Lockwood
- School of General Studies, Columbia University, New York, NY 10027, USA
| | - Virginia W. Cornish
- Department of Chemistry, Columbia University, New York, NY 10027, USA
- Department of Systems Biology, Columbia University, New York, NY 10027, USA
| |
Collapse
|
15
|
Ichhpujani P, Singh RB, Dhillon HK, Kumar S. Ocular manifestations of COVID-19 in pediatric patients. Ther Adv Ophthalmol 2023; 15:25158414221149916. [PMID: 36938488 PMCID: PMC10015281 DOI: 10.1177/25158414221149916] [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: 03/16/2022] [Accepted: 12/16/2022] [Indexed: 03/15/2023] Open
Abstract
The coronavirus disease-19 (COVID-19) infection may remain asymptomatic or may have several different presentations. Although this disease primarily affects the respiratory system, systemic manifestations affecting the gastrointestinal, cardiovascular, neurological, otorhinolaryngologic, and ophthalmic systems have been reported. Ophthalmic signs may be the first and only sign of COVID-19 infection in children. In the current narrative review, we report the ophthalmic manifestations of COVID-19 in the pediatric age cohort. We performed a comprehensive literature search for the publications on ophthalmic manifestations of COVID-19 in children between 1 March 2020 and 1 January 2022 and compiled the ophthalmic manifestations of this entity among the pediatric population. Conjunctivitis is the most common ophthalmic manifestation in children and can develop at any stage of the disease. Ophthalmic manifestations are seen more commonly in children with severe systemic disease. Long-term and indirect consequence of the COVID-19 disease is the rise of myopia among children. Ophthalmic signs may be the first and only sign of COVID-19 infection in children. Pediatricians, as well as ophthalmologists, must keep observing all children with COVID-19 closely for ophthalmic signs.
Collapse
Affiliation(s)
- Parul Ichhpujani
- Glaucoma Service, Department of Ophthalmology, Government Medical College and Hospital, Sector-32, Chandigarh 160036, India
| | - Rohan Bir Singh
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
- Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
- Discipline of Ophthalmology and Visual Sciences, Faculty of Health and Medical Sciences, Adelaide Medical School, The University of Adelaide, Adelaide, SA, Australia
| | - Hennaav Kaur Dhillon
- Department of Ophthalmology, Government Medical College and Hospital, Chandigarh, India
| | - Suresh Kumar
- Department of Ophthalmology, Government Medical College and Hospital, Chandigarh, India
| |
Collapse
|
16
|
Khan MZI, Nazli A, Al-furas H, Asad MI, Ajmal I, Khan D, Shah J, Farooq MA, Jiang W. An overview of viral mutagenesis and the impact on pathogenesis of SARS-CoV-2 variants. Front Immunol 2022; 13:1034444. [PMID: 36518757 PMCID: PMC9742215 DOI: 10.3389/fimmu.2022.1034444] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 11/04/2022] [Indexed: 11/29/2022] Open
Abstract
Viruses are submicroscopic, obligate intracellular parasites that carry either DNA or RNA as their genome, protected by a capsid. Viruses are genetic entities that propagate by using the metabolic and biosynthetic machinery of their hosts and many of them cause sickness in the host. The ability of viruses to adapt to different hosts and settings mainly relies on their ability to create de novo variety in a short interval of time. The size and chemical composition of the viral genome have been recognized as important factors affecting the rate of mutations. Coronavirus disease 2019 (Covid-19) is a novel viral disease that has quickly become one of the world's leading causes of mortality, making it one of the most serious public health problems in recent decades. The discovery of new medications to cope with Covid-19 is a difficult and time-consuming procedure, as new mutations represent a serious threat to the efficacy of recently developed vaccines. The current article discusses viral mutations and their impact on the pathogenicity of newly developed variants with a special emphasis on Covid-19. The biology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), its mutations, pathogenesis, and treatment strategies are discussed in detail along with the statistical data.
Collapse
Affiliation(s)
| | - Adila Nazli
- Faculty of Biological Sciences, Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Hawaa Al-furas
- International Cooperative Laboratory of Traditional Chinese Medicine Modernization and Innovative Drug Development, Ministry of Education (MOE) of China, Guangzhou City Key Laboratory of Precision Chemical Drug Development, School of Pharmacy, Jinan University, Guangzhou, China
| | - Muhammad Imran Asad
- Faculty of Biological Sciences, Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Iqra Ajmal
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, Shanghai, China
| | - Dildar Khan
- Faculty of Biological Sciences, Department of Pharmacy, Quaid-i-Azam University, Islamabad, Pakistan
| | - Jaffer Shah
- Department of Health, New York, NY, United States,*Correspondence: Jaffer Shah, ; Muhammad Asad Farooq, ; Wenzheng Jiang,
| | - Muhammad Asad Farooq
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, Shanghai, China,*Correspondence: Jaffer Shah, ; Muhammad Asad Farooq, ; Wenzheng Jiang,
| | - Wenzheng Jiang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, East China Normal University, Shanghai, China,*Correspondence: Jaffer Shah, ; Muhammad Asad Farooq, ; Wenzheng Jiang,
| |
Collapse
|
17
|
Evolution of SARS-CoV-2: BA.4/BA.5 Variants Continues to Pose New Challenges. Viruses 2022; 14:v14122610. [PMID: 36560614 PMCID: PMC9781647 DOI: 10.3390/v14122610] [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: 09/20/2022] [Revised: 10/25/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
The acquisition of a high number of mutations, notably, the gain of two mutations L452R and F486V in RBD, and the ability to evade vaccine/natural infection-induced immunity suggests that Omicron is continuing to use "immune-escape potential" as an evolutionary space to maintain a selection advantage within the population. Despite the low hospitalizations and lower death rate, the surges by these variants may offset public health measures and disrupt health care facilities as seen recently in Portugal and the USA. Interestingly these BA.4/BA.5 variants have been found to be more severe than the earlier-emerged Omicron variants. We believe that aggressive COVID-19 surveillance using affordable testing strategies might actually help understand the evolution and transmission pattern of new variants. The sudden dip in reporting of new cases in some of the low- and middle-income countries is an alarming situation and needs to be addressed as this could lead to undetected transmission of future variants of interest/concern of SARS-CoV-2 in large population settings, including advent of a 'super' virus. It would be interesting to examine the possible role/influence, if any, of the two different kinds of vaccines, the spike protein-based versus the inactivated whole virus, in the evolution of BA.4/BA.5.
Collapse
|
18
|
Singh J, Vashishtha S, Rahman SA, Ehtesham NZ, Alam A, Kundu B, Dobrindt U. Energetics of Spike Protein Opening of SARS-CoV-1 and SARS-CoV-2 and Its Variants of Concern: Implications in Host Receptor Scanning and Transmission. Biochemistry 2022; 61:2188-2197. [PMID: 36166360 DOI: 10.1021/acs.biochem.2c00301] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The receptor binding domain(s) (RBD) of spike (S) proteins of SARS-CoV-1 and SARS-CoV-2 (severe acute respiratory syndrome coronavirus) undergoes closed to open transition to engage with host ACE2 receptors. In this study, using multi atomistic (equilibrium) and targeted (non-equilibrium) molecular dynamics simulations, we have compared energetics of RBD opening pathways in full-length (modeled from cryo-EM structures) S proteins of SARS-CoV-1 and SARS-CoV-2. Our data indicate that amino acid variations at the RBD interaction interface can culminate into distinct free energy landscapes of RBD opening in these S proteins. We further report that mutations in the S protein of SARS-CoV-2 variants of concern can reduce the protein-protein interaction affinity of RBD(s) with its neighboring domains and could favor its opening to access ACE2 receptors. The findings can also aid in predicting the impact of future mutations on the rate of S protein opening for rapid host receptor scanning.
Collapse
Affiliation(s)
- Jasdeep Singh
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi 110016, India
| | - Shubham Vashishtha
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi 110016, India
| | - Syed Asad Rahman
- BioInception Pvt. Ltd., Future Business Centre, Kings Hedges Road, Cambridge CB4 2HY, U.K
| | - Nasreen Zafar Ehtesham
- ICMR National Institute of Pathology, Safdarjung Hospital Campus, New Delhi 110029, India
| | - Anwar Alam
- ICMR National Institute of Pathology, Safdarjung Hospital Campus, New Delhi 110029, India
| | - Bishwajit Kundu
- Kusuma School of Biological Sciences, Indian Institute of Technology-Delhi, Hauz Khas, New Delhi 110016, India
| | - Ulrich Dobrindt
- Institute of Hygiene, University of Münster, Münster 48149, Germany
| |
Collapse
|
19
|
Kang B, Lee Y, Lim J, Yong D, Ki Choi Y, Woo Yoon S, Seo S, Jang S, Uk Son S, Kang T, Jung J, Lee KS, Kim MH, Lim EK. FRET-based hACE2 receptor mimic peptide conjugated nanoprobe for simple detection of SARS-CoV-2. CHEMICAL ENGINEERING JOURNAL (LAUSANNE, SWITZERLAND : 1996) 2022; 442:136143. [PMID: 35382003 PMCID: PMC8969299 DOI: 10.1016/j.cej.2022.136143] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 03/15/2022] [Accepted: 03/29/2022] [Indexed: 05/19/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection has led to a pandemic of acute respiratory disease, namely coronavirus disease (COVID-19). This disease threatens human health and public safety. Early diagnosis, isolation, and prevention are important to suppress the outbreak of COVID 19 given the lack of specific antiviral drugs to treat this disease and the emergence of various variants of the virus that cause breakthrough infections even after vaccine administration. Simple and prompt testing is paramount to preventing further spread of the virus. However, current testing methods, namely RT-PCR, is time-consuming. Binding of the SARS-CoV-2 spike (S) glycoprotein to human angiotensin-converting enzyme 2 (hACE2) receptor plays a pivotal role in host cell entry. In the present study, we developed a hACE2 mimic peptide beacon (COVID19-PEB) for simple detection of SARS-CoV-2 using a fluorescence resonance energy transfer system. COVID19-PEB exhibits minimal fluorescence in its ''closed'' hairpin structure; however, in the presence of SARS-CoV-2, the specific recognition of the S protein receptor-binding domain by COVID19-PEB causes the beacon to assume an ''open'' structure that emits strong fluorescence. COVID19-PEB can detect SARS-CoV-2 within 3 h or even 50 min and exhibits strong fluorescence even at low viral concentrations, with a detection limit of 4 × 103 plaque-forming unit/test. Furthermore, in SARS-CoV-2-infected patient samples confirmed using polymerase chain reaction, COVID19-PEB accurately detected the virus. COVID19-PEB could be developed as a rapid and accurate diagnostic tool for COVID-19.
Collapse
Affiliation(s)
- Byunghoon Kang
- BioNanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Youngjin Lee
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Jaewoo Lim
- BioNanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Dongeun Yong
- Department of Laboratory Medicine and Research Institute of Bacterial Resistance, College of Medicine, Yonsei University, 50-1 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Young Ki Choi
- Department of Microbiology, College of Medicine and Medical Research Institute, Chungbuk National University, 776 1sunhwan-ro, Seowon-gu, Cheongju 28644, Republic of Korea
- Center for Study of Emerging and Re-emerging Viruses, Korea Virus Research Institute, Institute for Basic Science (IBS), Daejeon 34126, Republic of Korea
| | - Sun Woo Yoon
- BioNanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Seungbeom Seo
- BioNanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Cogno-Mechatronics Engineering, Pusan National University, 2 Busandaehak-ro, Gumjeong-gu, Busan 46241, Republic of Korea
| | - Soojin Jang
- BioNanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Seong Uk Son
- BioNanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Taejoon Kang
- BioNanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Juyeon Jung
- BioNanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| | - Kyu-Sun Lee
- BioNanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Myung Hee Kim
- Metabolic Regulation Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Eun-Kyung Lim
- BioNanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
- Department of Nanobiotechnology, KRIBB School of Biotechnology, University of Science and Technology (UST), 217 Gajeong-ro, Yuseong-gu, Daejeon 34113, Republic of Korea
| |
Collapse
|
20
|
Sui Y, Li J, Andersen H, Zhang R, Prabhu SK, Hoang T, Venzon D, Cook A, Brown R, Teow E, Velasco J, Pessaint L, Moore IN, Lagenaur L, Talton J, Breed MW, Kramer J, Bock KW, Minai M, Nagata BM, Choo-Wosoba H, Lewis MG, Wang LX, Berzofsky JA. An intranasally administrated SARS-CoV-2 beta variant subunit booster vaccine prevents beta variant replication in rhesus macaques. PNAS NEXUS 2022; 1:pgac091. [PMID: 35873792 PMCID: PMC9295201 DOI: 10.1093/pnasnexus/pgac091] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/08/2022] [Indexed: 02/05/2023]
Abstract
Emergence of SARS-CoV-2 variants and waning of vaccine/infection-induced immunity pose threats to curbing the COVID-19 pandemic. Effective, safe, and convenient booster vaccines are in need. We hypothesized that a variant-modified mucosal booster vaccine might induce local immunity to prevent SARS-CoV-2 infection at the port of entry. The beta-variant is one of the hardest to cross-neutralize. Herein, we assessed the protective efficacy of an intranasal booster composed of beta variant-spike protein S1 with IL-15 and TLR agonists in previously immunized macaques. The macaques were first vaccinated with Wuhan strain S1 with the same adjuvant. A total of 1 year later, negligibly detectable SARS-CoV-2-specific antibody remained. Nevertheless, the booster induced vigorous humoral immunity including serum- and bronchoalveolar lavage (BAL)-IgG, secretory nasal- and BAL-IgA, and neutralizing antibody against the original strain and/or beta variant. Beta-variant S1-specific CD4+ and CD8+ T cell responses were also elicited in PBMC and BAL. Following SARS-CoV-2 beta variant challenge, the vaccinated group demonstrated significant protection against viral replication in the upper and lower respiratory tracts, with almost full protection in the nasal cavity. The fact that one intranasal beta-variant booster administrated 1 year after the first vaccination provoked protective immunity against beta variant infections may inform future SARS-CoV-2 booster design and administration timing.
Collapse
Affiliation(s)
- Yongjun Sui
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jianping Li
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | - Roushu Zhang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Sunaina K Prabhu
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Tanya Hoang
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - David Venzon
- Biostatistics and Data Management Section, Center of for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | | | | | | | | | - Ian N Moore
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Laurel Lagenaur
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jim Talton
- Alchem Laboratories, Alachua, FL 32615, USA
| | - Matthew W Breed
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Rockville, MD 20850, USA
| | - Josh Kramer
- Laboratory Animal Sciences Program, Frederick National Laboratory for Cancer Research, Rockville, MD 20850, USA
| | - Kevin W Bock
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Bianca M Nagata
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, Rockville, MD 20852, USA
| | - Hyoyoung Choo-Wosoba
- Biostatistics and Data Management Section, Center of for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | | | - Lai-Xi Wang
- Department of Chemistry and Biochemistry, University of Maryland, College Park, MD 20742, USA
| | - Jay A Berzofsky
- Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| |
Collapse
|
21
|
Shariq M, Sheikh JA, Quadir N, Sharma N, Hasnain SE, Ehtesham NZ. COVID-19 and tuberculosis: the double whammy of respiratory pathogens. Eur Respir Rev 2022; 31:31/164/210264. [PMID: 35418488 DOI: 10.1183/16000617.0264-2021] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 02/07/2022] [Indexed: 12/19/2022] Open
Abstract
Prior to coronavirus disease 2019 (COVID-19), tuberculosis (TB) was the worst killer among infectious diseases. The union of these two obnoxious respiratory diseases can be devastating, with severe public health implications. The COVID-19 pandemic has affected all TB-elimination programmes due to the severe burden on healthcare systems and the diversion of funds and attention towards controlling the pandemic. The emerging data show that the COVID-19 pandemic caused a marked decrease in case notifications and bacille Calmette-Guérin immunisations, ultimately promoting disease transmission and increasing the susceptible population. The similarity between the clinical characteristics of TB and COVID-19 adds to the public health complications, with evidence of immune dysregulation in both cases leading to severe consequences. Clinical evidence suggests that severe acute respiratory syndrome coronavirus 2 infection predisposes patients to TB infection or may lead to reactivation of latent disease. Similarly, underlying TB disease can worsen COVID-19. Treatment options are limited in COVID-19; therefore, using immunosuppressive and immunomodulatory regimens that can modulate the concomitant bacterial infection and interaction with anti-TB drugs requires caution. Thus, considering the synergistic impact of these two respiratory diseases, it is crucial to manage both diseases to combat the syndemic of TB and COVID-19.
Collapse
Affiliation(s)
- Mohd Shariq
- ICMR-National Institute of Pathology, New Delhi, India.,These authors contributed equally to this work
| | - Javaid A Sheikh
- Dept of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi, India.,These authors contributed equally to this work
| | - Neha Quadir
- ICMR-National Institute of Pathology, New Delhi, India.,Jamia Hamdard-Institute of Molecular Medicine, New Delhi, India
| | - Neha Sharma
- ICMR-National Institute of Pathology, New Delhi, India.,Jamia Hamdard-Institute of Molecular Medicine, New Delhi, India
| | - Seyed E Hasnain
- Dept of Biochemical Engineering and Biotechnology, Indian Institute of Technology, New Delhi, India.,Dept of Life Science, School of Basic Sciences and Research, Sharda University, Greater Noida, India.,These authors contributed equally to this article as lead authors and supervised the work
| | - Nasreen Z Ehtesham
- ICMR-National Institute of Pathology, New Delhi, India .,These authors contributed equally to this article as lead authors and supervised the work
| |
Collapse
|
22
|
Kim Y, Gaudreault NN, Meekins DA, Perera KD, Bold D, Trujillo JD, Morozov I, McDowell CD, Chang KO, Richt JA. Effects of Spike Mutations in SARS-CoV-2 Variants of Concern on Human or Animal ACE2-Mediated Virus Entry and Neutralization. Microbiol Spectr 2022; 10:e0178921. [PMID: 35638818 PMCID: PMC9241865 DOI: 10.1128/spectrum.01789-21] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 04/15/2022] [Indexed: 12/31/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a zoonotic agent capable of infecting humans and a wide range of animal species. Over the duration of the pandemic, mutations in the SARS-CoV-2 spike (S) protein have arisen, culminating in the spread of several variants of concern (VOCs) with various degrees of altered virulence, transmissibility, and neutralizing antibody escape. In this study, we used pseudoviruses that express specific SARS-CoV-2 S protein substitutions and cell lines that express angiotensin-converting enzyme 2 (ACE2) from nine different animal species to gain insights into the effects of VOC mutations on viral entry and antibody neutralization capability. All animal ACE2 receptors tested, except mink, support viral cell entry for pseudoviruses expressing the ancestral prototype S at levels comparable to human ACE2. Most single S substitutions did not significantly change virus entry, although 614G and 484K resulted in a decreased efficiency. Conversely, combinatorial VOC substitutions in the S protein were associated with increased entry of pseudoviruses. Neutralizing titers in sera from various animal species were significantly reduced against pseudoviruses expressing the S proteins of Beta, Delta, or Omicron VOCs compared to the parental S protein. Especially, substitutions in the S protein of the Omicron variant significantly reduced the neutralizing titers of the sera. This study reveals important insights into the host range of SARS-CoV-2 and the effect of recently emergent S protein substitutions on viral entry, virus replication, and antibody-mediated viral neutralization. IMPORTANCE The ongoing coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), continues to have devastating impacts on global health and socioeconomics. The recent emergence of SARS-CoV-2 variants of concern, which contain mutations that can affect the virulence, transmission, and effectiveness of licensed vaccines and therapeutic antibodies, are currently becoming the common strains circulating in humans worldwide. In addition, SARS-CoV-2 has been shown to infect a wide variety of animal species, which could result in additional mutations of the SARS-CoV-2 virus. In this study, we investigate the effect of mutations present in SARS-CoV-2 variants of concern and determine the effects of these mutations on cell entry, virulence, and antibody neutralization activity in humans and a variety of animals that might be susceptible to SARS-CoV-2 infection. This information is essential to understand the effects of important SARS-CoV-2 mutations and to inform public policy to create better strategies to control the COVID-19 pandemic.
Collapse
Affiliation(s)
- Yunjeong Kim
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Natasha N. Gaudreault
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - David A. Meekins
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Krishani D. Perera
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Dashzeveg Bold
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Jessie D. Trujillo
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Igor Morozov
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Chester D. McDowell
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Kyeong-Ok Chang
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| | - Juergen A. Richt
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, USA
| |
Collapse
|
23
|
Delobel D, Furutani Y, Nagoshi S, Tsubota A, Miyasaka A, Watashi K, Wakita T, Matsuura T, Usui K. SEB genotyping: SmartAmp-Eprimer binary code genotyping for complex, highly variable targets applied to HBV. BMC Infect Dis 2022; 22:516. [PMID: 35659601 PMCID: PMC9164387 DOI: 10.1186/s12879-022-07458-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/09/2022] [Indexed: 11/10/2022] Open
Abstract
Background SmartAmp-Eprimer Binary code (SEB) Genotyping is a novel isothermal amplification method for rapid genotyping of any variable target of interest. Methods After in silico alignment of a large number of sequences and computational analysis to determine the smallest number of regions to be targeted by SEB Genotyping, SmartAmp primer sets were designed to obtain a binary code of On/Off fluorescence signals, each code corresponding to a unique genotype. Results Applied to HBV, we selected 4 targets for which fluorescence amplification signals produce a specific binary code unique to each of the 8 main genotypes (A–H) found in patients worldwide. Conclusions We present here the proof of concept of a new genotyping method specifically designed for complex and highly variable targets. Applied here to HBV, SEB Genotyping can be adapted to any other pathogen or disease carrying multiple known mutations. Using simple preparation steps, SEB Genotyping provides accurate results quickly and will enable physicians to choose the best adapted treatment for each of their patients. Supplementary Information The online version contains supplementary material available at 10.1186/s12879-022-07458-4.
Collapse
|
24
|
Sun X, Wandelt S, Zhang A. Air transportation as a puzzle piece of COVID-19 in Africa? RESEARCH IN TRANSPORTATION BUSINESS & MANAGEMENT 2022; 43:100780. [PMID: 38620873 PMCID: PMC8743508 DOI: 10.1016/j.rtbm.2022.100780] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/01/2022] [Accepted: 01/04/2022] [Indexed: 01/04/2023]
Abstract
COVID-19 has hit our society hard, with more than 242 million cases reported worldwide and more than 4.9 million directly related fatalities. The role of Africa throughout the pandemic has been puzzling, since the African continent seems to have gone through the pandemic better than other continents; clearly better than predicted by the public during the emergence of COVID-19 one year ago. While several factors have been proposed in the literature to explain the unexpected role of Africa, including a relatively young population, more historical-driven preparedness to other types of coronavirus and diseases, and a limited amount of testing, the puzzle is not considered to be solved. In this study, we aim to answer the question whether air transportation indicators can support us in explaining the evolution of COVID-19 in Africa? Using flight data for the year 2020, we explore how changes in the air transportation system correlate with evolution of epidemiological indicators. Our results suggest that air transportation could indeed play a critical role for the spread of COVID-19 in Africa as well. Overall, we hope that our analysis contributes towards a better understanding of COVID-19 and the role air transportation plays in an under-researched region of the world.
Collapse
Affiliation(s)
- Xiaoqian Sun
- National Key Laboratory of CNS/ATM, School of Electronic and Information Engineering, Beihang University, 100191 Beijing, China
- National Engineering Laboratory of Multi-Modal Transportation Big Data, 100191 Beijing, China
| | - Sebastian Wandelt
- National Key Laboratory of CNS/ATM, School of Electronic and Information Engineering, Beihang University, 100191 Beijing, China
- National Engineering Laboratory of Multi-Modal Transportation Big Data, 100191 Beijing, China
| | - Anming Zhang
- Sauder School of Business, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
25
|
Agrawal S, Orschler L, Schubert S, Zachmann K, Heijnen L, Tavazzi S, Gawlik BM, de Graaf M, Medema G, Lackner S. Prevalence and circulation patterns of SARS-CoV-2 variants in European sewage mirror clinical data of 54 European cities. WATER RESEARCH 2022; 214:118162. [PMID: 35193077 PMCID: PMC8817224 DOI: 10.1016/j.watres.2022.118162] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/28/2022] [Accepted: 02/03/2022] [Indexed: 05/04/2023]
Abstract
For community-level monitoring, the European Commission under the EU Sewage Sentinel System recommends wastewater-based SARS-CoV-2 surveillance. Tracking SARS-CoV-2 variants in a community is pivotal for appropriate public health response. Genome sequencing of SARS-CoV-2 in wastewater samples for tracking variants is challenging, often resulting in low coverage genome sequences, thereby impeding the detection of the SARS-CoV-2 mutations. Therefore, we aimed at high-coverage SARS-CoV-2 genome sequences from sewage samples which we successfully accomplished. This first pan-European surveillance compared the mutation profiles associated with the variants of concerns: B.1.1.7, P.1, B.1.351 and B.1.617.2 across 20 European countries, including 54 municipalities. The results highlight that SARS-CoV-2 variants detected in the wastewater samples mirror the variants profiles reported in clinical data. This study demonstrated that >98% coverage of SARS-CoV-2 genomic sequences is possible and can be used to track SARS-CoV-2 mutations in wastewater to support identifying variants circulating in a city at the community level.
Collapse
Affiliation(s)
- Shelesh Agrawal
- Department of Civil and Environmental Engineering Sciences, Institute IWAR, Chair of Water and Environmental Biotechnology, Technical University of Darmstadt, Darmstadt, Germany.
| | - Laura Orschler
- Department of Civil and Environmental Engineering Sciences, Institute IWAR, Chair of Water and Environmental Biotechnology, Technical University of Darmstadt, Darmstadt, Germany
| | - Selina Schubert
- Department of Civil and Environmental Engineering Sciences, Institute IWAR, Chair of Water and Environmental Biotechnology, Technical University of Darmstadt, Darmstadt, Germany
| | - Kira Zachmann
- Department of Civil and Environmental Engineering Sciences, Institute IWAR, Chair of Water and Environmental Biotechnology, Technical University of Darmstadt, Darmstadt, Germany
| | - Leo Heijnen
- KWR Water Research Institute, Nieuwegein, the Netherland
| | - Simona Tavazzi
- European Commission, Joint Research Centre, Ispra, VA, Italy
| | | | - Miranda de Graaf
- Department of Viroscience, Erasmus Medical Center, Rotterdam, the Netherland
| | - Gertjan Medema
- KWR Water Research Institute, Nieuwegein, the Netherland
| | - Susanne Lackner
- Department of Civil and Environmental Engineering Sciences, Institute IWAR, Chair of Water and Environmental Biotechnology, Technical University of Darmstadt, Darmstadt, Germany
| |
Collapse
|
26
|
Panneer Selvam S, Ramani P, R R, Sundar S, T A L. COVID-19 Vaccines and the Efficacy of Currently Available Vaccines Against COVID-19 Variants. Cureus 2022; 14:e24927. [PMID: 35706739 PMCID: PMC9187843 DOI: 10.7759/cureus.24927] [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] [Accepted: 05/11/2022] [Indexed: 11/06/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 is the seventh member of the Coronaviridiae family of viruses, which are thought to be transmitted by Chinese horseshoe bats. The virus undergoes mutations leading to variants such as B.1.1.7 (alpha), B.1.351 (beta), P.1 (gamma), and B.1.617 (delta), as well as the recent variant B.1.1.529 (omicron), which has around 30 deletions, making it a severely mutated form that lowers vaccination-induced protection. Vaccine efficacy is usually expressed as relative risk reduction, which is based on the ratio of attack rates with and without a vaccine, whereas absolute risk reduction is based on the entire population. Rather than two doses, recent research suggests that a third dose/booster dose may aid in protection against future variants. The constant influx of mutant variations is putting a strain on vaccine production. Despite the challenges, we are optimistic that the epidemic will be eradicated by achieving mass immunity and by ensuring that everyone receives vaccines at a faster rate.
Collapse
Affiliation(s)
| | - Pratibha Ramani
- Oral Pathology, Saveetha Dental College & Hospital, Chennai, IND
| | - Ramya R
- Oral Biology, Saveetha Dental College & Hospital, Chennai, IND
| | - Sandhya Sundar
- Oral Pathology, Saveetha Dental College & Hospital, Chennai, IND
| | - Lakshmi T A
- Oral Pathology, Saveetha Dental College & Hospital, Chennai, IND
| |
Collapse
|
27
|
Jamir E, Sarma H, Priyadarsinee L, Nagamani S, Kiewhuo K, Gaur AS, Rawal RK, Murugan NA, Subramanian V, Sastry GN. Applying polypharmacology approach for drug repurposing for SARS-CoV2. J CHEM SCI 2022; 134:57. [PMID: 35498548 PMCID: PMC9028909 DOI: 10.1007/s12039-022-02046-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 02/06/2023]
Abstract
Exploring the new therapeutic indications of known drugs for treating COVID-19, popularly known as drug repurposing, is emerging as a pragmatic approach especially owing to the mounting pressure to control the pandemic. Targeting multiple targets with a single drug by employing drug repurposing known as the polypharmacology approach may be an optimised strategy for the development of effective therapeutics. In this study, virtual screening has been carried out on seven popular SARS-CoV-2 targets (3CLpro, PLpro, RdRp (NSP12), NSP13, NSP14, NSP15, and NSP16). A total of 4015 approved drugs were screened against these targets. Four drugs namely venetoclax, tirilazad, acetyldigitoxin, and ledipasvir have been selected based on the docking score, ability to interact with four or more targets and having a reasonably good number of interactions with key residues in the targets. The MD simulations and MM-PBSA studies showed reasonable stability of protein-drug complexes and sustainability of key interactions between the drugs with their respective targets throughout the course of MD simulations. The identified four drug molecules were also compared with the known drugs namely elbasvir and nafamostat. While the study has provided a detailed account of the chosen protein-drug complexes, it has explored the nature of seven important targets of SARS-CoV-2 by evaluating the protein-drug complexation process in great detail.
Collapse
Affiliation(s)
- Esther Jamir
- Advanced Computation and Data Sciences Division, CSIR – North East Institute of Science and Technology, Jorhat, Assam 785006 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Himakshi Sarma
- Advanced Computation and Data Sciences Division, CSIR – North East Institute of Science and Technology, Jorhat, Assam 785006 India
| | - Lipsa Priyadarsinee
- Advanced Computation and Data Sciences Division, CSIR – North East Institute of Science and Technology, Jorhat, Assam 785006 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Selvaraman Nagamani
- Advanced Computation and Data Sciences Division, CSIR – North East Institute of Science and Technology, Jorhat, Assam 785006 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Kikrusenuo Kiewhuo
- Advanced Computation and Data Sciences Division, CSIR – North East Institute of Science and Technology, Jorhat, Assam 785006 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Anamika Singh Gaur
- Advanced Computation and Data Sciences Division, CSIR – North East Institute of Science and Technology, Jorhat, Assam 785006 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ravindra K Rawal
- Advanced Computation and Data Sciences Division, CSIR – North East Institute of Science and Technology, Jorhat, Assam 785006 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Natarajan Arul Murugan
- Department of Computer Science, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology, 106 91 Stockholm, Sweden
| | - Venkatesan Subramanian
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
- Center for High Computing, CSIR- Central Leather Research Institute (CLRI), Chennai, India
| | - G Narahari Sastry
- Advanced Computation and Data Sciences Division, CSIR – North East Institute of Science and Technology, Jorhat, Assam 785006 India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
28
|
Vindeirinho JM, Pinho E, Azevedo NF, Almeida C. SARS-CoV-2 Diagnostics Based on Nucleic Acids Amplification: From Fundamental Concepts to Applications and Beyond. Front Cell Infect Microbiol 2022; 12:799678. [PMID: 35402302 PMCID: PMC8984495 DOI: 10.3389/fcimb.2022.799678] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 02/18/2022] [Indexed: 02/06/2023] Open
Abstract
COVID-19 pandemic ignited the development of countless molecular methods for the diagnosis of SARS-CoV-2 based either on nucleic acid, or protein analysis, with the first establishing as the most used for routine diagnosis. The methods trusted for day to day analysis of nucleic acids rely on amplification, in order to enable specific SARS-CoV-2 RNA detection. This review aims to compile the state-of-the-art in the field of nucleic acid amplification tests (NAATs) used for SARS-CoV-2 detection, either at the clinic level, or at the Point-Of-Care (POC), thus focusing on isothermal and non-isothermal amplification-based diagnostics, while looking carefully at the concerning virology aspects, steps and instruments a test can involve. Following a theme contextualization in introduction, topics about fundamental knowledge on underlying virology aspects, collection and processing of clinical samples pave the way for a detailed assessment of the amplification and detection technologies. In order to address such themes, nucleic acid amplification methods, the different types of molecular reactions used for DNA detection, as well as the instruments requested for executing such routes of analysis are discussed in the subsequent sections. The benchmark of paradigmatic commercial tests further contributes toward discussion, building on technical aspects addressed in the previous sections and other additional information supplied in that part. The last lines are reserved for looking ahead to the future of NAATs and its importance in tackling this pandemic and other identical upcoming challenges.
Collapse
Affiliation(s)
- João M. Vindeirinho
- National Institute for Agrarian and Veterinarian Research (INIAV, I.P), Vairão, Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, Porto, Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Porto, Portugal
| | - Eva Pinho
- National Institute for Agrarian and Veterinarian Research (INIAV, I.P), Vairão, Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, Porto, Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Porto, Portugal
| | - Nuno F. Azevedo
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, Porto, Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Porto, Portugal
| | - Carina Almeida
- National Institute for Agrarian and Veterinarian Research (INIAV, I.P), Vairão, Portugal
- Laboratory for Process Engineering, Environment, Biotechnology and Energy (LEPABE), Faculty of Engineering, University of Porto, Porto, Portugal
- Associate Laboratory in Chemical Engineering (ALiCE), Faculty of Engineering, University of Porto, Porto, Portugal
- Centre of Biological Engineering (CEB), University of Minho, Braga, Portugal
| |
Collapse
|
29
|
Fung KM, Lai SJ, Lin TL, Tseng TS. Antigen–Antibody Complex-Guided Exploration of the Hotspots Conferring the Immune-Escaping Ability of the SARS-CoV-2 RBD. Front Mol Biosci 2022; 9:797132. [PMID: 35392535 PMCID: PMC8981523 DOI: 10.3389/fmolb.2022.797132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 03/02/2022] [Indexed: 11/24/2022] Open
Abstract
The COVID-19 pandemic resulting from the spread of SARS-CoV-2 spurred devastating health and economic crises around the world. Neutralizing antibodies and licensed vaccines were developed to combat COVID-19, but progress was slow. In addition, variants of the receptor-binding domain (RBD) of the spike protein confer resistance of SARS-CoV-2 to neutralizing antibodies, nullifying the possibility of human immunity. Therefore, investigations into the RBD mutations that disrupt neutralization through convalescent antibodies are urgently required. In this study, we comprehensively and systematically investigated the binding stability of RBD variants targeting convalescent antibodies and revealed that the RBD residues F456, F490, L452, L455, and K417 are immune-escaping hotspots, and E484, F486, and N501 are destabilizing residues. Our study also explored the possible modes of actions of emerging SARS-CoV-2 variants. All results are consistent with experimental observations of attenuated antibody neutralization and clinically emerging SARS-CoV-2 variants. We identified possible immune-escaping hotspots that could further promote resistance to convalescent antibodies. The results provide valuable information for developing and designing novel monoclonal antibody drugs to combat emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Kit-Man Fung
- Academia Sinica, Institute of Biological Chemistry, Taipei, Taiwan
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Shu-Jung Lai
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Tzu-Lu Lin
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
| | - Tien-Sheng Tseng
- Institute of Molecular Biology, National Chung Hsing University, Taichung, Taiwan
- *Correspondence: Tien-Sheng Tseng,
| |
Collapse
|
30
|
Heraud JM, Lavergne A, Njouom R. Special Issue: “Viral Genetic Diversity”. Viruses 2022; 14:v14030570. [PMID: 35336977 PMCID: PMC8954497 DOI: 10.3390/v14030570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022] Open
Affiliation(s)
- Jean-Michel Heraud
- National Reference Laboratory for Rabies and Viral Encephalitis, Institut Pasteur de Dakar, Dakar 12900, Senegal
- Correspondence: ; Tel.: +221-770923244
| | - Anne Lavergne
- Laboratoire des Interactions Virus-Hôtes, Institut Pasteur de la Guyane, Cayenne 97306, French Guiana;
| | - Richard Njouom
- Virology Department, Centre Pasteur du Cameroun, Yaounde, Cameroon;
| |
Collapse
|
31
|
Putri ND, Johar E, Dewi YP, Indrasari ND, Wulandari D, br Pasaribu MM, Sari TT, Cakti FP, Jasin MR, Tartila T, Yudhaputri FA, Malik SG, Myint KSA. Whole-Genome Sequencing of SARS-CoV-2 Infection in a Cluster of Immunocompromised Children in Indonesia. Front Med (Lausanne) 2022; 9:835998. [PMID: 35308495 PMCID: PMC8930830 DOI: 10.3389/fmed.2022.835998] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 01/26/2022] [Indexed: 12/24/2022] Open
Abstract
Background Thus far, Indonesia has recorded over 4,000,000 confirmed COVID-19 cases and 144,000 fatalities; 12.8% of cases have been in children under 18 years. Whole-genome viral sequencing (WGS) of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been demonstrated to help differentiate hospital-acquired infection from community-acquired coronavirus disease 2019 (COVID-19) infection. Our study highlighted the use of WGS to investigate the origin of infection among pediatric oncology patients in Jakarta. The aim of our study was to evaluate clinical and laboratory characteristics and also the efficacy of using WGS to confirm hospital-acquired COVID-19 infection in a cluster of immunocompromised children within a single ward of a tertiary hospital in metropolitan Jakarta based on quasispecies, viral load, and admission dates. Method Real-time reverse-transcription polymerase chain reaction (RT-PCR) from nasopharyngeal (NP) swabs was used to diagnose the patients and also guardians and healthcare workers (HCWs) in the ward, followed by WGS of RT-PCR positive cases to establish their phylogenetic relationships. Result Using WGS, we showed that SARS-CoV-2 transmission in a cluster of children with underlying malignancy was characterized by high similarity of whole virus genome, which suggests nosocomial transmission.
Collapse
Affiliation(s)
- Nina Dwi Putri
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Edison Johar
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia
| | | | - Nuri Dyah Indrasari
- Department of Clinical Pathology, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Dewi Wulandari
- Department of Clinical Pathology, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Merci Monica br Pasaribu
- Department of Clinical Pathology, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Teny Tjitra Sari
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Fitri Prima Cakti
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Madeline Ramdhani Jasin
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Tartila Tartila
- Department of Paediatrics, Dr. Cipto Mangunkusumo National Central Hospital, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | | | | | - Khin Saw Aye Myint
- Eijkman Institute for Molecular Biology, Jakarta, Indonesia,*Correspondence: Khin Saw Aye Myint
| |
Collapse
|
32
|
Pérez-Cataluña A, Chiner-Oms Á, Cuevas-Ferrando E, Díaz-Reolid A, Falcó I, Randazzo W, Girón-Guzmán I, Allende A, Bracho MA, Comas I, Sánchez G. Spatial and temporal distribution of SARS-CoV-2 diversity circulating in wastewater. WATER RESEARCH 2022; 211:118007. [PMID: 35033744 PMCID: PMC8702378 DOI: 10.1016/j.watres.2021.118007] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 11/15/2021] [Accepted: 12/21/2021] [Indexed: 05/03/2023]
Abstract
Wastewater-based epidemiology (WBE) has proven to be an effective tool for epidemiological surveillance of SARS-CoV-2 during the current COVID-19 pandemic. Furthermore, combining WBE together with high-throughput sequencing techniques can be useful for the analysis of SARS-CoV-2 viral diversity present in a given sample. The present study focuses on the genomic analysis of SARS-CoV-2 in 76 sewage samples collected during the three epidemiological waves that occurred in Spain from 14 wastewater treatment plants distributed throughout the country. The results obtained demonstrate that the metagenomic analysis of SARS-CoV-2 in wastewater allows the detection of mutations that define the B.1.1.7 lineage and the ability of the technique to anticipate the detection of certain mutations before they are detected in clinical samples. The study proves the usefulness of sewage sequencing to track Variants of Concern that can complement clinical testing to help in decision-making and in the analysis of the evolution of the pandemic.
Collapse
Affiliation(s)
- Alba Pérez-Cataluña
- VISAFELab, Department of Preservation and Food Safety Technologies, Institute of Agrochemistry and Food Technology, IATA-CSIC, Av. Agustín Escardino 7, Paterna, Valencia 46980, Spain.
| | - Álvaro Chiner-Oms
- Instituto de Biomedicina de Valencia (IBV-CSIC), C/ Jaume Roig, 11, Valencia 46010, Spain
| | - Enric Cuevas-Ferrando
- VISAFELab, Department of Preservation and Food Safety Technologies, Institute of Agrochemistry and Food Technology, IATA-CSIC, Av. Agustín Escardino 7, Paterna, Valencia 46980, Spain
| | - Azahara Díaz-Reolid
- VISAFELab, Department of Preservation and Food Safety Technologies, Institute of Agrochemistry and Food Technology, IATA-CSIC, Av. Agustín Escardino 7, Paterna, Valencia 46980, Spain
| | - Irene Falcó
- VISAFELab, Department of Preservation and Food Safety Technologies, Institute of Agrochemistry and Food Technology, IATA-CSIC, Av. Agustín Escardino 7, Paterna, Valencia 46980, Spain
| | - Walter Randazzo
- VISAFELab, Department of Preservation and Food Safety Technologies, Institute of Agrochemistry and Food Technology, IATA-CSIC, Av. Agustín Escardino 7, Paterna, Valencia 46980, Spain
| | - Inés Girón-Guzmán
- VISAFELab, Department of Preservation and Food Safety Technologies, Institute of Agrochemistry and Food Technology, IATA-CSIC, Av. Agustín Escardino 7, Paterna, Valencia 46980, Spain
| | - Ana Allende
- Department of Food Science and Technology, CEBAS-CSIC, Research Group on Quality and Safety of Fruits and Vegetables, Campus Universitario de Espinardo, 25, Murcia 30100, Spain
| | - María A Bracho
- FISABIO - Public Health, Department of Genomics and Health, Av. Catalunya, 21, Valencia 46020, Spain; Joint Research Unit "Infection and Public Health" FISABIO-Universitat de Valencia I2SysBio, Av. Catalunya, 21, Valencia 46020, Spain; CIBER in Epidemiology and Public Health (CIBERESP), Valencia, Spain
| | - Iñaki Comas
- Instituto de Biomedicina de Valencia (IBV-CSIC), C/ Jaume Roig, 11, Valencia 46010, Spain; CIBER in Epidemiology and Public Health (CIBERESP), Valencia, Spain
| | - Gloria Sánchez
- VISAFELab, Department of Preservation and Food Safety Technologies, Institute of Agrochemistry and Food Technology, IATA-CSIC, Av. Agustín Escardino 7, Paterna, Valencia 46980, Spain
| |
Collapse
|
33
|
Asrani P, Tiwari K, Eapen MS, Hassan MI, Sohal SS. Containment strategies for COVID-19 in India: lessons from the second wave. Expert Rev Anti Infect Ther 2022; 20:829-835. [DOI: 10.1080/14787210.2022.2036605] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Purva Asrani
- Department of Microbiology, University of Delhi, South Campus, New Delhi, 110021, India
| | - Keshav Tiwari
- ICAR-National Institute for Plant Biotechnology, New Delhi, India-110012
| | - Mathew Suji Eapen
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, 7248, Australia
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi-110025, India
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, College of Health and Medicine, University of Tasmania, Launceston, Tasmania, 7248, Australia
| |
Collapse
|
34
|
Rahman S, Rahman MM, Miah M, Begum MN, Sarmin M, Mahfuz M, Hossain ME, Rahman MZ, Chisti MJ, Ahmed T, Arifeen SE, Rahman M. COVID-19 reinfections among naturally infected and vaccinated individuals. Sci Rep 2022; 12:1438. [PMID: 35082344 PMCID: PMC8792012 DOI: 10.1038/s41598-022-05325-5] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/03/2022] [Indexed: 12/17/2022] Open
Abstract
The protection against emerging SARS-CoV-2 variants by pre-existing antibodies elicited due to the current vaccination or natural infection is a global concern. We aimed to investigate the rate of SARS-CoV-2 infection and its clinical features among infection-naïve, infected, vaccinated, and post-infection-vaccinated individuals. A cohort was designed among icddr,b staff registered for COVID-19 testing by real-time reverse transcriptase-polymerase chain reaction (rRT-PCR). Reinfection cases were confirmed by whole-genome sequencing. From 19 March 2020 to 31 March 2021, 1644 (mean age, 38.4 years and 57% male) participants were enrolled; where 1080 (65.7%) were tested negative and added to the negative cohort. The positive cohort included 750 positive patients (564 from baseline and 186 from negative cohort follow-up), of whom 27.6% were hospitalized and 2.5% died. Among hospitalized patients, 45.9% had severe to critical disease and 42.5% required oxygen support. Hypertension and diabetes mellitus were found significantly higher among the hospitalised patients compared to out-patients; risk ratio 1.3 and 1.6 respectively. The risk of infection among positive cohort was 80.2% lower than negative cohort (95% CI 72.6-85.7%; p < 0.001). Genome sequences showed that genetically distinct SARS-CoV-2 strains were responsible for reinfections. Naturally infected populations were less likely to be reinfected by SARS-CoV-2 than the infection-naïve and vaccinated individuals. Although, reinfected individuals did not suffer severe disease, a remarkable proportion of naturally infected or vaccinated individuals were (re)-infected by the emerging variants.
Collapse
Affiliation(s)
- Sezanur Rahman
- Virology Laboratory, Infectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - M Mahfuzur Rahman
- Virology Laboratory, Infectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Mojnu Miah
- Virology Laboratory, Infectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Mst Noorjahan Begum
- Virology Laboratory, Infectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Monira Sarmin
- Nutrition and Clinical Services Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, 1212, Bangladesh
| | - Mustafa Mahfuz
- Nutrition and Clinical Services Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, 1212, Bangladesh
| | - Mohammad Enayet Hossain
- Virology Laboratory, Infectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Mohammed Ziaur Rahman
- Virology Laboratory, Infectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Mohammod Jobayer Chisti
- Nutrition and Clinical Services Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, 1212, Bangladesh
| | - Tahmeed Ahmed
- Nutrition and Clinical Services Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, 1212, Bangladesh
| | - Shams El Arifeen
- Maternal and Child Health Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, Mohakhali, Dhaka, 1212, Bangladesh
| | - Mustafizur Rahman
- Virology Laboratory, Infectious Diseases Division, icddr,b: International Centre for Diarrhoeal Disease Research, Bangladesh, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh.
| |
Collapse
|
35
|
Johnson R, Sharma JR, Ramharack P, Mangwana N, Kinnear C, Viraragavan A, Glanzmann B, Louw J, Abdelatif N, Reddy T, Surujlal-Naicker S, Nkambule S, Mahlangeni N, Webster C, Mdhluli M, Gray G, Mathee A, Preiser W, Muller C, Street R. Tracking the circulating SARS-CoV-2 variant of concern in South Africa using wastewater-based epidemiology. Sci Rep 2022; 12:1182. [PMID: 35064174 PMCID: PMC8783013 DOI: 10.1038/s41598-022-05110-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 12/31/2021] [Indexed: 12/13/2022] Open
Abstract
This study uses wastewater-based epidemiology (WBE) to rapidly and, through targeted surveillance, track the geographical distribution of SARS-CoV-2 variants of concern (Alpha, Beta and Delta) within 24 wastewater treatment plants (WWTPs) in the Western Cape of South Africa. Information obtained was used to identify the circulating variant of concern (VOC) within a population and retrospectively trace when the predominant variant was introduced. Genotyping analysis of SARS-CoV-2 showed that 50% of wastewater samples harbored signature mutations linked to the Beta variant before the third wave, with the Delta variant absent within the population. Over time, the prevalence of the beta variant decreased steadily. The onset of the third wave resulted in the Delta variant becoming the predominant variant, with a 100% prevalence supporting the theory that the Delta variant was driving the third wave. In silico molecular docking analysis showed that the signature mutations of the Delta variant increased binding to host proteins, suggesting a possible molecular mechanism that increased viral infectivity of the Delta variant.
Collapse
Affiliation(s)
- Rabia Johnson
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa.
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Centre for Cardiometabolic Research in Africa, Stellenbosch University, Stellenbosch, South Africa.
| | - Jyoti R Sharma
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa
| | - Pritika Ramharack
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa
- Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban, 4001, South Africa
| | - Noluxabiso Mangwana
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa
| | - Craig Kinnear
- Genomics Centre, South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Amsha Viraragavan
- Genomics Centre, South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa
| | - Brigitte Glanzmann
- Genomics Centre, South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Stellenbosch University, Cape Town, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa
| | - Nada Abdelatif
- Biostatistics Research Unit, South African Medical Research Council (SAMRC), Durban, South Africa
| | - Tarylee Reddy
- Biostatistics Research Unit, South African Medical Research Council (SAMRC), Durban, South Africa
| | - Swastika Surujlal-Naicker
- Scientific Services, Water and Sanitation Department, City of Cape Town Metropolitan Municipality, Cape Town, South Africa
| | - Sizwe Nkambule
- Environment and Health Research Unit, South African Medical Research Council (SAMRC), Durban, South Africa
| | - Nomfundo Mahlangeni
- Environment and Health Research Unit, South African Medical Research Council (SAMRC), Durban, South Africa
| | - Candice Webster
- Environment and Health Research Unit, South African Medical Research Council (SAMRC), Johannesburg, South Africa
| | - Mongezi Mdhluli
- Office of the President, South African Medical Research Council, Tygerberg, 7050, South Africa
| | - Glenda Gray
- Office of the President, South African Medical Research Council, Tygerberg, 7050, South Africa
| | - Angela Mathee
- Environment and Health Research Unit, South African Medical Research Council (SAMRC), Johannesburg, South Africa
| | - Wolfgang Preiser
- Division of Medical Virology at NHLS Tygerberg Hospital and Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Christo Muller
- Biomedical Research and Innovation Platform (BRIP), South African Medical Research Council (SAMRC), Tygerberg, 7505, South Africa
- Division of Medical Physiology, Faculty of Medicine and Health Sciences, Centre for Cardiometabolic Research in Africa, Stellenbosch University, Stellenbosch, South Africa
| | - Renee Street
- Environment and Health Research Unit, South African Medical Research Council (SAMRC), Durban, South Africa
| |
Collapse
|
36
|
Afrin SZ, Islam MT, Paul SK, Kobayashi N, Parvin R. Dynamics of SARS-CoV-2 variants of concern (VOC) in Bangladesh during the first half of 2021. Virology 2022; 565:29-37. [PMID: 34700068 PMCID: PMC8531988 DOI: 10.1016/j.virol.2021.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 10/02/2021] [Accepted: 10/18/2021] [Indexed: 01/04/2023]
Abstract
Bangladesh is the second-worst-affected country in South Asia by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The aim of this study is to examine genome sequences from Bangladesh from January 2021 to June 2021 in order to monitor the SARS-CoV-2 VOC and the clades or lineages that are prevalent in the country. Within the study timeframe, at least eight Nextstrain clades were found: 20A, 20B, 20C, 20H (Beta, V2), 20I (Alpha, V1), 20 J (Gamma, V3), 21A (Delta), 21D (Eta), and six GISAID clades: four main (G, GH, GR, GRY) and two minors (GV, O) with an introduction of VOC B.1.1.7/Alpha, B.1.351/Beta and B.1.617.2/Delta. The introduction and recent occurrence of VOCs with substantial alterations in the receptor binding site of spike protein (K417 N, K417T, L452R, T478K, E484K, S494P, N501Y) are of particular importance. Specifically, VOC B.1.617.2/Delta has surpassed all prior VOCs in Bangladesh, posing a challenge to the existing disease management.
Collapse
Affiliation(s)
| | - Md Taohidul Islam
- Population Medicine and AMR Laboratory, Department of Medicine, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Shyamal Kumar Paul
- Department of Microbiology, Netrokona Medical College, Netrokona, Bangladesh
| | - Nobumichi Kobayashi
- Department of Hygiene, Sapporo Medical University School of Medicine, Sapporo, Japan
| | - Rokshana Parvin
- Department of Pathology, Faculty of Veterinary Science, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh.
| |
Collapse
|
37
|
Hawkins SFC, Guest PC. Multiplex Quantitative Polymerase Chain Reaction Test to Identify SARS-CoV-2 Variants. Methods Mol Biol 2022; 2511:67-78. [PMID: 35838952 DOI: 10.1007/978-1-0716-2395-4_5] [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] [Indexed: 06/15/2023]
Abstract
Quantitative polymerase chain reaction (qPCR) is a routinely used method for detection and quantitation of gene expression in real time. This is achieved through the incorporation and measurement of fluorescent reporter probes in the amplified cDNA strands, since the fluorescent signals increase as the reaction progresses. The availability of multiple probes which fluoresce at different wavelengths allows for multiplexing as this gives rise to amplicons with unique fluorescent signatures. Here we describe a method using the Inhibitor-Tolerant RT-qPCR kit, developed by Meridian Bioscience kit which allows simultaneous real-time quantitation of the UK, South Africa, and Brazil SARS-CoV-2 variants.
Collapse
Affiliation(s)
| | - Paul C Guest
- Laboratory of Neuroproteomics, Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, Brazil.
| |
Collapse
|
38
|
Biby A, Wang X, Liu X, Roberson O, Henry A, Xia X. Rapid testing for coronavirus disease 2019 (COVID-19). MRS COMMUNICATIONS 2022; 12:12-23. [PMID: 35075405 PMCID: PMC8769796 DOI: 10.1557/s43579-021-00146-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Accepted: 09/29/2021] [Indexed: 05/03/2023]
Abstract
Rapid testing, generally refers to the paper-based diagnostic platform known as "lateral flow assay" (LFA), has emerged as a critical asset to the containment of coronavirus disease 2019 (COVID-19) around the world. LFA technology stands out amongst peer platforms due to its cost-effective design, user-friendly interface, and low sample-to-readout times. This article aims to introduce its design, use, and practicality for the purpose of diagnosing SARS-CoV-2 infection. A connection is made from the normal COVID-19 immune response to the design and efficacy of rapid testing. Interference in test results is a challenge shared by most diagnostic platforms and can be rooted in various underlying issues. The current knowledge and situation about interference in rapid COVID-19 tests due to variant strains as well as vaccination are discussed. The cost and societal impact are reviewed as they play important roles in determining how to properly implement public testing practices. Perspectives on improving the performance, especially detection sensitivity, of LFA for COVID-19 are provided.
Collapse
Affiliation(s)
- Alexander Biby
- Department of Chemistry, University of Central Florida, Orlando, FL 32816 USA
| | - Xiaochuan Wang
- School of Social Work, University of Central Florida, Orlando, FL 32816 USA
| | - Xinliang Liu
- School of Global Health Management & Informatics, University of Central Florida, Orlando, FL 32816 USA
| | - Olivia Roberson
- Department of Chemistry, University of Central Florida, Orlando, FL 32816 USA
| | - Allya Henry
- School of Social Work, University of Central Florida, Orlando, FL 32816 USA
| | - Xiaohu Xia
- Department of Chemistry, University of Central Florida, Orlando, FL 32816 USA
- NanoScience Technology Center, University of Central Florida, Orlando, FL 32816 USA
| |
Collapse
|
39
|
Verma M, Sharma S, Kumar A, Hakim A, Bhansali S, Meena R. Comorbidities and Vaccination Status of COVID-19 All-Cause Mortality at a Tertiary Care Center of Western India. Cureus 2022; 14:e21721. [PMID: 35251795 PMCID: PMC8886330 DOI: 10.7759/cureus.21721] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2022] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION COVID-19 vaccines have been found to be efficacious for preventing severe disease, yet breakthrough infections and deaths have occurred in a small proportion of vaccinated individuals. This study aimed to describe the vaccination status and comorbidities of COVID-19 all-cause deaths. METHODS This descriptive observational study was conducted at a tertiary care center in western India. A total of 310, RT-PCR positive COVID-19 deaths, aged 45 years and above irrespective of the cause of death (all-cause mortality), were included in the study. Death after breakthrough infection was defined as death in patient with disease onset after 14 days of the second dose of vaccine. RESULTS Diabetes was the most common comorbidity found in 17.1% of the deaths, followed by hypertension. Cardiovascular disease and renal disease were other common comorbidities seen in 8.7% and 4.83% deaths respectively. Other less common comorbidities include neurological disorders, HIV, autoimmune disorders. Out of these 310 deaths, 21.4% of patients developed disease within 14 days of the first dose. Death after true breakthrough infection (after 14 days of both doses) was seen in only two patients (0.6%). One of these two patients was aged 60 years and had diabetes, while the other was aged 72 years and had a history of smoking. CONCLUSION Diabetes and hypertension were the most common comorbidities, indicating a higher risk of mortality among comorbid patients. Only a small proportion of deaths (0.6%) occurred after breakthrough infection beyond 14 days of two doses. COVID-19 vaccines have shown promising efficacy against severe disease, thus high vaccination coverage needs to be achieved to prevent morbidity and mortality.
Collapse
Affiliation(s)
- Manoj Verma
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| | - Savitri Sharma
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| | - Arun Kumar
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| | - Afzal Hakim
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| | - Suman Bhansali
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| | - Rita Meena
- Community Medicine, Dr. Sampurnanand (S. N. Medical College), Jodhpur, IND
| |
Collapse
|
40
|
Review of Ribosome Interactions with SARS-CoV-2 and COVID-19 mRNA Vaccine. LIFE (BASEL, SWITZERLAND) 2022; 12:life12010057. [PMID: 35054450 PMCID: PMC8780073 DOI: 10.3390/life12010057] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 12/22/2021] [Accepted: 12/28/2021] [Indexed: 12/29/2022]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) is the causing pathogen of the unprecedented global Coronavirus Disease 19 (COVID-19) pandemic. Upon infection, the virus manipulates host cellular machinery and ribosomes to synthesize its own proteins for successful replication and to facilitate further infection. SARS-CoV-2 executes a multi-faceted hijacking of the host mRNA translation and cellular protein synthesis. Viral nonstructural proteins (NSPs) interact with a range of different ribosomal states and interfere with mRNA translation. Concurrent mutations on NSPs and spike proteins contribute to the epidemiological success of variants of concern (VOCs). The interactions between ribosomes and SARS-CoV-2 represent attractive targets for the development of antiviral therapeutics and vaccines. Recently approved COVID-19 mRNA vaccines also utilize the cellular machinery, to produce antigens and trigger immune responses. The design features of the mRNA vaccines are critical to efficient mRNA translation in ribosomes, and are directly related to the vaccine's efficacy, safety, and immunogenicity. This review describes recent knowledge of how the SARS-CoV-2 virus' genomic characteristics interfere with ribosomal function and mRNA translation. In addition, we discuss the current learning of the design features of mRNA vaccines and their impacts on translational activity in ribosomes. The understanding of ribosomal interactions with the virus and mRNA vaccines offers the foundation for antiviral therapeutic discovery and continuous mRNA vaccine optimization to lower the dose, to increase durability and/or to reduce adverse effects.
Collapse
|
41
|
Vaccine Breakthrough Infections by SARS-CoV-2 Variants after ChAdOx1 nCoV-19 Vaccination in Healthcare Workers. Vaccines (Basel) 2021; 10:vaccines10010054. [PMID: 35062715 PMCID: PMC8778656 DOI: 10.3390/vaccines10010054] [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: 12/08/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/17/2022] Open
Abstract
This study elucidated the clinical, humoral immune response and genomic analysis of vaccine breakthrough (VBT) infections after ChAdOx1 nCoV-19/Covishield vaccine in healthcare workers (HCWs). Amongst 1858 HCWs, 1639 had received either two doses (1346) or a single dose (293) of ChAdOx1 nCoV-19 vaccine. SARS-CoV-2 IgG antibodies and neutralizing antibodies were measured in the vaccinated group and the development of SARS-CoV-2 infection was monitored.Forty-six RT-PCR positive samples from the 203 positive samples were subjected to whole genome sequencing (WGS). Of the 203 (10.92%) infected HCWs, 21.46% (47/219) were non-vaccinated, which was significantly more than 9.52% (156/1639) who were vaccinated and infection was higher in doctors and nurses. Unvaccinated HCWs had 1.57 times higher risk compared to partially vaccinated HCWs and 2.49 times higher risk than those who were fully vaccinated.The partially vaccinated were at higher risk than the fully vaccinated (RR 1.58). Antibody non-response was seen in 3.44% (4/116), low antibody levels in 15.51% (18/116) and medium levels were found in 81.03% (94/116). Fully vaccinated HCWs had a higher antibody response at day 42 than those who were partially vaccinated (8.96 + 4.00 vs. 7.17 + 3.82). Whole genome sequencing of 46 samples revealed that the Delta variant (B.1.617.2) was predominant (69.5%). HCWs who had received two doses of vaccine showed better protection from mild, moderate, or severe infection, with a higher humoral immune response than those who had received a single dose. The genomic analysis revealed the predominance of the Delta variant (B.1.617.2) in the VBT infections.
Collapse
|
42
|
Comparison of the Performance of the PanBio COVID-19 Antigen Test in SARS-CoV-2 B.1.1.7 (Alpha) Variants versus non-B.1.1.7 Variants. Microbiol Spectr 2021; 9:e0088421. [PMID: 34817226 PMCID: PMC8612141 DOI: 10.1128/spectrum.00884-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
This study evaluates the performance of the PanBio COVID-19 antigen (Ag) test as part of a hospital infection control policy. Hospital staff was encouraged to get tested for COVID-19 when presenting with SARS-CoV-2-related symptoms. In a period of approximately 5 months, a steady decline in the performance of the Ag test was noted, epidemiologically coinciding with the rise of the SARS-CoV-2 B.1.1.7 (alpha) variant of concern (VOC) in the Netherlands. This led to the hypothesis that the diagnostic performance of the PanBio COVID-19 Ag test was influenced by the infecting viral variant. The results show a significantly lower sensitivity of the PanBio COVID-19 Ag test in persons infected with the B.1.1.7 (alpha) variant of SARS-CoV-2 in comparison with that in persons infected with non-B.1.1.7 variants, also after adjustment for viral load. IMPORTANCE Antigen tests for COVID-19 are widely used for rapid identification of COVID-19 cases, for example, for access to schools, festivals, and travel. There are several FDA- and CE-cleared tests on the market. Their performance has been evaluated mainly on the basis of infections by the classical variant of the causing virus, SARS-CoV-2. This paper provides evidence that the performance of one of the most widely used antigen tests detects significantly fewer cases of COVID-19 by the alpha variant than by the classical variants of SARS-CoV-2. This means that the role of antigen tests needs to be reevaluated in regions where other variants of SARS-CoV-2 predominate.
Collapse
|
43
|
Wagner GE, Totaro MG, Volland A, Lipp M, Saiger S, Lichtenegger S, Forstner P, von Laer D, Oberdorfer G, Steinmetz I. A Novel High-Throughput Nanopore-Sequencing-Based Strategy for Rapid and Automated S-Protein Typing of SARS-CoV-2 Variants. Viruses 2021; 13:2548. [PMID: 34960817 PMCID: PMC8704619 DOI: 10.3390/v13122548] [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: 11/29/2021] [Revised: 12/15/2021] [Accepted: 12/16/2021] [Indexed: 11/24/2022] Open
Abstract
Rapid molecular surveillance of SARS-CoV-2 S-protein variants leading to immune escape and/or increased infectivity is of utmost importance. Among global bottlenecks for variant monitoring in diagnostic settings are sequencing and bioinformatics capacities. In this study, we aimed to establish a rapid and user-friendly protocol for high-throughput S-gene sequencing and subsequent automated identification of variants. We designed two new primer pairs to amplify only the immunodominant part of the S-gene for nanopore sequencing. Furthermore, we developed an automated "S-Protein-Typer" tool that analyzes and reports S-protein mutations on the amino acid level including a variant of concern indicator. Validation of our primer panel using SARS-CoV-2-positive respiratory specimens covering a broad Ct range showed successful amplification for 29/30 samples. Restriction to the region of interest freed sequencing capacity by a factor of 12-13, compared with whole-genome sequencing. Using either the MinION or Flongle flow cell, our sequencing strategy reduced the time required to identify SARS-CoV-2 variants accordingly. The S-Protein-Typer tool identified all mutations correctly when challenged with our sequenced samples and 50 deposited sequences covering all VOCs (December 2021). Our proposed S-protein variant screening offers a simple, more rapid, and low-cost entry into NGS-based SARS-CoV-2 analysis, compared with current whole-genome approaches.
Collapse
Affiliation(s)
- Gabriel E. Wagner
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, 8010 Graz, Austria; (M.L.); (S.S.); (S.L.); (P.F.)
| | - Massimo G. Totaro
- Department of Biochemistry, Graz University of Technology, 8010 Graz, Austria; (M.G.T.); (G.O.)
| | - André Volland
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (A.V.); (D.v.L.)
| | - Michaela Lipp
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, 8010 Graz, Austria; (M.L.); (S.S.); (S.L.); (P.F.)
| | - Sabine Saiger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, 8010 Graz, Austria; (M.L.); (S.S.); (S.L.); (P.F.)
| | - Sabine Lichtenegger
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, 8010 Graz, Austria; (M.L.); (S.S.); (S.L.); (P.F.)
| | - Patrick Forstner
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, 8010 Graz, Austria; (M.L.); (S.S.); (S.L.); (P.F.)
| | - Dorothee von Laer
- Department of Hygiene, Microbiology and Public Health, Institute of Virology, Medical University of Innsbruck, 6020 Innsbruck, Austria; (A.V.); (D.v.L.)
| | - Gustav Oberdorfer
- Department of Biochemistry, Graz University of Technology, 8010 Graz, Austria; (M.G.T.); (G.O.)
| | - Ivo Steinmetz
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, 8010 Graz, Austria; (M.L.); (S.S.); (S.L.); (P.F.)
| |
Collapse
|
44
|
Neagu M, Constantin C, Surcel M. Testing Antigens, Antibodies, and Immune Cells in COVID-19 as a Public Health Topic—Experience and Outlines. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182413173. [PMID: 34948782 PMCID: PMC8700871 DOI: 10.3390/ijerph182413173] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/14/2022]
Abstract
The current COVID-19 pandemic has triggered an accelerated pace in all research domains, including reliable diagnostics methodology. Molecular diagnostics of the virus and its presence in biological samples relies on the RT-PCR method, the most used and validated worldwide. Nonconventional tests with improved parameters that are in the development stages will be presented, such as droplet digital PCR or CRISPR-based assays. These molecular tests were followed by rapid antigen testing along with the development of antibody tests, whether based on ELISA platform or on a chemiluminescent microparticle immunoassay. Less-conventional methods of testing antibodies (e.g., lateral flow immunoassay) are presented as well. Left somewhere in the backstage of COVID-19 research, immune cells and, furthermore, immune memory cells, are gaining the spotlight, more so in the vaccination context. Recently, methodologies using flow-cytometry evaluate circulating immune cells in infected/recovered patients. The appearance of new virus variants has triggered a surge for tests improvement. As the pandemic has entered an ongoing or postvaccination era, all methodologies that are used to monitor public health focus on diagnostic strategies and this review points out where gaps should be filled in both clinical and research settings.
Collapse
Affiliation(s)
- Monica Neagu
- Immunology Laboratory, Victor Babes National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (M.N.); (M.S.)
- Pathology Department, Colentina University Hospital, 19-21 Șoseaua Ștefan cel Mare, 020125 Bucharest, Romania
- Doctoral School of Biology, Faculty of Biology, University of Bucharest, 91-93 Splaiul Independentei, 050095 Bucharest, Romania
| | - Carolina Constantin
- Immunology Laboratory, Victor Babes National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (M.N.); (M.S.)
- Pathology Department, Colentina University Hospital, 19-21 Șoseaua Ștefan cel Mare, 020125 Bucharest, Romania
- Correspondence:
| | - Mihaela Surcel
- Immunology Laboratory, Victor Babes National Institute of Pathology, 99-101 Splaiul Independentei, 050096 Bucharest, Romania; (M.N.); (M.S.)
| |
Collapse
|
45
|
Maffei M, Montemiglio LC, Vitagliano G, Fedele L, Sellathurai S, Bucci F, Compagnone M, Chiarini V, Exertier C, Muzi A, Roscilli G, Vallone B, Marra E. The Nuts and Bolts of SARS-CoV-2 Spike Receptor-Binding Domain Heterologous Expression. Biomolecules 2021; 11:1812. [PMID: 34944456 PMCID: PMC8699011 DOI: 10.3390/biom11121812] [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: 10/28/2021] [Revised: 11/27/2021] [Accepted: 11/30/2021] [Indexed: 12/23/2022] Open
Abstract
COVID-19 is a highly infectious disease caused by a newly emerged coronavirus (SARS-CoV-2) that has rapidly progressed into a pandemic. This unprecedent emergency has stressed the significance of developing effective therapeutics to fight the current and future outbreaks. The receptor-binding domain (RBD) of the SARS-CoV-2 surface Spike protein is the main target for vaccines and represents a helpful "tool" to produce neutralizing antibodies or diagnostic kits. In this work, we provide a detailed characterization of the native RBD produced in three major model systems: Escherichia coli, insect and HEK-293 cells. Circular dichroism, gel filtration chromatography and thermal denaturation experiments indicated that recombinant SARS-CoV-2 RBD proteins are stable and correctly folded. In addition, their functionality and receptor-binding ability were further evaluated through ELISA, flow cytometry assays and bio-layer interferometry.
Collapse
Affiliation(s)
- Mariano Maffei
- Evvivax Biotech, Via di Castel Romano 100, 00128 Rome, Italy;
| | - Linda Celeste Montemiglio
- Institute of Molecular Biology and Pathology (IBPM), National Research Council, c/o Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Sapienza, University of Rome, P. le Aldo Moro, 5, 00185 Rome, Italy;
| | - Grazia Vitagliano
- Takis Biotech, Via di Castel Romano 100, 00128 Rome, Italy; (G.V.); (L.F.); (S.S.); (F.B.); (V.C.); (A.M.)
| | - Luigi Fedele
- Takis Biotech, Via di Castel Romano 100, 00128 Rome, Italy; (G.V.); (L.F.); (S.S.); (F.B.); (V.C.); (A.M.)
| | - Shaila Sellathurai
- Takis Biotech, Via di Castel Romano 100, 00128 Rome, Italy; (G.V.); (L.F.); (S.S.); (F.B.); (V.C.); (A.M.)
| | - Federica Bucci
- Takis Biotech, Via di Castel Romano 100, 00128 Rome, Italy; (G.V.); (L.F.); (S.S.); (F.B.); (V.C.); (A.M.)
| | | | - Valerio Chiarini
- Takis Biotech, Via di Castel Romano 100, 00128 Rome, Italy; (G.V.); (L.F.); (S.S.); (F.B.); (V.C.); (A.M.)
| | - Cécile Exertier
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Sapienza, University of Rome, P. le Aldo Moro, 5, 00185 Rome, Italy; (C.E.); (B.V.)
| | - Alessia Muzi
- Takis Biotech, Via di Castel Romano 100, 00128 Rome, Italy; (G.V.); (L.F.); (S.S.); (F.B.); (V.C.); (A.M.)
| | - Giuseppe Roscilli
- Evvivax Biotech, Via di Castel Romano 100, 00128 Rome, Italy;
- Takis Biotech, Via di Castel Romano 100, 00128 Rome, Italy; (G.V.); (L.F.); (S.S.); (F.B.); (V.C.); (A.M.)
| | - Beatrice Vallone
- Department of Biochemical Sciences “Alessandro Rossi Fanelli”, Sapienza, University of Rome, P. le Aldo Moro, 5, 00185 Rome, Italy; (C.E.); (B.V.)
| | - Emanuele Marra
- Evvivax Biotech, Via di Castel Romano 100, 00128 Rome, Italy;
- Takis Biotech, Via di Castel Romano 100, 00128 Rome, Italy; (G.V.); (L.F.); (S.S.); (F.B.); (V.C.); (A.M.)
| |
Collapse
|
46
|
Paniri A, Hosseini MM, Akhavan-Niaki H. Impact of new UK (B.1.1.7) SARS-Cov-2 variant on interacting with ACE2 and host immune response. GENE REPORTS 2021; 25:101342. [PMID: 34493993 PMCID: PMC8414842 DOI: 10.1016/j.genrep.2021.101342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 07/28/2021] [Accepted: 08/31/2021] [Indexed: 11/25/2022]
Affiliation(s)
- Alireza Paniri
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
- Genetics Department, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | | | - Haleh Akhavan-Niaki
- Genetics Department, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
- Zoonoses Research Center, Pasteur Institute of Iran, Amol, Iran
| |
Collapse
|
47
|
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: 11.7] [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
|
48
|
Role of multiple factors likely contributing to severity-mortality of COVID-19. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2021; 96:105101. [PMID: 34624542 PMCID: PMC8491954 DOI: 10.1016/j.meegid.2021.105101] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 02/08/2023]
Abstract
COVID-19 stalled the world in 2020 and continues to be the greatest health crisis of this generation. While the apparent case fatality rates across fluctuates around ~2% globally, associated mortality/death rate (deaths per million population) varies distinctly across regions from the global average of ~600 per million population. Heterogeneous factors have been linked with COVID-19 associated mortalities and these include age, share of geriatric population, comorbidities, trained immunity and climatic conditions. Apart from direct or indirect role of endemic diseases, dietary factors and host immunity in regulating COVID-19 severity, human behaviour will inevitably control outcome of this pandemic. Comprehensive understanding of these factors will have a bearing on management of future health crises.
Collapse
|
49
|
Bhattacharya M, Sharma AR, Ghosh P, Lee SS, Chakraborty C. A Next-Generation Vaccine Candidate Using Alternative Epitopes to Protect against Wuhan and All Significant Mutant Variants of SARS-CoV-2: An Immunoinformatics Approach. Aging Dis 2021; 12:2173-2195. [PMID: 34881093 PMCID: PMC8612605 DOI: 10.14336/ad.2021.0518] [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: 05/04/2021] [Accepted: 05/17/2021] [Indexed: 12/24/2022] Open
Abstract
Newly emerging significant SARS-CoV-2 variants such as B.1.1.7, B.1.351, and B.1.1.28 are the variant of concern (VOC) for the human race. These variants are getting challenging to contain from spreading worldwide. Because of these variants, the second wave has started in various countries and is threatening human civilization. Thus, we require efficient vaccines that can combat all emerging variants of SARS-CoV-2. Therefore, we took the initiative to develop a peptide-based next-generation vaccine using four variants (Wuhan variant, B.1.1.7, B.1.351, and B.1.1.28) that could potentially combat SARS-CoV-2 variants. We applied a series of computational tools, servers, and software to identify the most significant epitopes present on the mutagenic regions of SARS-CoV-2 variants. The immunoinformatics approaches were used to identify common B cell derived T cell epitopes, influencing the host immune system. Consequently, to develop a novel vaccine candidate, the antigenic epitopes were linked with a flexible and stable peptide linker, and the adjuvant was added at the N-terminal end. 3D vaccine candidate structure was refined, and quality was assessed using web servers. The physicochemical properties and safety parameters of the vaccine construct were assessed through bioinformatics and immunoinformatics tools. The molecular docking analysis between TLR4/MD2 and the proposed vaccine candidate demonstrated a satisfactory interaction. The molecular dynamics studies confirmed the stability of the vaccine candidate. Finally, we optimized the proposed vaccine through codon optimization and in silico cloning to study the expression. Our multi-epitopic next-generation peptide vaccine construct can boost immunity against the Wuhan variant and all significant mutant variants of SARS-CoV-2.
Collapse
Affiliation(s)
- Manojit Bhattacharya
- Department of Zoology, Fakir Mohan University, Vyasa Vihar, Balasore-756020, Odisha, India.
| | - Ashish Ranjan Sharma
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea.
| | - Pratik Ghosh
- Department of Zoology, Vidyasagar University, Midnapore, West Bengal 721102, India.
| | - Sang-Soo Lee
- Institute for Skeletal Aging & Orthopedic Surgery, Hallym University-Chuncheon Sacred Heart Hospital, Chuncheon-si, 24252, Gangwon-do, Republic of Korea.
| | - Chiranjib Chakraborty
- Department of Biotechnology, School of Life Science and Biotechnology, Adamas University, Barasat-Barrackpore Rd, Kolkata, West Bengal 700126, India.
| |
Collapse
|
50
|
Wang L, Li G, Yuan C, Yang Y, Ling G, Zheng J, Zhou Y, Zhang T, Lin W, Lin Z. Progress in the Diagnosis and Treatment of COVID-19 in Children: A Review. Int J Gen Med 2021; 14:8097-8108. [PMID: 34795516 PMCID: PMC8594783 DOI: 10.2147/ijgm.s335888] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Accepted: 10/27/2021] [Indexed: 12/14/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) has been circulating in many countries around the world, characterized by long incubation period, strong infectivity, strong variability, high population susceptibility and diversified transmission methods. Its causative agent is severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Compared with adult patients, the clinical manifestations of COVID-19 in children are often dominated by mild or asymptomatic infections, but children are also important virus carriers and play an important role in the transmission of the virus. In addition, some children will show excessive inflammatory response and experience serious complications such as multisystem inflammatory syndrome in children (MIS-C). At present, the research on COVID-19 in children is still imperfect. This article will review epidemiological characteristics, the mechanism of action, variant characteristics, clinical manifestations, auxiliary examinations and treatment of children with COVID-19, in order to provide help for the diagnosis, treatment and research of children with COVID-19.
Collapse
Affiliation(s)
- Libo Wang
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Gan Li
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Chang Yuan
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Yuele Yang
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Gongxia Ling
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Jinyu Zheng
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Yiyang Zhou
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Tianlei Zhang
- Department of Clinical Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Wei Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| | - Zhenlang Lin
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, People's Republic of China
| |
Collapse
|