1
|
Thimmiraju SR, Villar MJ, Kimata JT, Strych U, Bottazzi ME, Hotez PJ, Pollet J. Optimization of Cellular Transduction by the HIV-Based Pseudovirus Platform with Pan-Coronavirus Spike Proteins. Viruses 2024; 16:1492. [PMID: 39339968 PMCID: PMC11437443 DOI: 10.3390/v16091492] [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: 08/07/2024] [Revised: 09/05/2024] [Accepted: 09/17/2024] [Indexed: 09/30/2024] Open
Abstract
Over the past three years, new SARS-CoV-2 variants have continuously emerged, evolving to a point where an immune response against the original vaccine no longer provided optimal protection against these new strains. During this time, high-throughput neutralization assays based on pseudoviruses have become a valuable tool for assessing the efficacy of new vaccines, screening updated vaccine candidates against emerging variants, and testing the efficacy of new therapeutics such as monoclonal antibodies. Lentiviral vectors derived from HIV-1 are popular for developing pseudo and chimeric viruses due to their ease of use, stability, and long-term transgene expression. However, the HIV-based platform has lower transduction rates for pseudotyping coronavirus spike proteins than other pseudovirus platforms, necessitating more optimized methods. As the SARS-CoV-2 virus evolved, we produced over 18 variants of the spike protein for pseudotyping with an HIV-based vector, optimizing experimental parameters for their production and transduction. In this article, we present key parameters that were assessed to improve such technology, including (a) the timing and method of collection of pseudovirus supernatant; (b) the timing of host cell transduction; (c) cell culture media replenishment after pseudovirus adsorption; and (d) the centrifugation (spinoculation) parameters of the host cell+ pseudovirus mix, towards improved transduction. Additionally, we found that, for some pseudoviruses, the addition of a cationic polymer (polybrene) to the culture medium improved the transduction process. These findings were applicable across variant spike pseudoviruses that include not only SARS-CoV-2 variants, but also SARS, MERS, Alpha Coronavirus (NL-63), and bat-like coronaviruses. In summary, we present improvements in transduction efficiency, which can broaden the dynamic range of the pseudovirus titration and neutralization assays.
Collapse
Affiliation(s)
- Syamala Rani Thimmiraju
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maria Jose Villar
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Jason T. Kimata
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Ulrich Strych
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Maria Elena Bottazzi
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Peter J. Hotez
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jeroen Pollet
- Texas Children’s Hospital Center for Vaccine Development, Houston, TX 77030, USA; (S.R.T.); (M.J.V.); (U.S.); (M.E.B.); (P.J.H.)
- Department of Pediatrics, National School of Tropical Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA;
| |
Collapse
|
2
|
Dhankher S, Yadav P, Sharma S, Gupta E, Yadav RG, Dash PK, Parida M. Structural and genomic evolutionary dynamics of Omicron variant of SARS-CoV-2 circulating in Madhya Pradesh, India. Front Med (Lausanne) 2024; 11:1416006. [PMID: 39323472 PMCID: PMC11422100 DOI: 10.3389/fmed.2024.1416006] [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: 04/11/2024] [Accepted: 08/06/2024] [Indexed: 09/27/2024] Open
Abstract
The SARS-CoV-2 Omicron (B.1.1.529) variant emerged in early November 2021 and its rapid spread created fear worldwide. This was attributed to its increased infectivity and escaping immune mechanisms. The spike protein of Omicron has more mutations (>30) than any other previous variants and was declared as the variant of concern (VOC) by the WHO. The concern among the scientific community was huge about this variant, and a piece of updated information on circulating viral strains is important in order to better understand the epidemiology, virus pathogenicity, transmission, therapeutic interventions, and vaccine development. A total of 710 samples were processed for sequencing and identification up to a resolution of sub-lineage. The sequence analysis revealed Omicron variant with distribution as follows: B.1.1, B.1.1.529, BA.1, BA.2, BA.2.10, BA.2.10.1, BA.2.23, BA.2.37, BA.2.38, BA.2.43, BA.2.74, BA.2.75, BA.2.76, and BA.4 sub-lineages. There is a shift noted in circulating lineage from BA.1 to BA.2 to BA.4 over a period from January to September 2022. Multiple signature mutations were identified in S protein T376A, D405N, and R408S mutations, which were new and common to all BA.2 variants. Additionally, R346T was seen in emerging BA.2.74 and BA.2.76 variants. The emerging BA.4 retained the common T376A, D405N, and R408S mutations of BA.2 along with a new mutation F486V. The samples sequenced were from different districts of Madhya Pradesh and showed a predominance of BA.2 and its variants circulating in this region. The current study identified circulation of BA.1 and BA.1.1 variants during initial phase. The predominant Delta strain of the second wave has been replaced by the Omicron variant in this region over a period of time. This study successfully deciphers the dynamics of the emergence and replacement of various sub-lineages of SARS-CoV-2 in central India on real real-time basis.
Collapse
Affiliation(s)
| | | | | | | | | | - Paban Kumar Dash
- Virology Division, Defence Research and Development Establishment, Gwalior, India
| | | |
Collapse
|
3
|
Nugent JR, Wood MS, Liu L, Bullick T, Schapiro JM, Arunleung P, Gautham G, Getabecha S, Morales C, Amsden LB, Hsiao CA, Wadford DA, Wyman SK, Skarbinski J. SARS-CoV-2 Omicron subvariant genomic variation associations with immune evasion in Northern California: A retrospective cohort study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.08.21.24312253. [PMID: 39228703 PMCID: PMC11370498 DOI: 10.1101/2024.08.21.24312253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Background The possibility of association between SARS-CoV-2 genomic variation and immune evasion is not known among persons with Omicron variant SARS-CoV-2 infection. Methods In a retrospective cohort, using Poisson regression adjusting for sociodemographic variables and month of infection, we examined associations between individual non-lineage defining mutations and SARS-CoV-2 immunity status, defined as a) no prior recorded infection, b) not vaccinated but with at least one prior recorded infection, c) complete primary series vaccination, and/or d) primary series vaccination and ≥ 1 booster. We identified all non-synonymous single nucleotide polymorphisms (SNPs), insertions and deletions in SARS-CoV-2 genomes with ≥5% allelic frequency and population frequency of ≥5% and ≤95%. We also examined correlations between the presence of SNPs with each other, with subvariants, and over time. Results Seventy-nine mutations met inclusion criteria. Among 15,566 persons infected with Omicron SARS-CoV-2, 1,825 (12%) were unvaccinated with no prior recorded infection, 360 (2%) were unvaccinated with a recorded prior infection, 13,381 (86%) had a complete primary series vaccination, and 9,172 (58%) had at least one booster. After examining correlation between SNPs, 79 individual non-lineage defining mutations were organized into 38 groups. After correction for multiple testing, no individual SNPs or SNP groups were significantly associated with immunity status levels. Conclusions Genomic variation identified within SARS-CoV-2 Omicron specimens was not significantly associated with immunity status, suggesting that contribution of non-lineage defining SNPs to immune evasion is minimal. Larger-scale surveillance of SARS-CoV-2 genomes linked with clinical data can help provide information to inform future vaccine development.
Collapse
|
4
|
Merrett JE, Nolan M, Hartman L, John N, Flynn B, Baker L, Schang C, McCarthy D, Lister D, Cheng NN, Crosbie N, Poon R, Jex A. Highly sensitive wastewater surveillance of SARS-CoV-2 variants by targeted next-generation amplicon sequencing provides early warning of incursion in Victoria, Australia. Appl Environ Microbiol 2024; 90:e0149723. [PMID: 39012098 PMCID: PMC11337797 DOI: 10.1128/aem.01497-23] [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: 08/29/2023] [Accepted: 06/06/2024] [Indexed: 07/17/2024] Open
Abstract
The future of the COVID pandemic and its public health and societal impact will be determined by the profile and spread of emerging variants and the timely identification and response to them. Wastewater surveillance of SARS-CoV-2 has been widely adopted in many countries across the globe and has played an important role in tracking infection levels and providing useful epidemiological information that cannot be adequately captured by clinical testing alone. However, novel variants can emerge rapidly, spread globally, and markedly alter the trajectory of the pandemic, as exemplified by the Delta and Omicron variants. Most mutations linked to the emergence of new SARS-CoV-2 variants are found within variable regions of the SARS-CoV-2 Spike protein. We have developed a duplex hemi-nested PCR method that, coupled with short amplicon sequencing, allows simultaneous typing of two of the most highly variable and informative regions of the Spike gene: the N-terminal domain and the receptor binding motif. Using this method in an operationalized public health program, we identified the first known incursion of Omicron BA.1 into Victoria, Australia and demonstrated how sensitive amplicon sequencing methods can be combined with wastewater surveillance as a relatively low-cost solution for early warning of variant incursion and spread.IMPORTANCEThis study offers a rapid, cost-effective, and sensitive approach for monitoring SARS-CoV-2 variants in wastewater. The method's flexibility permits timely modifications, enabling the integration of emerging variants and adaptations to evolving SARS-CoV-2 genetics. Of particular significance for low- and middle-income regions with limited surveillance capabilities, this technique can potentially be utilized to study a range of pathogens or viruses that possess diverse genetic sequences, similar to influenza.
Collapse
Affiliation(s)
- James E. Merrett
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Monica Nolan
- Victorian Department of Health, Melbourne, Victoria, Australia
| | - Leon Hartman
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Nijoy John
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Faculty of Science, The University of Melbourne, Parkville, Victoria, Australia
| | - Brianna Flynn
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Louise Baker
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Christelle Schang
- Environmental and Public Health Microbiology Lab, Monash University, Clayton, Victoria, Australia
| | - David McCarthy
- Environmental and Public Health Microbiology Lab, Monash University, Clayton, Victoria, Australia
- School of Civil and Environmental Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
| | - David Lister
- Victorian Department of Health, Melbourne, Victoria, Australia
| | - Ngai Ning Cheng
- Victorian Department of Health, Melbourne, Victoria, Australia
- South Australian Water Corporation, Adelaide, South Australia, Australia
| | - Nick Crosbie
- Melbourne Water Corporation, Docklands, Victoria, Australia
| | - Rachael Poon
- Victorian Department of Health, Melbourne, Victoria, Australia
| | - Aaron Jex
- Population Health and Immunity Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Faculty of Science, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
5
|
D’Acunto E, Muzi A, Marchese S, Donnici L, Chiarini V, Bucci F, Pavoni E, Ferrara FF, Cappelletti M, Arriga R, Serrao SM, Peluzzi V, Principato E, Compagnone M, Pinto E, Luberto L, Stoppoloni D, Lahm A, Groß R, Seidel A, Wettstein L, Münch J, Goodhead A, Parisot J, De Francesco R, Ciliberto G, Marra E, Aurisicchio L, Roscilli G. Isolation and Characterization of Neutralizing Monoclonal Antibodies from a Large Panel of Murine Antibodies against RBD of the SARS-CoV-2 Spike Protein. Antibodies (Basel) 2024; 13:5. [PMID: 38247569 PMCID: PMC10801580 DOI: 10.3390/antib13010005] [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: 11/15/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/23/2024] Open
Abstract
The COVID-19 pandemic, once a global crisis, is now largely under control, a testament to the extraordinary global efforts involving vaccination and public health measures. However, the relentless evolution of SARS-CoV-2, leading to the emergence of new variants, continues to underscore the importance of remaining vigilant and adaptable. Monoclonal antibodies (mAbs) have stood out as a powerful and immediate therapeutic response to COVID-19. Despite the success of mAbs, the evolution of SARS-CoV-2 continues to pose challenges and the available antibodies are no longer effective. New variants require the ongoing development of effective antibodies. In the present study, we describe the generation and characterization of neutralizing mAbs against the receptor-binding domain (RBD) of the SARS-CoV-2 spike protein by combining plasmid DNA and recombinant protein vaccination. By integrating genetic immunization for rapid antibody production and the potent immune stimulation enabled by protein vaccination, we produced a rich pool of antibodies, each with unique binding and neutralizing specificities, tested with the ELISA, BLI and FACS assays and the pseudovirus assay, respectively. Here, we present a panel of mAbs effective against the SARS-CoV-2 variants up to Omicron BA.1 and BA.5, with the flexibility to target emerging variants. This approach ensures the preparedness principle is in place to address SARS-CoV-2 actual and future infections.
Collapse
Affiliation(s)
- Emanuela D’Acunto
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Alessia Muzi
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Silvia Marchese
- INGM-Istituto Nazionale di Genetica Molecolare “Romeo ed Erica Invernizzi”, 20122 Milan, Italy; (S.M.); (L.D.); (R.D.F.)
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy
| | - Lorena Donnici
- INGM-Istituto Nazionale di Genetica Molecolare “Romeo ed Erica Invernizzi”, 20122 Milan, Italy; (S.M.); (L.D.); (R.D.F.)
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy
| | | | - Federica Bucci
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Emiliano Pavoni
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Fabiana Fosca Ferrara
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Manuela Cappelletti
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Roberto Arriga
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Silvia Maria Serrao
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Valentina Peluzzi
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
- Department of Engineering, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Eugenia Principato
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
- Department of Experimental and Clinical Medicine, University Magna Graecia, 88100 Catanzaro, Italy
| | | | - Eleonora Pinto
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Laura Luberto
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Daniela Stoppoloni
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Armin Lahm
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (R.G.); (A.S.); (J.M.)
| | - Alina Seidel
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (R.G.); (A.S.); (J.M.)
| | - Lukas Wettstein
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (R.G.); (A.S.); (J.M.)
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, 89081 Ulm, Germany; (R.G.); (A.S.); (J.M.)
| | - Andrew Goodhead
- Carterra, 825 N. 300 W., Suite C309, Salt Lake City, UT 84103, USA; (A.G.); (J.P.)
| | - Judicael Parisot
- Carterra, 825 N. 300 W., Suite C309, Salt Lake City, UT 84103, USA; (A.G.); (J.P.)
| | - Raffaele De Francesco
- INGM-Istituto Nazionale di Genetica Molecolare “Romeo ed Erica Invernizzi”, 20122 Milan, Italy; (S.M.); (L.D.); (R.D.F.)
- Department of Pharmacological and Biomolecular Sciences (DiSFeB), University of Milan, 20133 Milan, Italy
| | - Gennaro Ciliberto
- Tumor Immunology and Immunotherapy Unit, IRCSS Regina Elena National Cancer Institute, 00144 Rome, Italy;
| | - Emanuele Marra
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Luigi Aurisicchio
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| | - Giuseppe Roscilli
- Takis Biotech, 00128 Rome, Italy; (A.M.); (F.B.); (E.P.); (F.F.F.); (M.C.); (R.A.); (S.M.S.); (V.P.); (E.P.); (E.P.); (L.L.); (D.S.); (A.L.); (E.M.); (L.A.)
| |
Collapse
|
6
|
Amin FG, Elfiky AA, Nassar AM. In silico targeting of SARS-CoV-2 spike receptor-binding domain from different variants with chaga mushroom terpenoids. J Biomol Struct Dyn 2024; 42:1079-1087. [PMID: 37042960 DOI: 10.1080/07391102.2023.2199084] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/30/2023] [Indexed: 04/13/2023]
Abstract
Terpenoids from the chaga mushroom have been identified as potential antiviral agents against SARS-CoV-2. This is because it can firmly bind to the viral spike receptor binding domain (RBD) and the auxiliary host cell receptor glucose-regulated protein 78 (GRP78). The current work examines the association of the chaga mushroom terpenoids with the RBD of various SARS-CoV-2 variants, including alpha, beta, gamma, delta, and omicron. This association was compared to the SARS-CoV-2 wild-type (WT) RBD using molecular docking analysis and molecular dynamics modeling. The outcomes demonstrated that the mutant RBDs, which had marginally greater average binding affinities (better binding) than the WT, were successfully inhibited by the chaga mushroom terpenoids. The results suggest that the chaga mushroom can be effective against various SARS-CoV-2 variants by targeting both the host-cell surface receptor GRP78 and the viral spike RBD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Fatma G Amin
- Physics Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Abdo A Elfiky
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Aaya M Nassar
- Biophysics Department, Faculty of Science, Cairo University, Giza, Egypt
- Department of Clinical Research and Leadership, School of Medicine and Health Sciences, George Washington University, Washington, DC, USA
| |
Collapse
|
7
|
Skarbinski J, Nugent JR, Wood MS, Liu L, Bullick T, Schapiro JM, Arunleung P, Morales C, Amsden LB, Hsiao CA, Wadford DA, Chai SJ, Reingold A, Wyman SK. Severe Acute Respiratory Syndrome Coronavirus 2 Delta Variant Genomic Variation Associated With Breakthrough Infection in Northern California: A Retrospective Cohort Study. J Infect Dis 2023; 228:878-888. [PMID: 37195913 PMCID: PMC11009495 DOI: 10.1093/infdis/jiad164] [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: 03/01/2023] [Revised: 05/09/2023] [Accepted: 05/15/2023] [Indexed: 05/19/2023] Open
Abstract
BACKGROUND The association between severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) genomic variation and breakthrough infection is not well defined among persons with Delta variant SARS-CoV-2 infection. METHODS In a retrospective cohort, we assessed whether individual nonlineage defining mutations and overall genomic variation (including low-frequency alleles) were associated with breakthrough infection, defined as SARS-CoV-2 infection after coronavirus disease 2019 primary vaccine series. We identified all nonsynonymous single-nucleotide polymorphisms, insertions, and deletions in SARS-CoV-2 genomes with ≥5% allelic frequency and population frequency of ≥5% and ≤95%. Using Poisson regression, we assessed the association with breakthrough infection for each individual mutation and a viral genomic risk score. RESULTS Thirty-six mutations met our inclusion criteria. Among 12 744 persons infected with Delta variant SARS-CoV-2, 5949 (47%) were vaccinated and 6795 (53%) were unvaccinated. Viruses with a viral genomic risk score in the highest quintile were 9% more likely to be associated with breakthrough infection than viruses in the lowest quintile, but including the risk score improved overall predictive model performance (measured by C statistic) by only +0.0006. CONCLUSIONS Genomic variation within SARS-CoV-2 Delta variant was weakly associated with breakthrough infection, but several potential nonlineage defining mutations were identified that might contribute to immune evasion by SARS-CoV-2.
Collapse
Affiliation(s)
- Jacek Skarbinski
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
- Department of Infectious Diseases, Oakland Medical Center, Kaiser Permanente Northern California, Oakland, California, USA
- Physician Researcher Program, Kaiser Permanente Northern California, Oakland, California, USA
- The Permanente Medical Group, Kaiser Permanente Northern California, Oakland, California, USA
| | - Joshua R Nugent
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Mariah S Wood
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Liyan Liu
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Teal Bullick
- Viral and Rickettsial Disease Laboratory, California Department of Public Health, Richmond, California, USA
| | - Jeffrey M Schapiro
- The Permanente Medical Group, Kaiser Permanente Northern California, Oakland, California, USA
| | - Phacharee Arunleung
- Viral and Rickettsial Disease Laboratory, California Department of Public Health, Richmond, California, USA
| | - Christina Morales
- Viral and Rickettsial Disease Laboratory, California Department of Public Health, Richmond, California, USA
| | - Laura B Amsden
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Crystal A Hsiao
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Debra A Wadford
- Viral and Rickettsial Disease Laboratory, California Department of Public Health, Richmond, California, USA
| | - Shua J Chai
- Career Epidemiology Field Officer, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Arthur Reingold
- School of Public Health, University of California, Berkeley, Berkeley, California, USA
| | - Stacia K Wyman
- Innovative Genomics Institute, University of California, Berkeley, Berkeley, California, USA
| |
Collapse
|
8
|
Kumar N, Taily IM, Singh C, Kumar S, Rajmani RS, Chakraborty D, Sharma A, Singh P, Thakur KG, Varadarajan R, Ringe RP, Banerjee P, Banerjee I. Identification of diphenylurea derivatives as novel endocytosis inhibitors that demonstrate broad-spectrum activity against SARS-CoV-2 and influenza A virus both in vitro and in vivo. PLoS Pathog 2023; 19:e1011358. [PMID: 37126530 PMCID: PMC10174524 DOI: 10.1371/journal.ppat.1011358] [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: 11/08/2022] [Revised: 05/11/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023] Open
Abstract
Rapid evolution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza A virus (IAV) poses enormous challenge in the development of broad-spectrum antivirals that are effective against the existing and emerging viral strains. Virus entry through endocytosis represents an attractive target for drug development, as inhibition of this early infection step should block downstream infection processes, and potentially inhibit viruses sharing the same entry route. In this study, we report the identification of 1,3-diphenylurea (DPU) derivatives (DPUDs) as a new class of endocytosis inhibitors, which broadly restricted entry and replication of several SARS-CoV-2 and IAV strains. Importantly, the DPUDs did not induce any significant cytotoxicity at concentrations effective against the viral infections. Examining the uptake of cargoes specific to different endocytic pathways, we found that DPUDs majorly affected clathrin-mediated endocytosis, which both SARS-CoV-2 and IAV utilize for cellular entry. In the DPUD-treated cells, although virus binding on the cell surface was unaffected, internalization of both the viruses was drastically reduced. Since compounds similar to the DPUDs were previously reported to transport anions including chloride (Cl-) across lipid membrane and since intracellular Cl- concentration plays a critical role in regulating vesicular trafficking, we hypothesized that the observed defect in endocytosis by the DPUDs could be due to altered Cl- gradient across the cell membrane. Using in vitro assays we demonstrated that the DPUDs transported Cl- into the cell and led to intracellular Cl- accumulation, which possibly affected the endocytic machinery by perturbing intracellular Cl- homeostasis. Finally, we tested the DPUDs in mice challenged with IAV and mouse-adapted SARS-CoV-2 (MA 10). Treatment of the infected mice with the DPUDs led to remarkable body weight recovery, improved survival and significantly reduced lung viral load, highlighting their potential for development as broad-spectrum antivirals.
Collapse
Affiliation(s)
- Nirmal Kumar
- Cellular Virology Lab, Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali (IISER Mohali), Mohali, India
| | - Irshad Maajid Taily
- Department of Chemistry, Indian Institute of Technology Ropar (IIT Ropar), Rupnagar, Punjab, India
| | - Charandeep Singh
- Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR-IMTECH), Chandigarh, India
| | - Sahil Kumar
- Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR-IMTECH), Chandigarh, India
| | - Raju S. Rajmani
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore (IISc), Bengaluru, India
| | - Debajyoti Chakraborty
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore (IISc), Bengaluru, India
| | - Anshul Sharma
- Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR-IMTECH), Chandigarh, India
| | - Priyanka Singh
- Department of Chemistry, Indian Institute of Technology Ropar (IIT Ropar), Rupnagar, Punjab, India
| | - Krishan Gopal Thakur
- Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR-IMTECH), Chandigarh, India
| | - Raghavan Varadarajan
- Molecular Biophysics Unit, Indian Institute of Science, Bangalore (IISc), Bengaluru, India
| | - Rajesh P. Ringe
- Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR-IMTECH), Chandigarh, India
| | - Prabal Banerjee
- Department of Chemistry, Indian Institute of Technology Ropar (IIT Ropar), Rupnagar, Punjab, India
| | - Indranil Banerjee
- Cellular Virology Lab, Department of Biological Sciences, Indian Institute of Science Education and Research, Mohali (IISER Mohali), Mohali, India
| |
Collapse
|
9
|
Gopalan V, Chandran A, Arumugam K, Sundaram M, Velladurai S, Govindan K, Azhagesan N, Jeyavel P, Dhandapani P, Sivasubramanian S, Kitambi SS. Distribution and Functional Analyses of Mutations in Spike Protein and Phylogenic Diversity of SARS-CoV-2 Variants Emerged during the Year 2021 in India. J Glob Infect Dis 2023; 15:43-51. [PMID: 37469462 PMCID: PMC10353649 DOI: 10.4103/jgid.jgid_178_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 11/27/2022] [Accepted: 01/04/2023] [Indexed: 07/21/2023] Open
Abstract
Introduction Prolonged COVID-19 pandemic accelerates the emergence and transmissibility of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) variants through the accumulation of adaptive mutations. Particularly, adaptive mutations in spike (S) protein of SARS-CoV-2 leads to increased viral infectivity, severe morbidity and mortality, and immune evasion. This study focuses on the phylodynamic distribution of SARS-CoV-2 variants during the year 2021 in India besides analyzing the functional significance of mutations in S-protein of SARS-CoV-2 variants. Methods Whole genome of SARS-CoV-2 sequences (n = 87957) from the various parts of India over the period of January to December 2021 was retrieved from Global Initiative on Sharing All Influenza Data. All the S-protein sequences were subjected to clade analysis, variant calling, protein stability, immune escape potential, structural divergence, Furin cleavage efficiency, and phylogenetic analysis using various in silico tools. Results Delta variant belonging to 21A, 21I, and 21J clades was found to be predominant throughout the year 2021 though many variants were also present. A total of 4639 amino acid mutations were found in S-protein. D614G was the most predominant mutation in the S-protein followed by P681R, L452R, T19R, T478K, and D950N. The highest number of mutations was found in the N-terminal domain of S-protein. Mutations in the crucial sites of S-protein impacting pathogenicity, immunogenicity, and fusogenicity were identified. Intralineage diversity analysis showed that certain variants of SARS-CoV-2 possess high diversification. Conclusions The study has disclosed the distribution of various variants including the Delta, the predominant variant, in India throughout the year 2021. The study has identified mutations in S-protein of each SARS-CoV-2 variant that can significantly impact the virulence, immune evasion, increased transmissibility, high morbidity, and mortality. In addition, it is found that mutations acquired during each viral replication cycle introduce new sub-lineages as studied by intralineage diversity analysis.
Collapse
Affiliation(s)
- Vidya Gopalan
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India
| | - Aswathi Chandran
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India
| | - Kishore Arumugam
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India
| | - Monisha Sundaram
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India
| | - Selvakumar Velladurai
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India
| | - Karthikeyan Govindan
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India
| | - Nivetha Azhagesan
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India
| | - Padmapriya Jeyavel
- Department of Virology, King Institute of Preventive Medicine and Research, Chennai, Tamil Nadu, India
| | - Prabu Dhandapani
- Department of Microbiology, Dr. ALM Post Graduate, Institute of Basic Medical Sciences, University of Madras, Chennai, Tamil Nadu, India
| | | | - Satish Srinivas Kitambi
- Department of Translational Sciences, Institute for Healthcare Education and Translational Sciences, Hyderabad, Telengana, India
| |
Collapse
|
10
|
Zhang H, Jin H, Yan F, Song Y, Dai J, Jiao C, Bai Y, Sun J, Liu D, Wang S, Zhang M, Lu J, Huang J, Huang P, Li Y, Xia X, Wang H. An inactivated recombinant rabies virus chimerically expressed RBD induces humoral and cellular immunity against SARS-CoV-2 and RABV. Virol Sin 2023; 38:244-256. [PMID: 36587795 PMCID: PMC9797420 DOI: 10.1016/j.virs.2022.12.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 12/26/2022] [Indexed: 12/30/2022] Open
Abstract
Many studies suggest that severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can infect various animals and transmit among animals, and even to humans, posing a threat to humans and animals. There is an urgent need to develop inexpensive and efficient animal vaccines to prevent and control coronavirus disease 2019 (COVID-19) in animals. Rabies virus (RABV) is another important zoonotic pathogen that infects almost all warm-blooded animals and poses a great public health threat. The present study constructed two recombinant chimeric viruses expressing the S1 and RBD proteins of the SARS-CoV-2 Wuhan01 strain based on a reverse genetic system of the RABV SRV9 strain and evaluated their immunogenicity in mice, cats and dogs. The results showed that both inactivated recombinant viruses induced durable neutralizing antibodies against SARS-CoV-2 and RABV and a strong cellular immune response in mice. Notably, inactivated SRV-nCoV-RBD induced earlier antibody production than SRV-nCoV-S1, which was maintained at high levels for longer periods. Inactivated SRV-nCoV-RBD induced neutralizing antibodies against both SARS-CoV-2 and RABV in cats and dogs, with a relatively broad-spectrum cross-neutralization capability against the SARS-CoV-2 pseudoviruses including Alpha, Beta, Gamma, Delta, and Omicron, showing potential to be used as a safe bivalent vaccine candidate against COVID-19 and rabies in animals.
Collapse
Affiliation(s)
- Haili Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Hongli Jin
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China; Changchun Sino Biotechnology Co., Ltd., Changchun, 130012, China
| | - Feihu Yan
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Yumeng Song
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Jiaxin Dai
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Cuicui Jiao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Yujie Bai
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Jingxuan Sun
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Di Liu
- Changchun Sino Biotechnology Co., Ltd., Changchun, 130012, China
| | - Shen Wang
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Mengyao Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Jilong Lu
- Changchun Sino Biotechnology Co., Ltd., Changchun, 130012, China
| | - Jingbo Huang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Pei Huang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Yuanyuan Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China
| | - Xianzhu Xia
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130122, China
| | - Hualei Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, 130062, China.
| |
Collapse
|
11
|
COVID-19 vaccination hesitancy is not all a conspiracy theory: A qualitative study from Iran. Acta Psychol (Amst) 2023; 233:103839. [PMID: 36652822 PMCID: PMC9841080 DOI: 10.1016/j.actpsy.2023.103839] [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: 04/25/2022] [Revised: 01/05/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The COVID-19 pandemic has burdened disastrous mortality and morbidity rates in society all over the world. While vaccination is one of the most effective immunization methods to control infectious diseases globally, some have avoided receiving the vaccine. We have aimed to investigate the reasons behind the hesitancy of vaccination among healthcare workers. METHOD We performed ten semi-structured interviews with volunteered healthcare workers of Rasoul Akram hospital. Then each interview was anonymized, and Braun and Clarke's thematic analysis method was used to analyze the interviews. RESULTS Our data analysis revealed thirty-eight different codes as reasons for vaccination hesitancy among our interviewees. All these thirty-eight codes were grouped into ten sub-themes, and these sub-themes were further grouped into our four main themes: 1. Fear of side effects, 2. Distrust, 3. Inefficiency, and 4. Non-necessity. Fear of side effects was the most frequent reason that interviewees mentioned. Also, Half of the interviewees mentioned distrust as a reason for COVID-19 vaccine hesitancy. At least once, all interviewees mentioned that they believed vaccination is inefficient. Some interviewees had beliefs and reasons that made them assume vaccination is simply not necessary. CONCLUSION Fear of side effects, distrust, inefficiency, and non-necessity were the reasons that our participants refused to get vaccinated. Fear of side effects and distrust were the most common reasons that led to non-vaccination.
Collapse
|
12
|
Hamburg M, Poland GA. The time is now for committed and comprehensive action to attain more broadly protective coronavirus vaccines: The coronavirus vaccines R&D roadmap. Vaccine 2023; 41:2645-2647. [PMID: 36828718 PMCID: PMC9941882 DOI: 10.1016/j.vaccine.2023.02.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 02/16/2023] [Indexed: 02/23/2023]
Affiliation(s)
| | - Gregory A. Poland
- Mayo Vaccine Research Group, Mayo Clinic, Rochester, MN, USA,Corresponding author at: Mayo Vaccine Research Group, Mayo Clinic and Foundation, 611C Guggenheim Building, 200 First Street, SW, Rochester, MN, USA
| |
Collapse
|
13
|
Kuo HC, Kuo KC, Du PX, Keskin BB, Su WY, Ho TS, Tsai PS, Pau CH, Shih HC, Huang YH, Weng KP, Syu GD. Profiling humoral immunity after mixing and matching COVID-19 vaccines using SARS-CoV-2 variant protein microarrays. Mol Cell Proteomics 2023; 22:100507. [PMID: 36787877 PMCID: PMC9922205 DOI: 10.1016/j.mcpro.2023.100507] [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: 06/07/2022] [Revised: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 02/16/2023] Open
Abstract
In November 2022, 68% of the population received at least one dose of COVID-19 vaccines. Due to the ongoing mutations, especially for the variants of concern (VOCs), it is important to monitor the humoral immune responses after different vaccination strategies. In this study, we developed a SARS-CoV-2 variant protein microarray that contained the spike proteins from the VOCs, e.g., alpha, beta, gamma, delta, and omicron, to quantify the binding antibody and surrogate neutralizing antibody. Plasmas were collected after two doses of matching AZD1222 (AZx2), two doses of matching mRNA-1273 (Mx2), or mixing AZD1222 and mRNA-1273 (AZ+M). The results showed a significant decrease of surrogate neutralizing antibodies against the receptor-binding domain in all VOCs in AZx2 and Mx2 but not AZ+M. A similar but minor reduction pattern of surrogate neutralizing antibodies against the extracellular domain was observed. While Mx2 exhibited a higher surrogate neutralizing level against all VOCs compared to AZx2, AZ+M showed an even higher surrogate neutralizing level in gamma and omicron compared to Mx2. It is worth noting that the binding antibody displayed a low correlation to the surrogate neutralizing antibody (R-square 0.130-0.382). This study delivers insights into humoral immunities, SARS-CoV-2 mutations, and mixing and matching vaccine strategies, which may provide a more effective vaccine strategy especially in preventing omicron.
Collapse
Affiliation(s)
- Ho-Chang Kuo
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan,Kawasaki Disease Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan,College of Medicine, Chang Gung University, Taoyuan, Taiwan 33302
| | - Kuang-Che Kuo
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan
| | - Pin-Xian Du
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Batuhan Birol Keskin
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Wen-Yu Su
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Tzong-Shiann Ho
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 701, Taiwan R.O.C.,Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan 701, Taiwan R.O.C.,Department of Pediatrics, Tainan Hospital, Ministry of Health and Welfare, Tainan 700, Taiwan R.O.C
| | - Pei-Shan Tsai
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Chi Ho Pau
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan
| | - Hsi-Chang Shih
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Ying-Hsien Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan,Kawasaki Disease Center, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 83301, Taiwan,College of Medicine, Chang Gung University, Taoyuan, Taiwan 33302
| | - Ken-Pen Weng
- Congenital Structural Heart Disease Center, Department of Pediatrics, Kaohsiung Veterans General Hospital, Kaohsiung 813414, Taiwan,School of Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Guan-Da Syu
- Department of Biotechnology and Bioindustry Sciences, National Cheng Kung University, Tainan 701, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan 701, Taiwan; Medical Device Innovation Center, National Cheng Kung University, Tainan 701, Taiwan.
| |
Collapse
|
14
|
Capistrano KJ, Richner J, Schwartz J, Mukherjee SK, Shukla D, Naqvi AR. Host microRNAs exhibit differential propensity to interact with SARS-CoV-2 and variants of concern. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166612. [PMID: 36481486 PMCID: PMC9721271 DOI: 10.1016/j.bbadis.2022.166612] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Revised: 10/19/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022]
Abstract
A significant number of SARS-CoV-2-infected individuals naturally overcome viral infection, suggesting the existence of a potent endogenous antiviral mechanism. As an innate defense mechanism, microRNA (miRNA) pathways in mammals have evolved to restrict viruses, besides regulating endogenous mRNAs. In this study, we systematically examined the complete repertoire of human miRNAs for potential binding sites on SARS-CoV-2 Wuhan-Hu-1, Beta, Delta, and Omicron. Human miRNA and viral genome interaction were analyzed using RNAhybrid 2.2 with stringent parameters to identify highly bonafide miRNA targets. Using publicly available data, we filtered for miRNAs expressed in lung epithelial cells/tissue and oral keratinocytes, concentrating on the miRNAs that target SARS-CoV-2 S protein mRNAs. Our results show a significant loss of human miRNA and SARS-CoV-2 interactions in Omicron (130 miRNAs) compared to Wuhan-Hu-1 (271 miRNAs), Beta (279 miRNAs), and Delta (275 miRNAs). In particular, hsa-miR-3150b-3p and hsa-miR-4784 show binding affinity for S protein of Wuhan strain but not Beta, Delta, and Omicron. Loss of miRNA binding sites on N protein was also observed for Omicron. Through Ingenuity Pathway Analysis (IPA), we examined the experimentally validated and highly predicted functional role of these miRNAs. We found that hsa-miR-3150b-3p and hsa-miR-4784 have several experimentally validated or highly predicted target genes in the Toll-like receptor, IL-17, Th1, Th2, interferon, and coronavirus pathogenesis pathways. Focusing on the coronavirus pathogenesis pathway, we found that hsa-miR-3150b-3p and hsa-miR-4784 are highly predicted to target MAPK13. Exploring miRNAs to manipulate viral genome/gene expression can provide a promising strategy with successful outcomes by targeting specific VOCs.
Collapse
Affiliation(s)
- Kristelle J Capistrano
- Mucosal Immunology Lab, College of Dentistry, University of Illinois Chicago, Chicago 60612, IL, USA
| | - Justin Richner
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago 60612, IL, USA
| | - Joel Schwartz
- Molecular Pathology Lab, College of Dentistry, University of Illinois at Chicago, Chicago, IL, USA
| | - Sunil K Mukherjee
- Division of Plant Pathology, Indian Agricultural Research Institute, New Delhi, India
| | - Deepak Shukla
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, Chicago 60612, IL, USA; Department of Ophthalmology and Visual Sciences, Ocular Virology Laboratory, University of Illinois Chicago, Chicago 60612, IL, USA
| | - Afsar R Naqvi
- Mucosal Immunology Lab, College of Dentistry, University of Illinois Chicago, Chicago 60612, IL, USA.
| |
Collapse
|
15
|
Cox M, Peacock TP, Harvey WT, Hughes J, Wright DW, Willett BJ, Thomson E, Gupta RK, Peacock SJ, Robertson DL, Carabelli AM. SARS-CoV-2 variant evasion of monoclonal antibodies based on in vitro studies. Nat Rev Microbiol 2023; 21:112-124. [PMID: 36307535 PMCID: PMC9616429 DOI: 10.1038/s41579-022-00809-7] [Citation(s) in RCA: 141] [Impact Index Per Article: 141.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/26/2022] [Indexed: 01/20/2023]
Abstract
Monoclonal antibodies (mAbs) offer a treatment option for individuals with severe COVID-19 and are especially important in high-risk individuals where vaccination is not an option. Given the importance of understanding the evolution of resistance to mAbs by SARS-CoV-2, we reviewed the available in vitro neutralization data for mAbs against live variants and viral constructs containing spike mutations of interest. Unfortunately, evasion of mAb-induced protection is being reported with new SARS-CoV-2 variants. The magnitude of neutralization reduction varied greatly among mAb-variant pairs. For example, sotrovimab retained its neutralization capacity against Omicron BA.1 but showed reduced efficacy against BA.2, BA.4 and BA.5, and BA.2.12.1. At present, only bebtelovimab has been reported to retain its efficacy against all SARS-CoV-2 variants considered here. Resistance to mAb neutralization was dominated by the action of epitope single amino acid substitutions in the spike protein. Although not all observed epitope mutations result in increased mAb evasion, amino acid substitutions at non-epitope positions and combinations of mutations also contribute to evasion of neutralization. This Review highlights the implications for the rational design of viral genomic surveillance and factors to consider for the development of novel mAb therapies.
Collapse
Affiliation(s)
- MacGregor Cox
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Cambridge, UK
| | - Thomas P Peacock
- Department of Infectious Disease, St Mary's Medical School, Imperial College London, London, UK
| | - William T Harvey
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Joseph Hughes
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Derek W Wright
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Brian J Willett
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Emma Thomson
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK
| | - Ravindra K Gupta
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Cambridge, UK
- Cambridge Institute of Therapeutic Immunology & Infectious Disease (CITIID), Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
| | - Sharon J Peacock
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Cambridge, UK
| | - David L Robertson
- MRC-University of Glasgow Centre for Virus Research, University of Glasgow, Glasgow, UK.
| | | |
Collapse
|
16
|
Sasaki M, Tabata K, Kishimoto M, Itakura Y, Kobayashi H, Ariizumi T, Uemura K, Toba S, Kusakabe S, Maruyama Y, Iida S, Nakajima N, Suzuki T, Yoshida S, Nobori H, Sanaki T, Kato T, Shishido T, Hall WW, Orba Y, Sato A, Sawa H. S-217622, a SARS-CoV-2 main protease inhibitor, decreases viral load and ameliorates COVID-19 severity in hamsters. Sci Transl Med 2023; 15:eabq4064. [PMID: 36327352 PMCID: PMC9765455 DOI: 10.1126/scitranslmed.abq4064] [Citation(s) in RCA: 33] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In parallel with vaccination, oral antiviral agents are highly anticipated to act as countermeasures for the treatment of the coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Oral antiviral medication demands not only high antiviral activity but also target specificity, favorable oral bioavailability, and high metabolic stability. Although a large number of compounds have been identified as potential inhibitors of SARS-CoV-2 infection in vitro, few have proven to be effective in vivo. Here, we show that oral administration of S-217622 (ensitrelvir), an inhibitor of SARS-CoV-2 main protease (Mpro; also known as 3C-like protease), decreases viral load and ameliorates disease severity in SARS-CoV-2-infected hamsters. S-217622 inhibited viral proliferation at low nanomolar to submicromolar concentrations in cells. Oral administration of S-217622 demonstrated favorable pharmacokinetic properties and accelerated recovery from acute SARS-CoV-2 infection in hamster recipients. Moreover, S-217622 exerted antiviral activity against SARS-CoV-2 variants of concern, including the highly pathogenic Delta variant and the recently emerged Omicron BA.5 and BA.2.75 variants. Overall, our study provides evidence that S-217622, an antiviral agent that is under evaluation in a phase 3 clinical trial (clinical trial registration no. jRCT2031210350), has remarkable antiviral potency and efficacy against SARS-CoV-2 and is a prospective oral therapeutic option for COVID-19.
Collapse
Affiliation(s)
- Michihito Sasaki
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan.,Corresponding author. (M.S.); (H.S.)
| | - Koshiro Tabata
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan
| | - Mai Kishimoto
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan
| | - Yukari Itakura
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan
| | - Hiroko Kobayashi
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan
| | - Takuma Ariizumi
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan
| | - Kentaro Uemura
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan.,Shionogi & Co., Ltd., Osaka 561-0825, Japan.,Laboratory of Biomolecular Science, Faculty of Pharmaceutical Science, Hokkaido University, Sapporo, 060-0812, Japan
| | - Shinsuke Toba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan.,Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Shinji Kusakabe
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan.,Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Yuki Maruyama
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan.,Shionogi & Co., Ltd., Osaka 561-0825, Japan
| | - Shun Iida
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Noriko Nakajima
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Tadaki Suzuki
- Department of Pathology, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | | | | | | | | | | | - William W. Hall
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan.,National Virus Reference Laboratory, School of Medicine, University College of Dublin, 4, Ireland.,Global Virus Network, Baltimore, MD, 21201, USA
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan.,International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan
| | - Akihiko Sato
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan.,Shionogi & Co., Ltd., Osaka 561-0825, Japan.,Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, 001-0021, Japan
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-220, Japan.,International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, 001-0020, Japan.,Global Virus Network, Baltimore, MD, 21201, USA.,Institute for Vaccine Research and Development (IVReD), Hokkaido University, Sapporo, 001-0021, Japan.,One Health Research Center, Hokkaido University, Sapporo, 001-0020, Japan.,Corresponding author. (M.S.); (H.S.)
| |
Collapse
|
17
|
Insight into free energy and dynamic cross-correlations of residue for binding affinity of antibody and receptor binding domain SARS-CoV-2. Heliyon 2023; 9:e12667. [PMID: 36618128 PMCID: PMC9809146 DOI: 10.1016/j.heliyon.2022.e12667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 11/12/2022] [Accepted: 12/20/2022] [Indexed: 01/05/2023] Open
Abstract
SARS-CoV-2 virus continues to evolve and mutate causing most of the mutated variants resist to many of the therapeutic monoclonal antibodies (mAbs). Despite several mAbs retained neutralizing capability for Omicron BA.1 and BA.2, reduction in neutralization potency was reported. Hence, effort of searching for mAb that is broader in neutralization breadth without losing the neutralizing ability is continued. MW06 was reported with capability in neutralizing most of the variants of concern (VOC) and it binds to the conserved region (left flank) near epitope mAb sotrovimab (S309). In this study, binding affinity of mAb MW06 and its cocktail formulation with MW05 for receptor binding domain (RBD) SARS-CoV-2 virus was investigated under molecular dynamics simulations (MDs). Binding free energies computed by Molecular Mechanics Generalised Born Surface Area (MM-GBSA) algorithm predicted the binding affinity of MW06 for RBD BA.1 (-53 kcal/mol) as strong as RBD wildtype (-58 kcal/mol) while deterioration was observed for RBD BA.2 (-43 kcal/mol). Alike S309 and MW06, simulated cocktail mAb (MW05 and MW06)-RBD interactions suggested the neutralizing capability of the cocktail formulation for RBD BA.1 and BA.2 reduced. Meanwhile, residue pairs that favour the communication between the mAb and RBD have been identified by decomposing the free energy per pairwise residue basis. Apart from understanding the effects of mutation occurred in the RBD region on human angiotensin-converting enzyme 2 (hACE2) binding, impact of heavily mutated RBD on mAb-RBD interactions was investigated in this study as well. In addition to energetic profile obtained from MDs, plotting the dynamics cross-correlation map of the mAb-RBD complex under elastic network model (ENM) was aimed to understand the cross-correlations between residue fluctuations. It allows simple and rapid analysis on the motions or dynamics of the protein residues of mAbs and RBD in complex. Protein residues having correlated motions are normally part of the structural domains of the protein and their respective motions and protein function are related. Motion of mutated RBD residues and mAb residues was less correlated while their respective interactions energy computed to be higher. The combined techniques of MDs and ENM offered simplicity in understanding dynamics and energy contribution that explain binding affinity of mAb-RBD complexes.
Collapse
|
18
|
Alp Onen E, Sonmez K, Yildirim F, Demirci EK, Gurel A. Development, analysis, and preclinical evaluation of inactivated vaccine candidate for prevention of Covid-19 disease. ALL LIFE 2022. [DOI: 10.1080/26895293.2022.2099468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
Affiliation(s)
- Engin Alp Onen
- Kocak Pharmaceuticals, Vaccine and Biotechnology R&D, Organize Sanayi Bölgesi, Kapakli/Tekirdag, Turkey
| | - Kivilcim Sonmez
- Faculty of Veterinary Medicine, Pathology Department, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Funda Yildirim
- Faculty of Veterinary Medicine, Pathology Department, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | | | - Aydin Gurel
- Faculty of Veterinary Medicine, Pathology Department, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
19
|
Lin Y, Huang Z, Xu X, Du W, Alias H, Hu Z, Wong LP. Multi-dimensional psychosocial factors influencing the willingness to receive a COVID-19 vaccine booster: A survey among the public in Mainland China. Hum Vaccin Immunother 2022; 18:2126667. [PMID: 36165498 DOI: 10.1080/21645515.2022.2126667] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
This study aimed to investigate multi-dimensional psychological and social factors that influence the willingness to receive a COVID-19 vaccine booster in China. A nationwide cross-sectional online survey was conducted between March and April 2022. A total of 6375 complete responses were received. The majority were of age 18 to 40 years old (80.0%) and college-educated (49.2%). In total, 79% responded extremely willing to receive a COVID-19 vaccine booster. By demographics, younger age, females, higher education, and participants with the lowest income reported higher willingness. Having a very good health status (odds ratio [OR] 3.56, 95% confidence interval [CI] 2.92-4.34) and a higher score of vaccine confidence (OR 3.50, 95% CI 2.98-4.11) were associated with an increased willingness to receive a booster shot. Experiencing no side effects with primary COVID-19 vaccination (OR 2.46, 95% CI 1.89-3.20) and higher perceived susceptibility of COVID-19 infection (OR 2.38, 95% CI 1.92-2.95) were also associated with an increased willingness to receive a booster shot. A variety of psychosocial factors, namely having no chronic diseases, lower perceived concern over the safety of a booster shot, higher perceived severity of COVID-19 infection, and a higher level of institutional trust, were also significantly associated with greater willingness to get a booster shot. In conclusion, the present study adds evidence to the significant role of psychosocial factors in predicting COVID-19 vaccine booster acceptance and provides insights to design interventions to increase booster uptake in certain targeted demographic groups.
Collapse
Affiliation(s)
- Yulan Lin
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, FJ, China
| | - Zhiwen Huang
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, FJ, China
| | - Xiaonan Xu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, FJ, China
| | - Wei Du
- School of Medicine and Health Management, Guizhou Medical University, Guiyang, GZ, China.,Centre for Epidemiology and Evidence-Based Practice, Department of Social and Preventive Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Haridah Alias
- Centre for Epidemiology and Evidence-Based Practice, Department of Social and Preventive Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | - Zhijian Hu
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, FJ, China
| | - Li Ping Wong
- Department of Epidemiology and Health Statistics, School of Public Health, Fujian Medical University, Fuzhou, FJ, China.,Centre for Epidemiology and Evidence-Based Practice, Department of Social and Preventive Medicine, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
20
|
Zhang J, Han ZB, Liang Y, Zhang XF, Jin YQ, Du LF, Shao S, Wang H, Hou JW, Xu K, Lei W, Lei ZH, Liu ZM, Zhang J, Hou YN, Liu N, Shen FJ, Wu JJ, Zheng X, Li XY, Li X, Huang WJ, Wu GZ, Su JG, Li QM. A mosaic-type trimeric RBD-based COVID-19 vaccine candidate induces potent neutralization against Omicron and other SARS-CoV-2 variants. eLife 2022; 11:e78633. [PMID: 36004719 PMCID: PMC9481243 DOI: 10.7554/elife.78633] [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: 03/14/2022] [Accepted: 08/22/2022] [Indexed: 11/29/2022] Open
Abstract
Large-scale populations in the world have been vaccinated with COVID-19 vaccines, however, breakthrough infections of SARS-CoV-2 are still growing rapidly due to the emergence of immune-evasive variants, especially Omicron. It is urgent to develop effective broad-spectrum vaccines to better control the pandemic of these variants. Here, we present a mosaic-type trimeric form of spike receptor-binding domain (mos-tri-RBD) as a broad-spectrum vaccine candidate, which carries the key mutations from Omicron and other circulating variants. Tests in rats showed that the designed mos-tri-RBD, whether used alone or as a booster shot, elicited potent cross-neutralizing antibodies against not only Omicron but also other immune-evasive variants. Neutralizing antibody ID50 titers induced by mos-tri-RBD were substantially higher than those elicited by homo-tri-RBD (containing homologous RBDs from prototype strain) or the BIBP inactivated COVID-19 vaccine (BBIBP-CorV). Our study indicates that mos-tri-RBD is highly immunogenic, which may serve as a broad-spectrum vaccine candidate in combating SARS-CoV-2 variants including Omicron.
Collapse
Affiliation(s)
- Jing Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Zi Bo Han
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Yu Liang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xue Feng Zhang
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Yu Qin Jin
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
| | - Li Fang Du
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Shuai Shao
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Hui Wang
- Beijing Institute of Biological Products Company LimitedBeijingChina
| | - Jun Wei Hou
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Ke Xu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Wenwen Lei
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Ze Hua Lei
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Zhao Ming Liu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Jin Zhang
- Beijing Institute of Biological Products Company LimitedBeijingChina
| | - Ya Nan Hou
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Ning Liu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Fu Jie Shen
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Jin Juan Wu
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xiang Zheng
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xin Yu Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Xin Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Wei Jin Huang
- National Institutes for Food and Drug Control (NIFDC)BeijingChina
| | - Gui Zhen Wu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention (China CDC)BeijingChina
| | - Ji Guo Su
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| | - Qi Ming Li
- The Sixth Laboratory, National Vaccine and Serum Institute (NVSI)BeijingChina
- National Engineering Center for New Vaccine ResearchBeijingChina
| |
Collapse
|
21
|
Clinical and Virological Features of Patients Hospitalized with Different Types of COVID-19 Vaccination in Mexico City. Vaccines (Basel) 2022; 10:vaccines10081181. [PMID: 35893830 PMCID: PMC9330015 DOI: 10.3390/vaccines10081181] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/27/2022] [Accepted: 07/04/2022] [Indexed: 02/05/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) vaccines effectively protect against severe disease and death. However, the impact of the vaccine used, viral variants, and host factors on disease severity remain poorly understood. This work aimed to compare COVID-19 clinical presentations and outcomes in vaccinated and unvaccinated patients in Mexico City. From March to September 2021, clinical, demographic characteristics, and viral variants were obtained from 1014 individuals with a documented SARS-CoV-2 infection. We compared unvaccinated, partially vaccinated, and fully vaccinated patients, stratifying by age groups. We also fitted multivariate statistical models to evaluate the impact of vaccination status, SARS-CoV-2 lineages, vaccine types, and clinical parameters. Most hospitalized patients were unvaccinated. In patients over 61 years old, mortality was significantly higher in unvaccinated compared to fully vaccinated individuals. In patients aged 31 to 60 years, vaccinated patients were more likely to be outpatients (46%) than unvaccinated individuals (6.1%). We found immune disease and age above 61 years old to be risk factors, while full vaccination was found to be the most protective factor against in-hospital death. This study suggests that vaccination is essential to reduce mortality in a comorbid population such as that of Mexico.
Collapse
|
22
|
Yang L, Tang L, Zhang M, Liu C. Recent Advances in the Molecular Design and Delivery Technology of mRNA for Vaccination Against Infectious Diseases. Front Immunol 2022; 13:896958. [PMID: 35928814 PMCID: PMC9345514 DOI: 10.3389/fimmu.2022.896958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 06/20/2022] [Indexed: 12/02/2022] Open
Abstract
Vaccines can prevent many millions of illnesses against infectious diseases and save numerous lives every year. However, traditional vaccines such as inactivated viral and live attenuated vaccines cannot adapt to emerging pandemics due to their time-consuming development. With the global outbreak of the COVID-19 epidemic, the virus continues to evolve and mutate, producing mutants with enhanced transmissibility and virulence; the rapid development of vaccines against such emerging global pandemics becomes more and more critical. In recent years, mRNA vaccines have been of significant interest in combating emerging infectious diseases due to their rapid development and large-scale production advantages. However, their development still suffers from many hurdles such as their safety, cellular delivery, uptake, and response to their manufacturing, logistics, and storage. More efforts are still required to optimize the molecular designs of mRNA molecules with increased protein expression and enhanced structural stability. In addition, a variety of delivery systems are also needed to achieve effective delivery of vaccines. In this review, we highlight the advances in mRNA vaccines against various infectious diseases and discuss the molecular design principles and delivery systems of associated mRNA vaccines. The current state of the clinical application of mRNA vaccine pipelines against various infectious diseases and the challenge, safety, and protective effect of associated vaccines are also discussed.
Collapse
Affiliation(s)
- Lu Yang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lin Tang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Ming Zhang
- Department of Pathology, Peking University International Hospital, Beijing, China
- *Correspondence: Chaoyong Liu, ; Ming Zhang,
| | - Chaoyong Liu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China
- *Correspondence: Chaoyong Liu, ; Ming Zhang,
| |
Collapse
|
23
|
Pang Z, Hu R, Tian L, Lou F, Chen Y, Wang S, He S, Zhu S, An X, Song L, Liu F, Tong Y, Fan H. Overview of Breastfeeding Under COVID-19 Pandemic. Front Immunol 2022; 13:896068. [PMID: 35711421 PMCID: PMC9192965 DOI: 10.3389/fimmu.2022.896068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 04/22/2022] [Indexed: 12/19/2022] Open
Abstract
During the global pandemic of coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), pregnant and lactating women are at higher risk of infection. The potential of viral intrauterine transmission and vertical transmission by breastfeeding has raised wide concerns. Breastmilk is rich in nutrients that contribute to infant growth and development, and reduce the incidence rate of infant illness and death, as well as inhibit pathogens significantly, and protect infants from infection. Although it is controversial whether mothers infected with COVID-19 should continue to breastfeed, many countries and international organizations have provided recommendations and guidance for breastfeeding. This review presents the risks and benefits of breastfeeding for mothers infected with COVID-19, and the reasons for the absence of SARS-CoV-2 active virus in human milk. In addition, the antiviral mechanisms of nutrients in breastmilk, the levels of SARS-CoV-2 specific antibodies in breastmilk from COVID-19 infected mothers and vaccinated mothers are also summarized and discussed, aiming to provide some support and recommendations for both lactating mothers and infants to better deal with the COVID-19 pandemic.
Collapse
Affiliation(s)
- Zehan Pang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Ruolan Hu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lili Tian
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Fuxing Lou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Yangzhen Chen
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Shuqi Wang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Shiting He
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Shaozhou Zhu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Xiaoping An
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lihua Song
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Feitong Liu
- Health & Happiness Group, Health & Happiness Research, China Aesearch and Innovation, Guangzhou, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
24
|
Koley T, Goswami A, Kumar M, Upadhyay N, Hariprasad G. Comparative Structural Analysis of Human ACE2 Receptor with Spike Protein of SARS-CoV-2 Variants: Implications to Understand Infectivity of the Virus. Adv Appl Bioinform Chem 2022; 15:21-27. [PMID: 35734581 PMCID: PMC9208465 DOI: 10.2147/aabc.s360787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 06/01/2022] [Indexed: 11/23/2022] Open
Abstract
Purpose Spike protein on SARS-CoV-2 virus plays an integral part during infection as cell entry depends on binding of this protein to human ACE2 receptor. Understanding of infectivity by these variants necessitates a comparative structural analysis of complexes of spike protein-receptor binding domain (RBD) of these variants to receptor. Methodology Wild type SARS-CoV-2 spike protein sequence was retrieved from the UniProt database, and mutations of five variants at receptor binding domain were manually incorporated and aligned using Clustal Omega. Crystal structure complexes of human ACE2 receptor with spike protein RBD domain of SARS-CoV-2 variants of wild type, α, β, and δ were extracted from the RCSB database. Wild type SARS-CoV-2 complex with receptor was used as template to generate model complexes of receptor with spike protein RBD of γ and omicron variants through WinCoot program. These were energy minimized and validated and molecular dynamic simulation was performed using Desmond simulation program. Results Mutations are distributed across the entire length of RBD, but the maximum number of mutations are seen at 11 positions within binding interface motifs of six variant sequences. Interface of spike protein RBDs with human ACE2-receptor shows different mix of hydrogen bonded and ionic interactions. Alpha and β variants have few interactions, while γ and δ variants have higher number of interactions compared to wild type variant. Omicron variant, with 10 polar interactions including two ionic bonds, has the highest binding energy. Conclusion Different mutations on RBD of spike protein results in varying quantity and quality of interactions, thereby affecting potency of each variant. Variations in binding are due to interactions of mutant residues and induced conformational changes on loops of RBDs. Variants α and β have a low potency, while, γ, δ, and omicron have a higher potency. These results correlate with viral infectivity and place clinical observations in the right perspective.
Collapse
Affiliation(s)
- Tirthankar Koley
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Arunima Goswami
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Manoj Kumar
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Neelam Upadhyay
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| | - Gururao Hariprasad
- Department of Biophysics, All India Institute of Medical Sciences, New Delhi, 110029, India
| |
Collapse
|
25
|
Plikusiene I, Maciulis V, Juciute S, Maciuleviciene R, Balevicius S, Ramanavicius A, Ramanaviciene A. Investigation and Comparison of Specific Antibodies' Affinity Interaction with SARS-CoV-2 Wild-Type, B.1.1.7, and B.1.351 Spike Protein by Total Internal Reflection Ellipsometry. BIOSENSORS 2022; 12:351. [PMID: 35624652 PMCID: PMC9139055 DOI: 10.3390/bios12050351] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/13/2022] [Accepted: 05/16/2022] [Indexed: 05/21/2023]
Abstract
SARS-CoV-2 vaccines provide strong protection against COVID-19. However, the emergence of SARS-CoV-2 variants has raised concerns about the efficacy of vaccines. In this study, we investigated the interactions of specific polyclonal human antibodies (pAb-SCoV2-S) produced after vaccination with the Vaxzevria vaccine with the spike proteins of three SARS-CoV-2 variants of concern: wild-type, B.1.1.7, and B.1.351. Highly sensitive, label-free, and real-time monitoring of these interactions was accomplished using the total internal reflection ellipsometry method. Thermodynamic parameters such as association and dissociation rate constants, the stable immune complex formation rate constant (kr), the equilibrium association and dissociation (KD) constants and steric factors (Ps) were calculated using a two-step irreversible binding mathematical model. The results obtained show that the KD values for the specific antibody interactions with all three types of spike protein are in the same nanomolar range. The KD values for B.1.1.7 and B.1.351 suggest that the antibody produced after vaccination can successfully protect the population from the alpha (B.1.1.7) and beta (B.1.351) SARS-CoV-2 mutations. The steric factors (Ps) obtained for all three types of spike proteins showed a 100-fold lower requirement for the formation of an immune complex when compared with nucleocapsid protein.
Collapse
Affiliation(s)
- Ieva Plikusiene
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, 03225 Vilnius, Lithuania; (I.P.); (V.M.); (S.J.); (R.M.); (S.B.); (A.R.)
| | - Vincentas Maciulis
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, 03225 Vilnius, Lithuania; (I.P.); (V.M.); (S.J.); (R.M.); (S.B.); (A.R.)
- State Research Institute Center for Physical and Technological Sciences, Sauletekio Ave. 3, 03225 Vilnius, Lithuania
| | - Silvija Juciute
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, 03225 Vilnius, Lithuania; (I.P.); (V.M.); (S.J.); (R.M.); (S.B.); (A.R.)
| | - Ruta Maciuleviciene
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, 03225 Vilnius, Lithuania; (I.P.); (V.M.); (S.J.); (R.M.); (S.B.); (A.R.)
| | - Saulius Balevicius
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, 03225 Vilnius, Lithuania; (I.P.); (V.M.); (S.J.); (R.M.); (S.B.); (A.R.)
- State Research Institute Center for Physical and Technological Sciences, Sauletekio Ave. 3, 03225 Vilnius, Lithuania
| | - Arunas Ramanavicius
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, 03225 Vilnius, Lithuania; (I.P.); (V.M.); (S.J.); (R.M.); (S.B.); (A.R.)
| | - Almira Ramanaviciene
- NanoTechnas—Center of Nanotechnology and Materials Science, Faculty of Chemistry and Geosciences, Vilnius University, Naugarduko Str. 24, 03225 Vilnius, Lithuania; (I.P.); (V.M.); (S.J.); (R.M.); (S.B.); (A.R.)
| |
Collapse
|
26
|
Qin H, Qiu H, He ST, Hong B, Liu K, Lou F, Li M, Hu P, Kong X, Song Y, Liu Y, Pu M, Han P, Li M, An X, Song L, Tong Y, Fan H, Wang R. Efficient disinfection of SARS-CoV-2-like coronavirus, pseudotyped SARS-CoV-2 and other coronaviruses using cold plasma induces spike protein damage. JOURNAL OF HAZARDOUS MATERIALS 2022; 430:128414. [PMID: 35149493 PMCID: PMC8813208 DOI: 10.1016/j.jhazmat.2022.128414] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/19/2022] [Accepted: 01/31/2022] [Indexed: 05/05/2023]
Abstract
Coronavirus disease 2019 (COVID-19) has become a worldwide public health emergency, and the high transmission of SARS-CoV-2 variants has raised serious concerns. Efficient disinfection methods are crucial for the prevention of viral transmission. Herein, pulse power-driven cold atmospheric plasma (CAP), a novel sterilization strategy, was found to potently inactivate SARS-CoV-2-like coronavirus GX_P2V, six strains of major epidemic SARS-CoV-2 variants and even swine coronavirus PEDV and SADS-CoV within 300 s (with inhibition rate more than 99%). We identified four dominant short-lived reactive species, ONOO-, 1O2, O2- and·OH, generated in response to CAP and distinguished their roles in the inactivation of GX_P2V and SARS-CoV-2 spike protein receptor binding domain (RBD), which is responsible for recognition and binding to human angiotensin-converting enzyme 2 (hACE2). Our study provides detailed evidence of a novel surface disinfection strategy for SARS-CoV-2 and other coronaviruses.
Collapse
Affiliation(s)
- Hongbo Qin
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China; College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Hengju Qiu
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Shi-Ting He
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Bixia Hong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Ke Liu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Fuxing Lou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Maochen Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Pan Hu
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xianghao Kong
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yujie Song
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yuchen Liu
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Mingfang Pu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Pengjun Han
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Mengzhe Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaoping An
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lihua Song
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China; Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China.
| | - Ruixue Wang
- College of Mechanical and Electrical Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
27
|
Li M, Wang H, Tian L, Pang Z, Yang Q, Huang T, Fan J, Song L, Tong Y, Fan H. COVID-19 vaccine development: milestones, lessons and prospects. Signal Transduct Target Ther 2022; 7:146. [PMID: 35504917 PMCID: PMC9062866 DOI: 10.1038/s41392-022-00996-y] [Citation(s) in RCA: 176] [Impact Index Per Article: 88.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 04/11/2022] [Accepted: 04/13/2022] [Indexed: 12/15/2022] Open
Abstract
With the constantly mutating of SARS-CoV-2 and the emergence of Variants of Concern (VOC), the implementation of vaccination is critically important. Existing SARS-CoV-2 vaccines mainly include inactivated, live attenuated, viral vector, protein subunit, RNA, DNA, and virus-like particle (VLP) vaccines. Viral vector vaccines, protein subunit vaccines, and mRNA vaccines may induce additional cellular or humoral immune regulations, including Th cell responses and germinal center responses, and form relevant memory cells, greatly improving their efficiency. However, some viral vector or mRNA vaccines may be associated with complications like thrombocytopenia and myocarditis, raising concerns about the safety of these COVID-19 vaccines. Here, we systemically assess the safety and efficacy of COVID-19 vaccines, including the possible complications and different effects on pregnant women, the elderly, people with immune diseases and acquired immunodeficiency syndrome (AIDS), transplant recipients, and cancer patients. Based on the current analysis, governments and relevant agencies are recommended to continue to advance the vaccine immunization process. Simultaneously, special attention should be paid to the health status of the vaccines, timely treatment of complications, vaccine development, and ensuring the lives and health of patients. In addition, available measures such as mix-and-match vaccination, developing new vaccines like nanoparticle vaccines, and optimizing immune adjuvant to improve vaccine safety and efficacy could be considered.
Collapse
Affiliation(s)
- Maochen Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Han Wang
- Laboratory for Clinical Immunology, Harbin Children's Hospital, Harbin, China
| | - Lili Tian
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zehan Pang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Qingkun Yang
- College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing, China
| | - Tianqi Huang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Junfen Fan
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Lihua Song
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China. .,Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing, China.
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China.
| |
Collapse
|
28
|
Jaiswal V, Lee HJ. Conservation and Evolution of Antigenic Determinants of SARS-CoV-2: An Insight for Immune Escape and Vaccine Design. Front Immunol 2022; 13:832106. [PMID: 35444664 PMCID: PMC9014086 DOI: 10.3389/fimmu.2022.832106] [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] [Received: 12/09/2021] [Accepted: 03/07/2022] [Indexed: 11/20/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is the most devastating pandemic of the century, which is still far from over. The remarkable success of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines is the working hope, but the evolving variants are the huge concern that can turn the tide. Potential immune escape mutations (PIEMs) in the past and circulating variants were not studied at large scale (all available data). Hence, the conservation of antigenic determinants (epitopes) was analyzed in all available sequences of SARS-CoV-2 according to time (months), proteins, hosts, and variants. Numerous highly conserved B- and T-cell epitopes were identified in 24 proteins of SARS-CoV-2. A decrease in the conservation of epitopes with time was observed in almost all proteins, which was more rapid in neutralizing epitopes. Delta variant still has the highest PIEM in the circulating strains, which pose threat to the effectiveness of current vaccines. The inclusion of identified, highly conserved, and important epitopes in subunit vaccines can increase vaccine effectiveness against evolving variants. Trends in the conservation of epitopes in different proteins, hosts, and variants with time may also help to inspire the counter measure against the current pandemic.
Collapse
Affiliation(s)
- Varun Jaiswal
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam-si, South Korea
| | - Hae-Jeung Lee
- Department of Food and Nutrition, College of BioNano Technology, Gachon University, Seongnam-si, South Korea.,Institute for Aging and Clinical Nutrition Research, Gachon University, Seongnam-si, South Korea.,Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, South Korea
| |
Collapse
|
29
|
Tian L, Pang Z, Li M, Lou F, An X, Zhu S, Song L, Tong Y, Fan H, Fan J. Molnupiravir and Its Antiviral Activity Against COVID-19. Front Immunol 2022; 13:855496. [PMID: 35444647 PMCID: PMC9013824 DOI: 10.3389/fimmu.2022.855496] [Citation(s) in RCA: 71] [Impact Index Per Article: 35.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/09/2022] [Indexed: 12/15/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) constitutes a major worldwide public health threat and economic burden. The pandemic is still ongoing and the SARS-CoV-2 variants are still emerging constantly, resulting in an urgent demand for new drugs to treat this disease. Molnupiravir, a biological prodrug of NHC (β-D-N(4)-hydroxycytidine), is a novel nucleoside analogue with a broad-spectrum antiviral activity against SARS-CoV, SARS-CoV-2, Middle East respiratory syndrome coronavirus (MERS-CoV), influenza virus, respiratory syncytial virus (RSV), bovine viral diarrhea virus (BVDV), hepatitis C virus (HCV) and Ebola virus (EBOV). Molnupiravir showed potent therapeutic and prophylactic activity against multiple coronaviruses including SARS-CoV-2, SARS-CoV, and MERS-CoV in animal models. In clinical trials, molnupiravir showed beneficial effects for mild to moderate COVID-19 patients with a favorable safety profile. The oral bioavailability and potent antiviral activity of molnupiravir highlight its potential utility as a therapeutic candidate against COVID-19. This review presents the research progress of molnupiravir starting with its discovery and synthesis, broad-spectrum antiviral effects, and antiviral mechanism. In addition, the preclinical studies, antiviral resistance, clinical trials, safety, and drug tolerability of molnupiravir are also summarized and discussed, aiming to expand our knowledge on molnupiravir and better deal with the COVID-19 epidemic.
Collapse
Affiliation(s)
- Lili Tian
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Zehan Pang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Maochen Li
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Fuxing Lou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Xiaoping An
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Shaozhou Zhu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lihua Song
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
- *Correspondence: Junfen Fan, ; Huahao Fan, ; Yigang Tong, ; Lihua Song,
| | - Yigang Tong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
- *Correspondence: Junfen Fan, ; Huahao Fan, ; Yigang Tong, ; Lihua Song,
| | - Huahao Fan
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
- *Correspondence: Junfen Fan, ; Huahao Fan, ; Yigang Tong, ; Lihua Song,
| | - Junfen Fan
- Department of Neurology, Institute of Cerebrovascular Disease Research, Xuanwu Hospital, Capital Medical University, Beijing, China
- *Correspondence: Junfen Fan, ; Huahao Fan, ; Yigang Tong, ; Lihua Song,
| |
Collapse
|
30
|
de León UAP, Avila-Vales E, Huang KL. Modeling COVID-19 dynamic using a two-strain model with vaccination. CHAOS, SOLITONS, AND FRACTALS 2022; 157:111927. [PMID: 35185299 PMCID: PMC8847090 DOI: 10.1016/j.chaos.2022.111927] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/05/2022] [Accepted: 02/15/2022] [Indexed: 05/21/2023]
Abstract
Multiple strains of the SARS-CoV-2 have arisen and jointly influence the trajectory of the coronavirus disease (COVID-19) pandemic. However, current models rarely account for this multi-strain dynamics and their different transmission rate and response to vaccines. We propose a new mathematical model that accounts for two virus variants and the deployment of a vaccination program. To demonstrate utility, we applied the model to determine the control reproduction number ( R c ) and the per day infection, death and recovery rates of each strain in the US pandemic. The model dynamics predicted the rise of the alpha variant and shed light on potential impact of the delta variant in 2021. We obtained the minimum percentage of fully vaccinated individuals to reduce the spread of the variants in combination with other intervention strategies to deaccelerate the rise of a multi-strain pandemic.
Collapse
Affiliation(s)
| | - Eric Avila-Vales
- Facultad de Matemáticas, Universidad Autónoma de Yucatán. Anillo Periférico Norte. Tablaje Catastral 13615, C.P. 97119. Mérida, Yucatán
| | - Kuan-Lin Huang
- Department of Genetics and Genomic Sciences. Center for Transformative Disease Modeling. Tisch Cancer Institute. Icahn Institute for Data Science and Genomic Technology. Icahn School of Medicine at Mount Sinai. New York. NY 10029. USA
| |
Collapse
|
31
|
Patiño LH, Castañeda S, Muñoz M, Ballesteros N, Ramirez AL, Luna N, Guerrero-Araya E, Pérez J, Correa-Cárdenas CA, Duque MC, Méndez C, Oliveros C, Shaban MV, Paniz-Mondolfi AE, Ramírez JD. Epidemiological Dynamics of SARS-CoV-2 Variants During Social Protests in Cali, Colombia. Front Med (Lausanne) 2022; 9:863911. [PMID: 35433760 PMCID: PMC9008484 DOI: 10.3389/fmed.2022.863911] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/10/2022] [Indexed: 01/29/2023] Open
Abstract
BackgroundThe third wave of the global health crisis attributed to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus reached Colombia in March 2021. Over the following 6 months, it was interpolated by manifestations of popular disapproval to the actual political regime—with multiple protests sprouting throughout the country. Large social gatherings seeded novel coronavirus disease 2019 (COVID-19) variants in big cities and propagated their facile spread, leading to increased rates of hospitalizations and deaths.MethodsIn this article, we evaluate the effective reproduction number (Rt) dynamics of SARS-CoV-2 in Cali, Colombia, between 4 April 2021 and 31 July 2021 based on the analysis of 228 genomes.ResultsOur results showed clear contrast in Rt values between the period of frequent protests (Rt > 1), and the preceding and following months (Rt < 1). Genomic analyses revealed 16 circulating SARS-CoV-2 lineages during the initial period—including variants of concern (VOCs) (Alpha, Gamma, and Delta) and variants of interest (VOIs) (Lambda and Mu). Furthermore, we noticed the Mu variant dominating the COVID-19 distribution schema as the months progressed. We identified four principal clusters through phylogenomic analyses—each one of potentially independent introduction to the city. Two of these were associated with the Mu variant, one associated with the Gamma variant, and one with the Lambda variant.ConclusionOur results chronicle the impact of large group assemblies on the epidemiology of COVID-19 during this intersection of political turmoil and sanitary crisis in Cali, Colombia. We emphasize upon the effects of limited biosecurity strategies (which had characterized this time period), on the spread of highly virulent strains throughout Cali and greater Colombia.
Collapse
Affiliation(s)
- Luz H. Patiño
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Sergio Castañeda
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Marina Muñoz
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Agencia Nacional de Investigación y Desarrollo (ANID)—Millennium Science Initiative Program—Millennium Nucleus in the Biology of the Intestinal Microbiota, Santiago, Chile
| | - Nathalia Ballesteros
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Angie L. Ramirez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Nicolas Luna
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Enzo Guerrero-Araya
- Agencia Nacional de Investigación y Desarrollo (ANID)—Millennium Science Initiative Program—Millennium Nucleus in the Biology of the Intestinal Microbiota, Santiago, Chile
- Microbiota-Host Interactions and Clostridia Research Group, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Julie Pérez
- Grupo de Investigación en Enfermedades Tropicales del Ejército (GINETEJ), Laboratorio de Referencia e Investigación, Dirección de Sanidad Ejército, Bogotá, Colombia
| | - Camilo A. Correa-Cárdenas
- Grupo de Investigación en Enfermedades Tropicales del Ejército (GINETEJ), Laboratorio de Referencia e Investigación, Dirección de Sanidad Ejército, Bogotá, Colombia
| | - Maria Clara Duque
- Grupo de Investigación en Enfermedades Tropicales del Ejército (GINETEJ), Laboratorio de Referencia e Investigación, Dirección de Sanidad Ejército, Bogotá, Colombia
| | - Claudia Méndez
- Grupo de Investigación en Enfermedades Tropicales del Ejército (GINETEJ), Laboratorio de Referencia e Investigación, Dirección de Sanidad Ejército, Bogotá, Colombia
| | - Carolina Oliveros
- Grupo de Investigación en Enfermedades Tropicales del Ejército (GINETEJ), Laboratorio de Referencia e Investigación, Dirección de Sanidad Ejército, Bogotá, Colombia
| | - Maryia V. Shaban
- Incubadora Venezolana de la Ciencia (IVC), Centro de Investigaciones Biomédicas IDB, Barquisimeto, Venezuela
| | - Alberto E. Paniz-Mondolfi
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- Molecular Microbiology Laboratory, Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- *Correspondence: Juan David Ramírez, ;
| |
Collapse
|
32
|
Nie C, Sahoo AK, Netz RR, Herrmann A, Ballauff M, Haag R. Charge Matters: Mutations in Omicron Variant Favor Binding to Cells. Chembiochem 2022; 23:e202100681. [PMID: 35020256 PMCID: PMC9015620 DOI: 10.1002/cbic.202100681] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 01/11/2022] [Indexed: 11/13/2022]
Abstract
Evidence is strengthening to suggest that the novel SARS-CoV-2 mutant Omicron, with its more than 60 mutations, will spread and dominate worldwide. Although the mutations in the spike protein are known, the molecular basis for why the additional mutations in the spike protein that have not previously occurred account for Omicron's higher infection potential, is not understood. We propose, based on chemical rational and molecular dynamics simulations, that the elevated occurrence of positively charged amino acids in certain domains of the spike protein (Delta: +4; Omicron: +5 vs. wild type) increases binding to cellular polyanionic receptors, such as heparan sulfate due to multivalent charge-charge interactions. This observation is a starting point for targeted drug development.
Collapse
Affiliation(s)
- Chuanxiong Nie
- Institut für Chemie und BiochemieFreie Universität BerlinTakustrasse 314195BerlinGermany
| | - Anil Kumar Sahoo
- Fachbereich PhysikFreie Universität Berlin14195BerlinGermany
- Max Planck Institute of Colloids and Interfaces14476PotsdamGermany
| | - Roland R. Netz
- Fachbereich PhysikFreie Universität Berlin14195BerlinGermany
| | - Andreas Herrmann
- Institut für Chemie und BiochemieFreie Universität BerlinTakustrasse 314195BerlinGermany
| | - Matthias Ballauff
- Institut für Chemie und BiochemieFreie Universität BerlinTakustrasse 314195BerlinGermany
| | - Rainer Haag
- Institut für Chemie und BiochemieFreie Universität BerlinTakustrasse 314195BerlinGermany
| |
Collapse
|
33
|
Vo GV, Bagyinszky E, An SSA. COVID-19 Genetic Variants and Their Potential Impact in Vaccine Development. Microorganisms 2022; 10:598. [PMID: 35336173 PMCID: PMC8954257 DOI: 10.3390/microorganisms10030598] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 02/28/2022] [Accepted: 03/07/2022] [Indexed: 01/18/2023] Open
Abstract
In the two years since the SARS-CoV-2 pandemic started, it has caused over 5 million deaths and 400 million infected cases, and the world continues to be on high alert for COVID-19. Among the variants of interest and concern of SARS-CoV-2, the current Omicron (B.1.1.529) and stealth Omicron (BA.2) raised serious concerns due to rapid rates of infection caused by numerous mutations in the spike protein, which could escape from the antibody-mediated neutralization and increase the risk of reinfections. Hence, this work aims to describe the most relevant mutations in the SARS-CoV-2 spike protein, discuss vaccine against variant of concerns, describe rare adverse events after COVID-19 vaccination, introduce the most available promising COVID-19 vaccine candidates, and provide few perspectives of the future variants.
Collapse
Affiliation(s)
- Giau Van Vo
- Department of Biomedical Engineering, School of Medicine, Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City 70000, Vietnam;
- Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, Vietnam National University, Ho Chi Minh City (VNU-HCM), Ho Chi Minh City 70000, Vietnam
- Vietnam National University Ho Chi Minh City (VNU-HCM), Ho Chi Minh City 70000, Vietnam
| | - Eva Bagyinszky
- Graduate School of Environment Department of Industrial and Environmental Engineering, Gachon University, Seongnam 13120, Korea
| | - Seong Soo A. An
- Department of Bionano Technology, Gachon University, Seongnam 13120, Korea
| |
Collapse
|
34
|
Acharjee A, Stephen Kingsly J, Kamat M, Kurlawala V, Chakraborty A, Vyas P, Vaishnav R, Srivastava S. Rise of the SARS-CoV-2 Variants: can proteomics be the silver bullet? Expert Rev Proteomics 2022; 19:197-212. [PMID: 35655386 DOI: 10.1080/14789450.2022.2085564] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The challenges posed by emergent strains of SARS-CoV-2 need to be tackled by contemporary scientific approaches, with proteomics playing a significant role. AREAS COVERED In this review, we provide a brief synthesis of the impact of proteomics technologies in elucidating disease pathogenesis and classifiers for the prognosis of COVID-19 and propose proteomics methodologies that could play a crucial role in understanding emerging variants and their altered disease pathology. From aiding the design of novel drug candidates to facilitating the identification of T cell vaccine targets, we have discussed the impact of proteomics methods in COVID-19 research. Techniques varied as mass spectrometry, single-cell proteomics, multiplexed ELISA arrays, high-density proteome arrays, surface plasmon resonance, immunopeptidomics, and in silico docking studies that have helped augment the fight against existing diseases were useful in preparing us to tackle SARS-CoV-2 variants. We also propose an action plan for a pipeline to combat emerging pandemics using proteomics technology by adopting uniform standard operating procedures and unified data analysis paradigms. EXPERT OPINION The knowledge about the use of diverse proteomics approaches for COVID-19 investigation will provide a framework for future basic research, better infectious disease prevention strategies, improved diagnostics, and targeted therapeutics.
Collapse
Affiliation(s)
- Arup Acharjee
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| | | | - Madhura Kamat
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be University), Mumbai, India
| | - Vishakha Kurlawala
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's NMIMS (Deemed-to-be University), Mumbai, India
| | | | - Priyanka Vyas
- Department of Biotechnology and Botany, Mahila PG Mahavidyalaya, J. N. V University, Jodhpur, India
| | - Radhika Vaishnav
- Department of Life Sciences, Ivy Tech Community College, Indianapolis, Indiana, USA
| | - Sanjeeva Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Mumbai, India
| |
Collapse
|
35
|
Singh BG, Gandhi VV, Phadnis PP, Kunwar A. Identification of a pyridine derivative of diselenides as a potent inhibitor of the main protease of SARS-CoV-2 through in silico screening and biochemical evaluation. NEW J CHEM 2022. [DOI: 10.1039/d2nj02744e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Among the 22 organoselenium compounds studied, 2-Py2Se2 & Nict2Se2 showed the highest affinity for Mpro. The biochemical studies confirmed their superiority as compared to standard compound like Ebselen in terms of the IC50 required for Mpro inhibition.
Collapse
Affiliation(s)
- B. G. Singh
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Mumbai-400085, India
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai-400085, India
| | - V. V. Gandhi
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Mumbai-400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai-400094, India
| | - P. P. Phadnis
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai-400085, India
- Homi Bhabha National Institute, Anushaktinagar, Mumbai-400094, India
| | - A. Kunwar
- Radiation & Photochemistry Division, Bhabha Atomic Research Centre, Mumbai-400085, India
- Chemistry Division, Bhabha Atomic Research Centre, Mumbai-400085, India
| |
Collapse
|