1
|
Choi Y, De Ridder D, Greub G. Genomic and spatial epidemiology: lessons learned from SARS-CoV-2 pandemic. Curr Opin HIV AIDS 2025; 20:287-293. [PMID: 40172549 PMCID: PMC11970598 DOI: 10.1097/coh.0000000000000936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
PURPOSE OF REVIEW The SARS-CoV-2 pandemic presented unprecedented challenges, particularly in understanding its complex spatial transmission patterns. The high transmissibility of the virus led to frequent super-spreading events. These events demonstrated clear spatial clustering patterns, often tied to specific events that facilitated transmission. The uneven geographic distribution of medical resources and varying access to care amplified the impact of SARS-CoV-2. Asymptomatic cases further complicated the situation, as infected individuals could silently spread the virus before being identified.Thus, this review examines how genomic and spatial epidemiology approaches can be integrated to answer some of the above-mentioned challenges. We first describe the methodological foundations of genomics and spatial epidemiology, detailing opportunities of their applications during the SARS-CoV-2 pandemic. We then present a novel interdisciplinary framework that combines these approaches to better guide public health interventions. RECENT FINDINGS During the pandemic, the genomic and spatial approaches were used to address key questions, including "how does the pathogen evolve and diversify?" and "how does the pathogen spread geographically?". Genomic epidemiology allows researchers to identify viral lineages and new variants. Conversely, spatial epidemiology focused on geographic distribution of infections, analyzing how the virus spread. However, despite their complementary nature, these approaches were largely applied independently during the pandemic. This separation limited our collective ability to fully understand the complex relationships between viral evolution and geographic spread. SUMMARY While phylogeography has traditionally combined phylogenetic and geographic data to understand long-term evolutionary patterns across large areas, events such as the recent SARS-CoV-2 pandemic demand frameworks that can inform public health interventions through joint analysis of genomic and local-scale spatial data.
Collapse
Affiliation(s)
- Yangji Choi
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne
| | - David De Ridder
- Group of Geospatial Molecular Epidemiology (GEOME), Laboratory for Biological Geochemistry (LGB), School of Architecture, Civil and Environmental Engineering (ENAC), École Polytechnique Fédérale de Lausanne (EPFL), Lausanne
- Group of Geographic Information Research and Analysis in Population Health (GIRAPH)
- Faculty of Medicine, University of Geneva (UNIGE)
- Division and Department of Primary Care Medicine, Geneva University Hospitals, Geneva
| | - Gilbert Greub
- Institute of Microbiology, Lausanne University Hospital and University of Lausanne
- Service of Infectious Diseases, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
2
|
Invenção MDCV, de Macêdo LS, de Moura IA, Santos LABDO, Espinoza BCF, de Pinho SS, Leal LRS, dos Santos DL, São Marcos BDF, Elsztein C, de Sousa GF, de Souza-Silva GA, Barros BRDS, Cruz LCDO, Maux JMDL, Silva Neto JDC, de Melo CML, Silva AJD, Batista MVDA, de Freitas AC. Design and Immune Profile of Multi-Epitope Synthetic Antigen Vaccine Against SARS-CoV-2: An In Silico and In Vivo Approach. Vaccines (Basel) 2025; 13:149. [PMID: 40006696 PMCID: PMC11861798 DOI: 10.3390/vaccines13020149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND The rapid advancement of the pandemic caused by SARS-CoV-2 and its variants reinforced the importance of developing easy-to-edit vaccines with fast production, such as multi-epitope DNA vaccines. The present study aimed to construct a synthetic antigen multi-epitope SARS-CoV-2 to produce a DNA vaccine. METHODS A database of previously predicted Spike and Nucleocapsid protein epitopes was created, and these epitopes were analyzed for immunogenicity, conservation, population coverage, and molecular docking. RESULTS A synthetic antigen with 15 epitopes considered immunogenic, conserved even in the face of variants and that were able to anchor themselves in the appropriate HLA site, together had more than 90% worldwide coverage. A multi-epitope construct was developed with the sequences of these peptides separated from each other by linkers, cloned into the pVAX1 vector. This construct was evaluated in vivo as a DNA vaccine and elicited T CD4+ and T CD8+ cell expansion in the blood and spleen. In hematological analyses, there was an increase in lymphocytes, monocytes, and neutrophils between the two doses. Furthermore, based on histopathological analysis, the vaccines did not cause any damage to the organs analyzed. CONCLUSIONS The present study generated a multi-epitope synthetic vaccine antigen capable of generating antibody-mediated and cellular immune responses.
Collapse
Affiliation(s)
- Maria da Conceição Viana Invenção
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (M.d.C.V.I.); (L.S.d.M.); (I.A.d.M.); (B.C.F.E.); (S.S.d.P.); (L.R.S.L.); (D.L.d.S.); (B.d.F.S.M.); (C.E.); (A.J.D.S.)
| | - Larissa Silva de Macêdo
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (M.d.C.V.I.); (L.S.d.M.); (I.A.d.M.); (B.C.F.E.); (S.S.d.P.); (L.R.S.L.); (D.L.d.S.); (B.d.F.S.M.); (C.E.); (A.J.D.S.)
| | - Ingrid Andrêssa de Moura
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (M.d.C.V.I.); (L.S.d.M.); (I.A.d.M.); (B.C.F.E.); (S.S.d.P.); (L.R.S.L.); (D.L.d.S.); (B.d.F.S.M.); (C.E.); (A.J.D.S.)
| | - Lucas Alexandre Barbosa de Oliveira Santos
- Laboratory of Molecular Genetics and Biotechnology (GMBio), Department of Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Brazil; (L.A.B.d.O.S.); (M.V.d.A.B.)
| | - Benigno Cristofer Flores Espinoza
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (M.d.C.V.I.); (L.S.d.M.); (I.A.d.M.); (B.C.F.E.); (S.S.d.P.); (L.R.S.L.); (D.L.d.S.); (B.d.F.S.M.); (C.E.); (A.J.D.S.)
| | - Samara Sousa de Pinho
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (M.d.C.V.I.); (L.S.d.M.); (I.A.d.M.); (B.C.F.E.); (S.S.d.P.); (L.R.S.L.); (D.L.d.S.); (B.d.F.S.M.); (C.E.); (A.J.D.S.)
| | - Lígia Rosa Sales Leal
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (M.d.C.V.I.); (L.S.d.M.); (I.A.d.M.); (B.C.F.E.); (S.S.d.P.); (L.R.S.L.); (D.L.d.S.); (B.d.F.S.M.); (C.E.); (A.J.D.S.)
| | - Daffany Luana dos Santos
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (M.d.C.V.I.); (L.S.d.M.); (I.A.d.M.); (B.C.F.E.); (S.S.d.P.); (L.R.S.L.); (D.L.d.S.); (B.d.F.S.M.); (C.E.); (A.J.D.S.)
| | - Bianca de França São Marcos
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (M.d.C.V.I.); (L.S.d.M.); (I.A.d.M.); (B.C.F.E.); (S.S.d.P.); (L.R.S.L.); (D.L.d.S.); (B.d.F.S.M.); (C.E.); (A.J.D.S.)
| | - Carolina Elsztein
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (M.d.C.V.I.); (L.S.d.M.); (I.A.d.M.); (B.C.F.E.); (S.S.d.P.); (L.R.S.L.); (D.L.d.S.); (B.d.F.S.M.); (C.E.); (A.J.D.S.)
| | - Georon Ferreira de Sousa
- Laboratory of Immunological and Antitumor Analysis, Keizo Asami Immunopathology Laboratory, Department of Antibiotics, Bioscience Center, Federal University of Pernambuco, Recife 50670-901, Brazil; (G.F.d.S.); (G.A.d.S.-S.); (B.R.d.S.B.); (L.C.d.O.C.); (C.M.L.d.M.)
| | - Guilherme Antonio de Souza-Silva
- Laboratory of Immunological and Antitumor Analysis, Keizo Asami Immunopathology Laboratory, Department of Antibiotics, Bioscience Center, Federal University of Pernambuco, Recife 50670-901, Brazil; (G.F.d.S.); (G.A.d.S.-S.); (B.R.d.S.B.); (L.C.d.O.C.); (C.M.L.d.M.)
| | - Bárbara Rafaela da Silva Barros
- Laboratory of Immunological and Antitumor Analysis, Keizo Asami Immunopathology Laboratory, Department of Antibiotics, Bioscience Center, Federal University of Pernambuco, Recife 50670-901, Brazil; (G.F.d.S.); (G.A.d.S.-S.); (B.R.d.S.B.); (L.C.d.O.C.); (C.M.L.d.M.)
| | - Leonardo Carvalho de Oliveira Cruz
- Laboratory of Immunological and Antitumor Analysis, Keizo Asami Immunopathology Laboratory, Department of Antibiotics, Bioscience Center, Federal University of Pernambuco, Recife 50670-901, Brazil; (G.F.d.S.); (G.A.d.S.-S.); (B.R.d.S.B.); (L.C.d.O.C.); (C.M.L.d.M.)
| | - Julliano Matheus de Lima Maux
- Laboratory of Cytological and Molecular Research, Department of Histology and Embriology, Federal University of Pernambuco, Recife 50670-901, Brazil; (J.M.d.L.M.); (J.d.C.S.N.)
| | - Jacinto da Costa Silva Neto
- Laboratory of Cytological and Molecular Research, Department of Histology and Embriology, Federal University of Pernambuco, Recife 50670-901, Brazil; (J.M.d.L.M.); (J.d.C.S.N.)
| | - Cristiane Moutinho Lagos de Melo
- Laboratory of Immunological and Antitumor Analysis, Keizo Asami Immunopathology Laboratory, Department of Antibiotics, Bioscience Center, Federal University of Pernambuco, Recife 50670-901, Brazil; (G.F.d.S.); (G.A.d.S.-S.); (B.R.d.S.B.); (L.C.d.O.C.); (C.M.L.d.M.)
| | - Anna Jéssica Duarte Silva
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (M.d.C.V.I.); (L.S.d.M.); (I.A.d.M.); (B.C.F.E.); (S.S.d.P.); (L.R.S.L.); (D.L.d.S.); (B.d.F.S.M.); (C.E.); (A.J.D.S.)
| | - Marcus Vinicius de Aragão Batista
- Laboratory of Molecular Genetics and Biotechnology (GMBio), Department of Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Brazil; (L.A.B.d.O.S.); (M.V.d.A.B.)
| | - Antonio Carlos de Freitas
- Laboratory of Molecular Studies and Experimental Therapy—LEMTE, Department of Genetics, Federal University of Pernambuco, Recife 50670-901, Brazil; (M.d.C.V.I.); (L.S.d.M.); (I.A.d.M.); (B.C.F.E.); (S.S.d.P.); (L.R.S.L.); (D.L.d.S.); (B.d.F.S.M.); (C.E.); (A.J.D.S.)
| |
Collapse
|
3
|
Sheehan J, Trauth AJ, Hagensee ME, Ramsay AJ. Characterization of Vaccine-Enhanced Humoral Immune Responses Against Emergent SARS-CoV-2 Variants in a Convalescent Cohort. Pathogens 2025; 14:44. [PMID: 39861005 PMCID: PMC11768806 DOI: 10.3390/pathogens14010044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/27/2025] Open
Abstract
Vaccination of COVID-19-convalescent individuals may generate 'hybrid' immunity of enhanced magnitude, durability, and cross-reactive breadth. Our primary goal was to characterize hybrid antibody (Ab) responses in a patient cohort infected with ancestral Wuhan-Hu-1 virus and vaccinated between 6 and 10 months later with the Wuhan-Hu-1-based BNT162b2 mRNA vaccine. We were particularly interested in determining the efficacy of neutralizing Ab responses against subsequently emergent SARS-CoV-2 variants. Sera collected at 3-monthly intervals over a period of 12 months were analyzed by ELISA for SARS-CoV-2 RBD-specific Ab responses, and also for neutralizing Ab activity using pseudovirus-based neutralization assays. We found that convalescent RBD-reactive IgG and IgA Ab responses did not decline significantly through 9 months post-diagnosis. These responses improved significantly following vaccination and remained elevated through at least 12-months. SARS-CoV-2 neutralizing Ab activity was detected in convalescent sera through 9 months post-diagnosis, although it trended downwards from 3 months. Neutralizing Ab activity against the Wuhan-Hu-1 strain was significantly improved by vaccination, to levels that persisted through the end of the study. However, sera collected from vaccinated convalescent subjects also had significant neutralization activity against Delta B.1.617.2 and Omicron variants that persisted for at least 2-3 months, unlike sera from unvaccinated convalescent controls. Thus, vaccination of Wuhan-Hu-1-convalescent individuals with the BNT162b2 vaccine improved and sustained protective neutralizing Ab activity against SARS-CoV-2, including cross-reactive neutralizing activity against variants that emerged months later.
Collapse
Affiliation(s)
- Jared Sheehan
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
| | - Amber J. Trauth
- Stanley S. Scott Cancer, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
| | - Michael E. Hagensee
- Department of Internal Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
| | - Alistair J. Ramsay
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA;
| |
Collapse
|
4
|
Barghash RF, Gemmati D, Awad AM, Elbakry MMM, Tisato V, Awad K, Singh AV. Navigating the COVID-19 Therapeutic Landscape: Unveiling Novel Perspectives on FDA-Approved Medications, Vaccination Targets, and Emerging Novel Strategies. Molecules 2024; 29:5564. [PMID: 39683724 PMCID: PMC11643501 DOI: 10.3390/molecules29235564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Amidst the ongoing global challenge of the SARS-CoV-2 pandemic, the quest for effective antiviral medications remains paramount. This comprehensive review delves into the dynamic landscape of FDA-approved medications repurposed for COVID-19, categorized as antiviral and non-antiviral agents. Our focus extends beyond conventional narratives, encompassing vaccination targets, repurposing efficacy, clinical studies, innovative treatment modalities, and future outlooks. Unveiling the genomic intricacies of SARS-CoV-2 variants, including the WHO-designated Omicron variant, we explore diverse antiviral categories such as fusion inhibitors, protease inhibitors, transcription inhibitors, neuraminidase inhibitors, nucleoside reverse transcriptase, and non-antiviral interventions like importin α/β1-mediated nuclear import inhibitors, neutralizing antibodies, and convalescent plasma. Notably, Molnupiravir emerges as a pivotal player, now licensed in the UK. This review offers a fresh perspective on the historical evolution of COVID-19 therapeutics, from repurposing endeavors to the latest developments in oral anti-SARS-CoV-2 treatments, ushering in a new era of hope in the battle against the pandemic.
Collapse
Affiliation(s)
- Reham F. Barghash
- Institute of Chemical Industries Research, National Research Centre, Dokki, Cairo 12622, Egypt
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Cairo 12451, Egypt
| | - Donato Gemmati
- Department of Translational Medicine, University of Ferrara, 44121 Ferrara, Italy
| | - Ahmed M. Awad
- Department of Chemistry, California State University Channel Islands, Camarillo, CA 93012, USA
| | - Mustafa M. M. Elbakry
- Faculty of Biotechnology, October University for Modern Sciences and Arts (MSA), Cairo 12451, Egypt
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo 11566, Egypt
| | - Veronica Tisato
- Centre Hemostasis & Thrombosis, University of Ferrara, 44121 Ferrara, Italy
| | - Kareem Awad
- Institute of Pharmaceutical and Drug Industries Research, National Research Center, Dokki, Cairo 12622, Egypt;
| | - Ajay Vikram Singh
- Department of Chemical and Product Safety, German Federal Institute for Risk Assessment (BfR), Max-Dohrn-Strasse 8-10, 10589 Berlin, Germany
| |
Collapse
|
5
|
Barazorda-Ccahuana HL, Cárcamo Rodriguez EG, Centeno-Lopez A, Paco-Chipana M, Goyzueta-Mamani LD, Chavez-Fumagalli MA. Identification of compounds from natural Peruvian sources as potential inhibitors of SARS-CoV-2 Mpro mutations by virtual screening and computational simulations. F1000Res 2024; 13:246. [PMID: 39583212 PMCID: PMC11585855 DOI: 10.12688/f1000research.143633.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/19/2024] [Indexed: 11/26/2024] Open
Abstract
Background Although the COVID-19 pandemic has diminished in intensity, the virus continues to circulate globally. The SARS-CoV-2 main protease (Mpro) is a key enzyme in the life cycle of the virus, making it important for the development of treatments against future variants of the virus. In this work, Peruvian natural compounds were evaluated against different mutations of the SARS-CoV-2 Mpro. Methods In silico techniques such as virtual screening, all-atom molecular dynamics simulations, and energy estimation analysis were applied. Results Of the tested compounds by virtual screening, rutin was identified as the best binding agent against the different proposed Mpro mutations. In addition, computational simulations and energy estimation analysis demonstrated the high structural and energetic stability between the Mpro-rutin systems. Conclusions Overall, our study identified rutin as the most promising compound with a strong affinity for various Mpro mutations, potentially playing a key role in the development of new treatments for emerging viral variants.
Collapse
Affiliation(s)
- Haruna Luz Barazorda-Ccahuana
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
| | - Eymi Gladys Cárcamo Rodriguez
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
- Facultad de Ciencias Farmaceuticas, Bioquímicas y Biotecnológicas, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
| | - Angela Emperatriz Centeno-Lopez
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
- Facultad de Ciencias Farmaceuticas, Bioquímicas y Biotecnológicas, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
| | - Margot Paco-Chipana
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
| | - Luis Daniel Goyzueta-Mamani
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
- Sustainable Innovative Biomaterials, Le Qara Research Center, Arequipa, Peru
| | - Miguel Angel Chavez-Fumagalli
- Computational Biology and Chemistry Research Group, Vicerrectorado de Investigación, Universidad Catolica de Santa Maria de Arequipa, Pedro Vilcapaza, Arequipa, 04000, Peru
| |
Collapse
|
6
|
Ntie-Kang F, Eni DB, Telukunta KK, Osamor VC, Egieyeh SA, Duran-Frigola M, Mishra P, Shadrack DM, Paul L, Musyoka TM, Blin K, Farid MM, Chen Y, Djogang LK, Betow JY, Ibezim A, Joshi D, Edwin AT, Chama MA, Ongagna JM, Kemdoum Sinda PV, Metuge JA, Bekono BD, Isa MA, Medina-Franco JL, Weber T, Dorrestein PC, Janezic D, Bishop ÖT, Ludwig-Müller J. The workshops on computational applications in secondary metabolite discovery (CAiSMD). PHYSICAL SCIENCES REVIEWS 2024; 9:3289-3304. [PMID: 39478877 PMCID: PMC11519840 DOI: 10.1515/psr-2024-0015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 02/03/2024] [Indexed: 11/02/2024]
Abstract
We report the outcomes of the second session of the free online open-access workshop "Computational Applications in Secondary Metabolite Discovery (CAiSMD) 2022" that took place from 09 to 11 March 2022. The first session was held from 08 to 10 March 2021 and drew the attention of many early career scientists from academia and industry. The 23 invited speakers of this year's workshop also came from academia and industry and 222 registered participants from five continents (Africa, Asia, Europe, South, and North America) took part in the workshop. The workshop highlighted the potential applications of computational methodologies in the search for secondary metabolites or natural products as drug candidates and drug leads. For three days, the participants of this online workshop discussed modern computer-based approaches for exploring NP discovery in the "omics" age. The invited experts gave keynote lectures, trained participants in hands-on sessions, and held round table discussions. These were followed by oral presentations during which much interaction between the speakers and the audience was observed. Selected applicants (early-career scientists) were offered the opportunity to give oral presentations (15 min) upon submission of an abstract. The final program available on the workshop website (https://indiayouth.info/index.php/caismd) comprised three keynote lectures, 14 oral presentations, two round table discussions, and four hands-on sessions. This meeting report also references internet resources for computational biology around secondary metabolites that are of use outside of the workshop areas and will constitute a long-term valuable source for the community.
Collapse
Affiliation(s)
- Fidele Ntie-Kang
- Center for Drug Discovery, University of Buea, P. O. Box 63Buea, Cameroon
- Department of Chemistry, University of Buea, P. O. Box 63Buea, Cameroon
- Institute of Pharmacy, Martin-Luther University of Halle-Wittenberg, Kurt-Mothes-Str. 3, 06120Halle, Germany
| | - Donatus B. Eni
- Center for Drug Discovery, University of Buea, P. O. Box 63Buea, Cameroon
- Department of Chemistry, University of Buea, P. O. Box 63Buea, Cameroon
| | - Kiran K. Telukunta
- Tarunavadaanenasaha Muktbharatonnayana Samstha Foundation, Hyderabad, India
| | | | - Samuel A. Egieyeh
- School of Pharmacy, University of the Western Cape, Cape Town, 7535South Africa
- South African Medical Research Council Bioinformatics Unit, South African National Bioinformatics Institute, University of the Western Cape, Cape Town7535, South Africa
| | | | - Pankaj Mishra
- Department of Education, Uresearcher Growth Labs Private Limited, 2/44-45, Wazir Hasan Road, Lucknow, Uttar Pradesh226001, India
| | - Daniel M. Shadrack
- Department of Chemistry, St. John’s University of Tanzania, P. O. Box 47Dodoma, Tanzania
| | - Lucas Paul
- Department of Chemistry, Dar es Salaam University College of Education (DUCE), P. O. Box 2329Dar es Salaam, Tanzania
| | - Thommas M. Musyoka
- Department of Biochemistry, Microbiology and Biotechnology, Kenyatta University, Nairobi, Kenya
| | - Kai Blin
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Mai M. Farid
- Department of Phytochemistry and Plant Systematics, Pharmaceutical and Drug Industries Research Institute, National Research Center, Cairo, Egypt
| | - Ya Chen
- Department of Pharmaceutical Sciences, Division of Pharmaceutical Chemistry, Faculty of Life Sciences, University of Vienna, 1090Vienna, Austria
| | | | - Jude Y. Betow
- Center for Drug Discovery, University of Buea, P. O. Box 63Buea, Cameroon
- Department of Chemistry, University of Buea, P. O. Box 63Buea, Cameroon
| | - Akachukwu Ibezim
- Department of Pharmaceutical and Medicinal Chemistry, Faculty of Pharmaceutical Sciences, University of Nigeria, Nsukka, Nigeria
| | - Darshana Joshi
- Tarunavadaanenasaha Muktbharatonnayana Samstha Foundation, Hyderabad, India
| | - Alanis T. Edwin
- Tarunavadaanenasaha Muktbharatonnayana Samstha Foundation, Hyderabad, India
| | - Mary A. Chama
- Department of Chemistry, School of Physical and Mathematical Sciences, University of Ghana, Accra, Ghana
| | | | | | - Jonathan A. Metuge
- Department of Natural Resources and Environmental Sciences, Alabama A & M University, Huntsville, USA
| | - Boris D. Bekono
- Department of Physics, Ecole Normale Supérieure, University of Yaoundé I, BP. 47, Yaoundé, Cameroon
| | - Mustafa A. Isa
- Bioinformatics and Computational Biology Lab, Department of Microbiology, Faculty of Sciences, University of Maiduguri, P.M.B. 1069, Maiduguri, Borno State, Nigeria
- School of Pharmacy, Taipei Medical University, Taipei, Taiwan
| | - José L. Medina-Franco
- DIFACQUIM Research Group, Department of Pharmacy, School of Chemistry, Universidad Nacional Autónoma de Mexico, Mexico City04510, Mexico
| | - Tilmann Weber
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Pieter C. Dorrestein
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, 9500 Gilman Drive, MC 0751, La Jolla, CA92093-0751, USA
| | - Dusanka Janezic
- Faculty of Mathematics, Natural Sciences and Information Technologies, University of Primorska, Glagoljaška 8, SI-6000Koper, Slovenia
| | - Özlem Tastan Bishop
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, Makhanda, 6140, South Africa
| | - Jutta Ludwig-Müller
- Faculty of Biology, Technische Universität Dresden, Zellescher Weg 20b, 01062Dresden, Germany
| |
Collapse
|
7
|
Sarkar M, Madabhavi I. COVID-19 mutations: An overview. World J Methodol 2024; 14:89761. [PMID: 39310238 PMCID: PMC11230071 DOI: 10.5662/wjm.v14.i3.89761] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/07/2024] [Accepted: 04/17/2024] [Indexed: 06/25/2024] Open
Abstract
The severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) belongs to the genus Beta coronavirus and the family of Coronaviridae. It is a positive-sense, non-segmented single-strand RNA virus. Four common types of human coronaviruses circulate globally, particularly in the fall and winter seasons. They are responsible for 10%-30% of all mild upper respiratory tract infections in adults. These are 229E, NL63 of the Alfacoronaviridae family, OC43, and HKU1 of the Betacoronaviridae family. However, there are three highly pathogenic human coronaviruses: SARS-CoV-2, Middle East respiratory syndrome coronavirus, and the latest pandemic caused by the SARS-CoV-2 infection. All viruses, including SARS-CoV-2, have the inherent tendency to evolve. SARS-CoV-2 is still evolving in humans. Additionally, due to the development of herd immunity, prior infection, use of medication, vaccination, and antibodies, the viruses are facing immune pressure. During the replication process and due to immune pressure, the virus may undergo mutations. Several SARS-CoV-2 variants, including the variants of concern (VOCs), such as B.1.1.7 (Alpha), B.1.351 (Beta), B.1.617/B.1.617.2 (Delta), P.1 (Gamma), and B.1.1.529 (Omicron) have been reported from various parts of the world. These VOCs contain several important mutations; some of them are on the spike proteins. These mutations may lead to enhanced infectivity, transmissibility, and decreased neutralization efficacy by monoclonal antibodies, convalescent sera, or vaccines. Mutations may also lead to a failure of detection by molecular diagnostic tests, leading to a delayed diagnosis, increased community spread, and delayed treatment. We searched PubMed, EMBASE, Covariant, the Stanford variant Database, and the CINAHL from December 2019 to February 2023 using the following search terms: VOC, SARS-CoV-2, Omicron, mutations in SARS-CoV-2, etc. This review discusses the various mutations and their impact on infectivity, transmissibility, and neutralization efficacy.
Collapse
Affiliation(s)
- Malay Sarkar
- Department of Pulmonary Medicine, Indira Gandhi Medical College, Shimla 171001, Himachal Pradesh, India
| | - Irappa Madabhavi
- Department of Medical and Pediatric Oncology and Hematology, J N Medical College, and KAHER, Belagavi, Karnataka 590010, India
- Department of Medical and Pediatric Oncology and Hematology, Kerudi Cancer Hospital, Bagalkot, Karnataka 587103, India
| |
Collapse
|
8
|
Das A, Pathak S, Premkumar M, Sarpparajan CV, Balaji ER, Duttaroy AK, Banerjee A. A brief overview of SARS-CoV-2 infection and its management strategies: a recent update. Mol Cell Biochem 2024; 479:2195-2215. [PMID: 37742314 PMCID: PMC11371863 DOI: 10.1007/s11010-023-04848-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/02/2023] [Indexed: 09/26/2023]
Abstract
The COVID-19 pandemic has become a global health crisis, inflicting substantial morbidity and mortality worldwide. A diverse range of symptoms, including fever, cough, dyspnea, and fatigue, characterizes COVID-19. A cytokine surge can exacerbate the disease's severity. This phenomenon involves an increased immune response, marked by the excessive release of inflammatory cytokines like IL-6, IL-8, TNF-α, and IFNγ, leading to tissue damage and organ dysfunction. Efforts to reduce the cytokine surge and its associated complications have garnered significant attention. Standardized management protocols have incorporated treatment strategies, with corticosteroids, chloroquine, and intravenous immunoglobulin taking the forefront. The recent therapeutic intervention has also assisted in novel strategies like repurposing existing medications and the utilization of in vitro drug screening methods to choose effective molecules against viral infections. Beyond acute management, the significance of comprehensive post-COVID-19 management strategies, like remedial measures including nutritional guidance, multidisciplinary care, and follow-up, has become increasingly evident. As the understanding of COVID-19 pathogenesis deepens, it is becoming increasingly evident that a tailored approach to therapy is imperative. This review focuses on effective treatment measures aimed at mitigating COVID-19 severity and highlights the significance of comprehensive COVID-19 management strategies that show promise in the battle against COVID-19.
Collapse
Affiliation(s)
- Alakesh Das
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Madhavi Premkumar
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Chitra Veena Sarpparajan
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Esther Raichel Balaji
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India
| | - Asim K Duttaroy
- Department of Nutrition, Faculty of Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway.
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute (CHRI), Chettinad Academy of Research and Education (CARE), Kelambakkam, Chennai, Tamil Nadu, 603103, India.
| |
Collapse
|
9
|
Chandy S, Kumar H, Pearl S, Basu S, M G, Sankar J, Manoharan A, Ramaiah S, Anbarasu A. Whole genome analysis reveals unique traits of SARS-CoV-2 in pediatric patients. Gene 2024; 919:148508. [PMID: 38670399 DOI: 10.1016/j.gene.2024.148508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/10/2024] [Accepted: 04/23/2024] [Indexed: 04/28/2024]
Abstract
Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) continues to challenge the global healthcare with emerging variants and higher infectivity as well as morbidities. This study investigated potential age-related variations through genomic characterization of the virus under common clinical settings. A cohort comprising 71 SARS-CoV-2 strains from both infected infants and accompanying adults, diagnosed via RT-PCR at a tertiary pediatric hospital and research center, underwent Illumina paired-end sequencing. The subsequent analysis involved standard genomic screening, phylogeny construction, and mutational analyses. The analyzed SARSCoV- 2 strains were compared with globally circulating variants. The overall distribution revealed 67.61 % Delta, 25.7 % Omicron, and 1 % either Kappa or Alpha variants. In 2021, Delta predominated at ∼ 94 %, with Alpha/Kappa accounting for around 5 %. However, in 2022, over 94 % of the samples were Omicron variants, signifying a substantial shift from Delta dominance. Delta variants constituted 69.5 % of infections in adults and 78.5 % in infants, while Omicron variants were responsible for 31 % of cases in infants and 18 % in adults. The Spike region harbored the majority of mutations, with T19R being the most prevalent mutation in the Delta lineage. Notably, the frequencies of this mutation varied between infants and adults. In Omicron samples, G142D emerged as the most prevalent mutation. Our dataset predominantly featured clade 21A and lineage B.1.617.2. This study underscores the differential clinical presentations and genomic characteristics of SARS-CoV-2 in pediatric patients and accompanying adults. Understanding the dynamic evolution of the SARS- CoV-2 in both pediatric and adults can help in strengthening prophylactic measures.
Collapse
Affiliation(s)
- Sara Chandy
- The CHILDS Trust Medical Research Foundation (CTMRF), 12-A, Nageswara Road, Nungambakkam, Chennai 600034, Tamil Nadu, India
| | - Hithesh Kumar
- Department of Bio-Sciences, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, India; Medical and Biological Computing Laboratory, SBST, VIT, Vellore 632014, India
| | - Sara Pearl
- Medical and Biological Computing Laboratory, SBST, VIT, Vellore 632014, India; Department of Integrative Biology, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, India
| | - Soumya Basu
- Medical and Biological Computing Laboratory, SBST, VIT, Vellore 632014, India; Department of Biotechnology, NIST University, Berhampore 761008, India
| | - Gurumoorthy M
- The CHILDS Trust Medical Research Foundation (CTMRF), 12-A, Nageswara Road, Nungambakkam, Chennai 600034, Tamil Nadu, India
| | - Janani Sankar
- The CHILDS Trust Medical Research Foundation (CTMRF), 12-A, Nageswara Road, Nungambakkam, Chennai 600034, Tamil Nadu, India
| | - Anand Manoharan
- Kanchi Kamakoti CHILDS Trust Hospital (KKCTH), 12-A, Nageswara Road, Nungambakkam, Chennai 600034, Tamil Nadu, India
| | - Sudha Ramaiah
- Department of Bio-Sciences, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, India; Medical and Biological Computing Laboratory, SBST, VIT, Vellore 632014, India
| | - Anand Anbarasu
- Medical and Biological Computing Laboratory, SBST, VIT, Vellore 632014, India; Department of Biotechnology, School of Biosciences and Technology (SBST), Vellore Institute of Technology (VIT), Vellore 632014, India.
| |
Collapse
|
10
|
El-Assaad AM, Hamieh T. SARS-CoV-2: Prediction of critical ionic amino acid mutations. Comput Biol Med 2024; 178:108688. [PMID: 38870723 DOI: 10.1016/j.compbiomed.2024.108688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 05/26/2024] [Accepted: 06/01/2024] [Indexed: 06/15/2024]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), that caused coronavirus disease 2019 (COVID-19), has been studied thoroughly, and several variants are revealed across the world with their corresponding mutations. Studies and vaccines development focus on the genetic mutations of the S protein due to its vital role in allowing the virus attach and fuse with the membrane of a host cell. In this perspective, we study the effects of all ionic amino acid mutations of the SARS-CoV-2 viral spike protein S1 when bound to Antibody CC12.1 within the SARS-CoV-2:CC12.1 complex model. Binding free energy calculations between SARS-CoV-2 and antibody CC12.1 are based on the Analysis of Electrostatic Similarities of Proteins (AESOP) framework, where the electrostatic potentials are calculated using Adaptive Poisson-Boltzmann Solver (APBS). The atomic radii and charges that feed into the APBS calculations are calculated using the PDB2PQR software. Our results are the first to propose in silico potential life-threatening mutations of SARS-CoV-2 beyond the present mutations found in the five common variants worldwide. We find each of the following mutations: K378A, R408A, K424A, R454A, R457A, K458A, and K462A, to play significant roles in the binding to Antibody CC12.1, since they are turned into strong inhibitors on both chains of the S1 protein, whereas the mutations D405A, D420A, and D427A, show to play important roles in this binding, as they are turned into mild inhibitors on both chains of the S1 protein.
Collapse
Affiliation(s)
- Atlal M El-Assaad
- Department of Electrical Engineering & Computer Science, University of Toledo (UT), Toledo OH 43606, USA; Department of Computer Science, Lebanese International University (LIU), Bekaa, Lebanon.
| | - Tayssir Hamieh
- Faculty of Science and Engineering, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands; Laboratory of Materials, Catalysis, Environment and Analytical Methods (MCEMA), Faculty of Sciences, Lebanese University, Hadath, Lebanon.
| |
Collapse
|
11
|
Perovic V, Glisic S, Veljkovic M, Paessler S, Veljkovic V. In Silico Exploration of CD200 as a Therapeutic Target for COVID-19. Microorganisms 2024; 12:1185. [PMID: 38930566 PMCID: PMC11205781 DOI: 10.3390/microorganisms12061185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 06/06/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024] Open
Abstract
SARS-CoV-2, the pathogen causing COVID-19, continues to pose a significant threat to public health and has had major economic implications. Developing safe and effective vaccines and therapies offers a path forward for overcoming the COVID-19 pandemic. The presented study, performed by using the informational spectrum method (ISM), representing an electronic biology-based tool for analysis of protein-protein interactions, identified the highly conserved region of spike protein (SP) from SARS-CoV-2 virus, which is essential for recognition and targeting between the virus and its protein interactors on the target cells. This domain is suggested as a promising target for the drug therapy and vaccines, which could be effective against all currently circulating variants of SARS-CoV-2 viruses. The analysis of the virus/host interaction, performed by the ISM, also revealed OX-2 membrane glycoprotein (CD200) as a possible interactor of SP, which could serve as a novel therapeutic target for COVID-19 disease.
Collapse
Affiliation(s)
- Vladimir Perovic
- Laboratory for Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences VINCA, University of Belgrade, 11001 Belgrade, Serbia;
| | - Sanja Glisic
- Laboratory for Bioinformatics and Computational Chemistry, Institute of Nuclear Sciences VINCA, University of Belgrade, 11001 Belgrade, Serbia;
| | - Milena Veljkovic
- Department of Clinical Laboratory Medicine, Hospital for Cerebrovascular Diseases Sveti Sava, 11000 Belgrade, Serbia
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | |
Collapse
|
12
|
Edner NM, Houghton LP, Ntavli E, Rees-Spear C, Petersone L, Wang C, Fabri A, Elfaki Y, Rueda Gonzalez A, Brown R, Kisand K, Peterson P, McCoy LE, Walker LSK. TIGIT +Tfh show poor B-helper function and negatively correlate with SARS-CoV-2 antibody titre. Front Immunol 2024; 15:1395684. [PMID: 38868776 PMCID: PMC11167088 DOI: 10.3389/fimmu.2024.1395684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/15/2024] [Indexed: 06/14/2024] Open
Abstract
Circulating follicular helper T cells (cTfh) can show phenotypic alterations in disease settings, including in the context of tissue-damaging autoimmune or anti-viral responses. Using severe COVID-19 as a paradigm of immune dysregulation, we have explored how cTfh phenotype relates to the titre and quality of antibody responses. Severe disease was associated with higher titres of neutralising S1 IgG and evidence of increased T cell activation. ICOS, CD38 and HLA-DR expressing cTfh correlated with serum S1 IgG titres and neutralising strength, and interestingly expression of TIGIT by cTfh showed a negative correlation. TIGIT+cTfh expressed increased IFNγ and decreased IL-17 compared to their TIGIT-cTfh counterparts, and showed reduced capacity to help B cells in vitro. Additionally, TIGIT+cTfh expressed lower levels of CD40L than TIGIT-cTfh, providing a potential explanation for their poor B-helper function. These data identify phenotypic changes in polyclonal cTfh that correlate with specific antibody responses and reveal TIGIT as a marker of cTfh with altered function.
Collapse
Affiliation(s)
- Natalie M. Edner
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Luke P. Houghton
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Elisavet Ntavli
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Chloe Rees-Spear
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Lina Petersone
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Chunjing Wang
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Astrid Fabri
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Yassin Elfaki
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Andrea Rueda Gonzalez
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Rachel Brown
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
- Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Kai Kisand
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Pärt Peterson
- Institute of Biomedicine and Translational Medicine, University of Tartu, Tartu, Estonia
| | - Laura E. McCoy
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| | - Lucy S. K. Walker
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, United Kingdom
| |
Collapse
|
13
|
Özçelik C, Araz CZ, Yılmaz Ö, Gülyüz S, Özdamar P, Salmanlı E, Özkul A, Şeker UÖŞ. Screening Peptide Drug Candidates To Neutralize Whole Viral Agents: A Case Study with Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). ACS Pharmacol Transl Sci 2024; 7:1032-1042. [PMID: 38633598 PMCID: PMC11020059 DOI: 10.1021/acsptsci.3c00317] [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: 11/08/2023] [Revised: 03/03/2024] [Accepted: 03/06/2024] [Indexed: 04/19/2024]
Abstract
The COVID-19 pandemic revealed the need for therapeutic and pharmaceutical molecule development in a short time with different approaches. Although boosting immunological memory by vaccination was the quickest and robust strategy, still medication is required for the immediate treatment of a patient. A popular approach is the mining of new therapeutic molecules. Peptide-based drug candidates are also becoming a popular avenue. To target whole pathogenic viral agents, peptide libraries can be employed. With this motivation, we have used the 12mer M13 phage display library for selecting SARS-CoV-2 targeting peptides as potential neutralizing molecules to prevent viral infections. Panning was applied with four iterative cycles to select SARS-CoV-2 targeting phage particles displaying 12-amino acid-long peptides. Randomly selected peptide sequences were synthesized by a solid-state peptide synthesis method. Later, selected peptides were analyzed by the quartz crystal microbalance method to characterize their molecular interaction with SARS-CoV-2's S protein. Finally, the neutralization activity of the selected peptides was probed with an in-house enzyme-linked immunosorbent assay. The results showed that scpep3, scpep8, and scpep10 peptides have both binding and neutralizing capacity for S1 protein as a candidate for therapeutic molecule. The results of this study have a translational potential with future in vivo and human studies.
Collapse
Affiliation(s)
- Cemile
Elif Özçelik
- UNAM—Institute
of Materials Science and Nanotechnology, Bilkent University, Ankara 06800, Turkey
| | - Cemre Zekiye Araz
- Synbiotik
Biotechnology and Biomedical Technology Bilkent Kümeevler, Çankaya, Ankara 06800, Turkey
| | - Özgür Yılmaz
- Material
Technologies, Marmara Research Center, TUBITAK, Gebze, Kocaeli 41470, Turkey
| | - Sevgi Gülyüz
- Material
Technologies, Marmara Research Center, TUBITAK, Gebze, Kocaeli 41470, Turkey
| | - Pınar Özdamar
- Faculty of Veterinary Medicine, Department of Virology, Graduate School of Health
Sciences, Department of Virology, Ankara
University, Ankara 06110, Turkey
| | - Ezgi Salmanlı
- Faculty of Veterinary Medicine, Department of Virology, Graduate School of Health
Sciences, Department of Virology, Ankara
University, Ankara 06110, Turkey
| | - Aykut Özkul
- Faculty of Veterinary Medicine, Department of Virology, Graduate School of Health
Sciences, Department of Virology, Ankara
University, Ankara 06110, Turkey
| | - Urartu Özgür Şafak Şeker
- UNAM—Institute
of Materials Science and Nanotechnology, Bilkent University, Ankara 06800, Turkey
- Interdisciplinary
Program in Neuroscience, Bilkent University, Ankara 06800, Turkey
| |
Collapse
|
14
|
Pegg CL, Modhiran N, Parry RH, Liang B, Amarilla AA, Khromykh AA, Burr L, Young PR, Chappell K, Schulz BL, Watterson D. The role of N-glycosylation in spike antigenicity for the SARS-CoV-2 gamma variant. Glycobiology 2024; 34:cwad097. [PMID: 38048640 PMCID: PMC10969516 DOI: 10.1093/glycob/cwad097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 11/10/2023] [Accepted: 11/28/2023] [Indexed: 12/06/2023] Open
Abstract
The emergence of SARS-CoV-2 variants alters the efficacy of existing immunity towards the viral spike protein, whether acquired from infection or vaccination. Mutations that impact N-glycosylation of spike may be particularly important in influencing antigenicity, but their consequences are difficult to predict. Here, we compare the glycosylation profiles and antigenicity of recombinant viral spike of ancestral Wu-1 and the Gamma strain, which has two additional N-glycosylation sites due to amino acid substitutions in the N-terminal domain (NTD). We found that a mutation at residue 20 from threonine to asparagine within the NTD caused the loss of NTD-specific antibody COVA2-17 binding. Glycan site-occupancy analyses revealed that the mutation resulted in N-glycosylation switching to the new sequon at N20 from the native N17 site. Site-specific glycosylation profiles demonstrated distinct glycoform differences between Wu-1, Gamma, and selected NTD variant spike proteins, but these did not affect antibody binding. Finally, we evaluated the specificity of spike proteins against convalescent COVID-19 sera and found reduced cross-reactivity against some mutants, but not Gamma spike compared to Wuhan spike. Our results illustrate the impact of viral divergence on spike glycosylation and SARS-CoV-2 antibody binding profiles.
Collapse
Affiliation(s)
- Cassandra L Pegg
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, Building 75, Corner College Road and Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Rhys H Parry
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Benjamin Liang
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Alberto A Amarilla
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
| | - Alexander A Khromykh
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland 4072 and 4006, Australia
| | - Lucy Burr
- Department of Respiratory Medicine, Mater Health Services, Raymond Terrace, South Brisbane, Queensland 4101, Australia
| | - Paul R Young
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, Building 75, Corner College Road and Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland 4072 and 4006, Australia
| | - Keith Chappell
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, Building 75, Corner College Road and Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland 4072 and 4006, Australia
| | - Benjamin L Schulz
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland 4072 and 4006, Australia
| | - Daniel Watterson
- School of Chemistry and Molecular Bioscience, Chemistry Building 68, Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Institute for Bioengineering and Nanotechnology, Building 75, Corner College Road and Cooper Road, University of Queensland, St Lucia, Queensland 4072, Australia
- Australian Infectious Disease Research Centre, Global Virus Network Centre of Excellence, Brisbane, Queensland 4072 and 4006, Australia
| |
Collapse
|
15
|
Han G, Deng W, Lyu Q, Ma Q, Qiao L. Multiplexed discrimination of SARS-CoV-2 variants via duplex-specific nuclease combined MALDI-TOF MS. Anal Bioanal Chem 2024; 416:1833-1842. [PMID: 38367041 DOI: 10.1007/s00216-024-05202-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/19/2024]
Abstract
The frequent mutations in SARS-CoV-2 significantly increase the virus's pathogenicity and transmissibility while also diminishing the effectiveness of vaccines. Consequently, assays capable of rapidly and simultaneously identifying multiple SARS-CoV-2 variants are essential for large-scale applications that aim to monitor the evolution of the virus. In this work, we propose a method combining duplex-specific nuclease (DSN)-assisted cyclic amplification with matrix-assisted laser desorption/ionization time-of-flight mass spectrometry (MALDI-TOF MS) detection, enabling the simultaneous identification of multiple SARS-CoV-2 variants at high-throughput. Due to the high specificity of DSN, single-base mutations can be resolved by the method. With ultra-sensitive detection by MALDI-TOF MS, a limit of detection of 100 pM viral RNA fragment was demonstrated. The assay was used for simultaneous identification and typing of SARS-CoV-2 Alpha, Beta, and Delta variants. The whole assay can be accomplished within 3 h, and the amplification is performed under constant temperature, making the technique simple in operation and efficient. It is also feasible to extend the technique to the detection of many other variants of the virus. We expect that the method can add value to the rapid screening of viral variants and can play an important role in pandemic control.
Collapse
Affiliation(s)
- Guobin Han
- Department of Chemistry, and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China
| | - Wenchan Deng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, Zhejiang, China
| | - Qian Lyu
- Bioyong Technologics Inc, Beijing, 100176, China
| | - Qingwei Ma
- Bioyong Technologics Inc, Beijing, 100176, China
| | - Liang Qiao
- Department of Chemistry, and Shanghai Stomatological Hospital, Fudan University, Shanghai, 200000, China.
| |
Collapse
|
16
|
Ahmed N, Athavale A, Tripathi AH, Subramaniam A, Upadhyay SK, Pandey AK, Rai RC, Awasthi A. To be remembered: B cell memory response against SARS-CoV-2 and its variants in vaccinated and unvaccinated individuals. Scand J Immunol 2024; 99:e13345. [PMID: 38441373 DOI: 10.1111/sji.13345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 03/07/2024]
Abstract
COVID-19 disease has plagued the world economy and affected the overall well-being and life of most of the people. Natural infection as well as vaccination leads to the development of an immune response against the pathogen. This involves the production of antibodies, which can neutralize the virus during future challenges. In addition, the development of cellular immune memory with memory B and T cells provides long-lasting protection. The longevity of the immune response has been a subject of intensive research in this field. The extent of immunity conferred by different forms of vaccination or natural infections remained debatable for long. Hence, understanding the effectiveness of these responses among different groups of people can assist government organizations in making informed policy decisions. In this article, based on the publicly available data, we have reviewed the memory response generated by some of the vaccines against SARS-CoV-2 and its variants, particularly B cell memory in different groups of individuals.
Collapse
Affiliation(s)
- Nafees Ahmed
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Atharv Athavale
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Ankita H Tripathi
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, India
| | - Adarsh Subramaniam
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Santosh K Upadhyay
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, India
| | | | - Ramesh Chandra Rai
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Amit Awasthi
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
17
|
Keller MW, Keong LM, Rambo-Martin BL, Hassell N, Lacek KA, Wilson MM, Kirby MK, Liddell J, Owuor DC, Sheth M, Madden J, Lee JS, Kondor RJ, Wentworth DE, Barnes JR. Targeted amplification and genetic sequencing of the severe acute respiratory syndrome coronavirus 2 surface glycoprotein. Microbiol Spectr 2024; 12:e0298223. [PMID: 38084972 PMCID: PMC10783008 DOI: 10.1128/spectrum.02982-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/09/2023] [Indexed: 01/13/2024] Open
Abstract
IMPORTANCE The COVID-19 pandemic was accompanied by an unprecedented surveillance effort. The resulting data were and will continue to be critical for surveillance and control of SARS-CoV-2. However, some genomic surveillance methods experienced challenges as the virus evolved, resulting in incomplete and poor quality data. Complete and quality coverage, especially of the S-gene, is important for supporting the selection of vaccine candidates. As such, we developed a robust method to target the S-gene for amplification and sequencing. By focusing on the S-gene and imposing strict coverage and quality metrics, we hope to increase the quality of surveillance data for this continually evolving gene. Our technique is currently being deployed globally to partner laboratories, and public health representatives from 79 countries have received hands-on training and support. Expanding access to quality surveillance methods will undoubtedly lead to earlier detection of novel variants and better inform vaccine strain selection.
Collapse
Affiliation(s)
- Matthew W. Keller
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Lisa M. Keong
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Benjamin L. Rambo-Martin
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Norman Hassell
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Kristine A. Lacek
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Malania M. Wilson
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Marie K. Kirby
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Jimma Liddell
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - D. Collins Owuor
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - Mili Sheth
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Joseph Madden
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Justin S. Lee
- Biotechnology Core Facility Branch, Division of Scientific Resources, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Rebecca J. Kondor
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - David E. Wentworth
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| | - John R. Barnes
- Influenza Division, National Center for Immunization and Respiratory Diseases (NCIRD), Centers for Disease Control and Prevention (CDC), Atlanta, Georgia, USA
| |
Collapse
|
18
|
Plans-Rubió P. Effectiveness of Adapted COVID-19 Vaccines and Ability to Establish Herd Immunity against Omicron BA.1 and BA4-5 Variants of SARS-CoV-2. Vaccines (Basel) 2023; 11:1836. [PMID: 38140240 PMCID: PMC10747774 DOI: 10.3390/vaccines11121836] [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: 10/28/2023] [Revised: 12/06/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
The emergence of novel SARS-CoV-2 variants has raised concerns about the ability of COVID-19 vaccination programs to establish adequate herd immunity levels in the population. This study assessed the effectiveness of adapted vaccines in preventing SARS-CoV-2 infection and the ability of the adapted vaccines to establish herd immunity against emerging Omicron variants. A systematic literature review was conducted to estimate the absolute vaccine effectiveness (aVE) in preventing SARS-CoV-2 infection using adapted vaccines targeting Omicron variants. The ability of the adapted vaccines to establish herd immunity was assessed by taking into account the following factors: aVE, Ro values of SARS-CoV-2 and the use of non-pharmacological interventions (NPIs). This study found meta-analysis-based aVEs in preventing severe disease and SARS-CoV-2 infection of 56-60% and 36-39%, respectively. Adapted vaccines could not establish herd immunity against the Omicron BA.1 and BA.4-5 variants without using non-pharmacological interventions (NPIs). The adapted vaccines could establish herd immunity only by achieving >80% vaccination coverage, using NPIs with greater effectiveness and when 20-30% of individuals were already protected against SARS-CoV-2 in the population. New adapted COVID-19 vaccines with greater effectiveness in preventing SARS-CoV-2 infection must be developed to increase herd immunity levels against emerging SARS-CoV-2 variants in the population.
Collapse
Affiliation(s)
- Pedro Plans-Rubió
- Public Health Agency of Catalonia, Department of Health of Catalonia, 08005 Barcelona, Spain;
- Ciber of Epidemiology and Public Health (CIBERESP), 28028 Madrid, Spain
| |
Collapse
|
19
|
Sheehan J, Ardizzone CM, Khanna M, Trauth AJ, Hagensee ME, Ramsay AJ. Dynamics of Serum-Neutralizing Antibody Responses in Vaccinees through Multiple Doses of the BNT162b2 Vaccine. Vaccines (Basel) 2023; 11:1720. [PMID: 38006052 PMCID: PMC10675463 DOI: 10.3390/vaccines11111720] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 11/08/2023] [Accepted: 11/11/2023] [Indexed: 11/26/2023] Open
Abstract
SARS-CoV-2 mRNA vaccines are administered as effective prophylactic measures for reducing virus transmission rates and disease severity. To enhance the durability of post-vaccination immunity and combat SARS-CoV-2 variants, boosters have been administered to two-dose vaccinees. However, long-term humoral responses following booster vaccination are not well characterized. A 16-member cohort of healthy SARS-CoV-2 naïve participants were enrolled in this study during a three-dose BNT162b2 vaccine series. Serum samples were collected from vaccinees over 420 days and screened for antigen (Ag)-specific antibody titers, IgG subclass distribution, and neutralizing antibody (nAb) responses. Vaccine boosting restored peak Ag-specific titers with sustained α-RBD IgG and IgA antibody responses when measured at six months post-boost. RBD- and spike-specific IgG4 antibody levels were markedly elevated in three-dose but not two-dose immune sera. Although strong neutralization responses were detected in two- and three-dose vaccine sera, these rapidly decayed to pre-immune levels by four and six months, respectively. While boosters enhanced serum IgG Ab reactivity and nAb responses against variant strains, all variants tested showed resistance to two- and three-dose immune sera. Our data reflect the poor durability of vaccine-induced nAb responses which are a strong predictor of protection from symptomatic SARS-CoV-2 infection. The induction of IgG4-switched humoral responses may permit extended viral persistence via the downregulation of Fc-mediated effector functions.
Collapse
Affiliation(s)
- Jared Sheehan
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Caleb M. Ardizzone
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Mayank Khanna
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Amber J. Trauth
- Department of Internal Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Michael E. Hagensee
- Department of Internal Medicine, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Alistair J. Ramsay
- Department of Microbiology, Immunology, and Parasitology, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
20
|
Diniz MO, Maini MK, Swadling L. T cell control of SARS-CoV-2: When, which, and where? Semin Immunol 2023; 70:101828. [PMID: 37651850 DOI: 10.1016/j.smim.2023.101828] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 08/13/2023] [Indexed: 09/02/2023]
Abstract
Efficient immune protection against viruses such as SARS-CoV-2 requires the coordinated activity of innate immunity, B and T cells. Accumulating data point to a critical role for T cells not only in the clearance of established infection, but also for aborting viral replication independently of humoral immunity. Here we review the evidence supporting the contribution of antiviral T cells and consider which of their qualitative features favour efficient control of infection. We highlight how studies of SARS-CoV-2 and other coronaviridae in animals and humans have provided important lessons on the optimal timing (When), functionality and specificity (Which), and location (Where) of antiviral T cells. We discuss the clinical implications, particularly for the development of next-generation vaccines, and emphasise areas requiring further study.
Collapse
Affiliation(s)
- Mariana O Diniz
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK
| | - Mala K Maini
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK.
| | - Leo Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, Pears Building, London WC1E 6BT, UK.
| |
Collapse
|
21
|
Cozzini P, Agosta F, Dolcetti G, Dal Palù A. A Computational Workflow to Predict Biological Target Mutations: The Spike Glycoprotein Case Study. Molecules 2023; 28:7082. [PMID: 37894561 PMCID: PMC10609230 DOI: 10.3390/molecules28207082] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/03/2023] [Accepted: 10/09/2023] [Indexed: 10/29/2023] Open
Abstract
The biological target identification process, a pivotal phase in the drug discovery workflow, becomes particularly challenging when mutations affect proteins' mechanisms of action. COVID-19 Spike glycoprotein mutations are known to modify the affinity toward the human angiotensin-converting enzyme ACE2 and several antibodies, compromising their neutralizing effect. Predicting new possible mutations would be an efficient way to develop specific and efficacious drugs, vaccines, and antibodies. In this work, we developed and applied a computational procedure, combining constrained logic programming and careful structural analysis based on the Structural Activity Relationship (SAR) approach, to predict and determine the structure and behavior of new future mutants. "Mutations rules" that would track statistical and functional types of substitutions for each residue or combination of residues were extracted from the GISAID database and used to define constraints for our software, having control of the process step by step. A careful molecular dynamics analysis of the predicted mutated structures was carried out after an energy evaluation of the intermolecular and intramolecular interactions using the HINT (Hydrophatic INTeraction) force field. Our approach successfully predicted, among others, known Spike mutants.
Collapse
Affiliation(s)
- Pietro Cozzini
- Molecular Modeling Lab, Food and Drug Department, University of Parma, Parco Area delle Scienze 17/A, 43121 Parma, Italy;
| | - Federica Agosta
- Molecular Modeling Lab, Food and Drug Department, University of Parma, Parco Area delle Scienze 17/A, 43121 Parma, Italy;
| | - Greta Dolcetti
- Department of Mathematical, Physical and Computer Sciences, University of Parma, 43121 Parma, Italy; (G.D.); (A.D.P.)
| | - Alessandro Dal Palù
- Department of Mathematical, Physical and Computer Sciences, University of Parma, 43121 Parma, Italy; (G.D.); (A.D.P.)
| |
Collapse
|
22
|
Upasani V, Townsend K, Wu MY, Carr EJ, Hobbs A, Dowgier G, Ragno M, Herman LS, Sharma S, Shah D, Lee SFK, Chauhan N, Glanville JM, Neave L, Hanson S, Ravichandran S, Tynan A, O’Sullivan M, Moreira F, Workman S, Symes A, Burns SO, Tadros S, Hart JCL, Beale RCL, Gandhi S, Wall EC, McCoy L, Lowe DM. Commercial Immunoglobulin Products Contain Neutralizing Antibodies Against Severe Acute Respiratory Syndrome Coronavirus 2 Spike Protein. Clin Infect Dis 2023; 77:950-960. [PMID: 37338118 PMCID: PMC10552578 DOI: 10.1093/cid/ciad368] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/05/2023] [Accepted: 06/16/2023] [Indexed: 06/21/2023] Open
Abstract
BACKGROUND Patients with antibody deficiency respond poorly to coronavirus disease 2019 (COVID-19) vaccination and are at risk of severe or prolonged infection. They are given long-term immunoglobulin replacement therapy (IRT) prepared from healthy donor plasma to confer passive immunity against infection. Following widespread COVID-19 vaccination alongside natural exposure, we hypothesized that immunoglobulin preparations will now contain neutralizing severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) spike antibodies, which confer protection against COVID-19 disease and may help to treat chronic infection. METHODS We evaluated anti-SARS-CoV-2 spike antibody in a cohort of patients before and after immunoglobulin infusion. Neutralizing capacity of patient samples and immunoglobulin products was assessed using in vitro pseudovirus and live-virus neutralization assays, the latter investigating multiple batches against current circulating Omicron variants. We describe the clinical course of 9 patients started on IRT during treatment of COVID-19. RESULTS In 35 individuals with antibody deficiency established on IRT, median anti-spike antibody titer increased from 2123 to 10 600 U/mL postinfusion, with corresponding increase in pseudovirus neutralization titers to levels comparable to healthy donors. Testing immunoglobulin products directly in the live-virus assay confirmed neutralization, including of BQ1.1 and XBB variants, but with variation between immunoglobulin products and batches.Initiation of IRT alongside remdesivir in patients with antibody deficiency and prolonged COVID-19 infection (median 189 days, maximum >900 days with an ancestral viral strain) resulted in clearance of SARS-CoV-2 at a median of 20 days. CONCLUSIONS Immunoglobulin preparations now contain neutralizing anti-SARS-CoV-2 antibodies that are transmitted to patients and help to treat COVID-19 in individuals with failure of humoral immunity.
Collapse
Affiliation(s)
- Vinit Upasani
- Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Katie Townsend
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Mary Y Wu
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
| | - Edward J Carr
- Francis Crick Institute, London, United Kingdom
- Department of Renal Medicine, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Agnieszka Hobbs
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
| | - Giulia Dowgier
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
| | - Martina Ragno
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
| | - Lou S Herman
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
| | - Sonal Sharma
- Department of Elderly Medicine, Barnet Hospital, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Devesh Shah
- Department of Elderly Medicine, Barnet Hospital, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Simon F K Lee
- Department of Infectious Diseases, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Neil Chauhan
- Department of Haematology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Julie M Glanville
- Department of Haematology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Lucy Neave
- Department of Haematology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Steven Hanson
- Department of Haematology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Sriram Ravichandran
- Department of Haematology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Aoife Tynan
- Department of Pharmacy, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Mary O’Sullivan
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Fernando Moreira
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Sarita Workman
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Andrew Symes
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Siobhan O Burns
- Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Susan Tadros
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| | - Jennifer C L Hart
- Department of Virology, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Rupert C L Beale
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
- Department of Renal Medicine, Royal Free London NHS Foundation Trust, London, United Kingdom
| | - Sonia Gandhi
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
- UCL Hospitals Biomedical Research Centre, London, United Kingdom
| | - Emma C Wall
- COVID Surveillance Unit, Francis Crick Institute, London, United Kingdom
- UCL Hospitals Biomedical Research Centre, London, United Kingdom
| | - Laura McCoy
- Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - David M Lowe
- Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
- Department of Clinical Immunology, Royal Free London National Health Service (NHS) Foundation Trust, London, United Kingdom
| |
Collapse
|
23
|
Liu Y, Ye Q. The Key Site Variation and Immune Challenges in SARS-CoV-2 Evolution. Vaccines (Basel) 2023; 11:1472. [PMID: 37766148 PMCID: PMC10537874 DOI: 10.3390/vaccines11091472] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/07/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a worldwide public health and economic threat, and virus variation amplifies the difficulty in epidemic prevention and control. The structure of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been studied extensively and is now well defined. The S protein is the most distinguishing feature in terms of infection and immunity, mediating virus entrance and inducing neutralizing antibodies. The S protein and its essential components are also the most promising target to develop vaccines and antibody-based drugs. Therefore, the key site mutation in the S gene is of high interest. Among them, RBD, NTD, and furin cleavage sites are the most mutable regions with the most mutation sites and the most serious consequences for SARS-CoV-2 biological characteristics, including infectivity, pathogenicity, natural immunity, vaccine efficacy, and antibody therapeutics. We are also aware that this outbreak may not be the last. Therefore, in this narrative review, we summarized viral variation and prevalence condition, discussed specific amino acid replacement and associated immune challenges and attempted to sum up some prevention and control strategies by reviewing the literature on previously published research about SARS-CoV-2 variation to assist in clarifying the mutation pathway and consequences of SARS-CoV-2 for developing countermeasures against such viruses as soon as possible.
Collapse
Affiliation(s)
| | - Qing Ye
- Department of ‘A’, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310052, China;
| |
Collapse
|
24
|
Pinder CL, Jankovic D, Fox TA, Kirkwood A, Enfield L, Alrubayyi A, Touizer E, Ford R, Pocock R, Shin J, Ziegler J, Thomson KJ, Ardeshna KM, Peppa D, McCoy LE, Morris EC. Humoral and cellular responses to SARS-CoV-2 in patients with B-cell haematological malignancies improve with successive vaccination. Br J Haematol 2023; 202:1091-1103. [PMID: 37402627 PMCID: PMC10953351 DOI: 10.1111/bjh.18962] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 07/06/2023]
Abstract
Patients with haematological malignancies are more likely to have poor responses to vaccination. Here we provide detailed analysis of the humoral and cellular responses to COVID-19 vaccination in 69 patients with B-cell malignancies. Measurement of anti-spike IgG in serum demonstrated a low seroconversion rate with 27.1% and 46.8% of patients seroconverting after the first and second doses of vaccine, respectively. In vitro pseudoneutralisation assays demonstrated a poor neutralising response, with 12.5% and 29.5% of patients producing a measurable neutralising titre after the first and second doses, respectively. A third dose increased seropositivity to 54.3% and neutralisation to 51.5%, while a fourth dose further increased both seropositivity and neutralisation to 87.9%. Neutralisation titres post-fourth dose showed a positive correlation with the size of the B-cell population measured by flow cytometry, suggesting an improved response correlating with recovery of the B-cell compartment after B-cell depletion treatments. In contrast, interferon gamma ELISpot analysis showed a largely intact T-cell response, with the percentage of patients producing a measurable response boosted by the second dose to 75.5%. This response was maintained thereafter, with only a small increase following the third and fourth doses, irrespective of the serological response at these timepoints.
Collapse
Affiliation(s)
| | - Dylan Jankovic
- Division of Infection and ImmunityUniversity College LondonLondonUK
| | - Thomas A. Fox
- Division of Infection and ImmunityUniversity College LondonLondonUK
- Department of Clinical HaematologyUniversity College London Hospitals, NHS Foundation TrustLondonUK
| | - Amy Kirkwood
- CR UK and UCL Cancer Trials CentreUCL Cancer Institute, UCLLondonUK
| | - Louise Enfield
- Department of Clinical HaematologyUniversity College London Hospitals, NHS Foundation TrustLondonUK
| | | | - Emma Touizer
- Division of Infection and ImmunityUniversity College LondonLondonUK
| | - Rosemarie Ford
- Division of Infection and ImmunityUniversity College LondonLondonUK
| | - Rachael Pocock
- Department of Clinical HaematologyUniversity College London Hospitals, NHS Foundation TrustLondonUK
| | - Jin‐Sup Shin
- Department of Clinical HaematologyUniversity College London Hospitals, NHS Foundation TrustLondonUK
| | - Joseph Ziegler
- Department of Clinical HaematologyUniversity College London Hospitals, NHS Foundation TrustLondonUK
| | - Kirsty J. Thomson
- Department of Clinical HaematologyUniversity College London Hospitals, NHS Foundation TrustLondonUK
| | - Kirit M. Ardeshna
- Department of Clinical HaematologyUniversity College London Hospitals, NHS Foundation TrustLondonUK
| | - Dimitra Peppa
- Division of Infection and ImmunityUniversity College LondonLondonUK
| | - Laura E. McCoy
- Division of Infection and ImmunityUniversity College LondonLondonUK
| | - Emma C. Morris
- Division of Infection and ImmunityUniversity College LondonLondonUK
- Department of Clinical HaematologyUniversity College London Hospitals, NHS Foundation TrustLondonUK
| |
Collapse
|
25
|
Hederman AP, Natarajan H, Heyndrickx L, Ariën KK, Wiener JA, Wright PF, Bloch EM, Tobian AAR, Redd AD, Blankson JN, Rottenstreich A, Zarbiv G, Wolf D, Goetghebuer T, Marchant A, Ackerman ME. SARS-CoV-2 vaccination elicits broad and potent antibody effector functions to variants of concern in vulnerable populations. Nat Commun 2023; 14:5171. [PMID: 37620337 PMCID: PMC10449910 DOI: 10.1038/s41467-023-40960-0] [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: 09/28/2022] [Accepted: 08/17/2023] [Indexed: 08/26/2023] Open
Abstract
SARS-CoV-2 variants have continuously emerged in the face of effective vaccines. Reduced neutralization against variants raises questions as to whether other antibody functions are similarly compromised, or if they might compensate for lost neutralization activity. Here, the breadth and potency of antibody recognition and effector function is surveyed following either infection or vaccination. Considering pregnant women as a model cohort with higher risk of severe illness and death, we observe similar binding and functional breadth for healthy and immunologically vulnerable populations, but considerably greater functional antibody breadth and potency across variants associated with vaccination. In contrast, greater antibody functional activity targeting the endemic coronavirus OC43 is noted among convalescent individuals, illustrating a dichotomy in recognition between close and distant human coronavirus strains associated with exposure history. This analysis of antibody functions suggests the differential potential for antibody effector functions to contribute to protecting vaccinated and convalescent subjects as novel variants continue to evolve.
Collapse
Affiliation(s)
| | - Harini Natarajan
- Department of Immunology and Microbiology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA
| | - Leo Heyndrickx
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Kevin K Ariën
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Joshua A Wiener
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Peter F Wright
- Department of Pediatrics, Geisel School of Medicine at Dartmouth, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Evan M Bloch
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Aaron A R Tobian
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Andrew D Redd
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Joel N Blankson
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Amihai Rottenstreich
- Department of Obstetrics and Gynecology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Gila Zarbiv
- Clinical Virology Unit, Hadassah University Medical Center, Jerusalem, Israel
| | - Dana Wolf
- Clinical Virology Unit, Hadassah University Medical Center, Jerusalem, Israel
| | - Tessa Goetghebuer
- Institute for Medical Immunology, Université libre de Bruxelles, Charleroi, Belgium
- Pediatric Department, CHU St Pierre, Brussels, Belgium
| | - Arnaud Marchant
- Institute for Medical Immunology, Université libre de Bruxelles, Charleroi, Belgium
| | - Margaret E Ackerman
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
- Department of Immunology and Microbiology, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
26
|
Loke J, Upasani V, Gaskell C, Fox S, Fletcher R, Thomas C, Hopkins L, Kumari A, Tang T, Yafai E, Boucher R, Homer V, Toth A, Chan YLT, Randall K, Rider T, O'Nions J, Drew V, Pillai A, Dungarwalla M, Murray D, Khan A, Wandroo F, Moore S, Krishnamurthy P, Huang YWJ, Knapper S, Byrne J, Zhao R, Craddock C, Parry H, Moss P, Stanworth SJ, Lowe DM. Defective T-cell response to COVID-19 vaccination in acute myeloid leukaemia and myelodysplastic syndromes. Br J Haematol 2023; 202:498-503. [PMID: 37303189 DOI: 10.1111/bjh.18894] [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: 02/25/2023] [Revised: 05/18/2023] [Accepted: 05/19/2023] [Indexed: 06/13/2023]
Abstract
Limited data exist on COVID-19 vaccination efficacy in patients with acute myeloid leukemia and myelodysplasia with excess blasts (AML/MDS-EB2). We report results from a prospective study, PACE (Patients with AML and COVID-19 Epidemiology). 93 patients provided samples post-vaccine 2 or 3 (PV2, PV3). Antibodies against SARS-COV-2 spike antigen were detectable in all samples. Neutralization of the omicron variant was poorer than ancestral variants but improved PV3. In contrast, adequate T-cell reactivity to SARS-COV-2 spike protein was seen in only 16/47 (34%) patients PV2 and 23/52 (44%) PV3. Using regression models, disease response (not in CR/Cri), and increasing age predicted poor T cell response.
Collapse
Affiliation(s)
- Justin Loke
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
- Dana Farber Cancer Institute, Boston, Massachusetts, USA
| | | | | | - Sonia Fox
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Rachel Fletcher
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Catherine Thomas
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Louise Hopkins
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Anita Kumari
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Tina Tang
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Emily Yafai
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Rebecca Boucher
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Victoria Homer
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Arpad Toth
- Clatterbridge Cancer Hospital, Liverpool, UK
| | | | - Katie Randall
- South Warwickshire University NHS Foundation Trust, Warwick, UK
| | - Tom Rider
- Brighton and Sussex University Hospitals NHS Trust, Brighton, UK
| | | | | | | | | | | | | | - Farooq Wandroo
- Sandwell and West Birmingham Hospitals NHS Trust, West Bromwich, UK
| | - Sally Moore
- Royal United Hospital Bath NHS Foundation Trust, Bath, UK
| | | | | | | | - Jenny Byrne
- Nottingham University Hospitals Trust, Nottingham, UK
| | | | - Charles Craddock
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
- CRUK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Helen Parry
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
- University of Birmingham, Birmingham, UK
| | - Paul Moss
- Centre for Clinical Haematology, Queen Elizabeth Hospital, Birmingham, UK
- University of Birmingham, Birmingham, UK
| | - Simon J Stanworth
- Oxford University Hospitals, Oxford, UK
- University of Oxford, Oxford, UK
- NHS Blood and Transplant, Oxford, UK
| | - David M Lowe
- University College London, London, UK
- Royal Free London NHS Foundation Trust, London, UK
| |
Collapse
|
27
|
Devaux CA, Fantini J. ACE2 receptor polymorphism in humans and animals increases the risk of the emergence of SARS-CoV-2 variants during repeated intra- and inter-species host-switching of the virus. Front Microbiol 2023; 14:1199561. [PMID: 37520374 PMCID: PMC10373931 DOI: 10.3389/fmicb.2023.1199561] [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: 04/03/2023] [Accepted: 06/23/2023] [Indexed: 08/01/2023] Open
Abstract
Like other coronaviruses, SARS-CoV-2 has ability to spread through human-to-human transmission and to circulate from humans to animals and from animals to humans. A high frequency of SARS-CoV-2 mutations has been observed in the viruses isolated from both humans and animals, suggesting a genetic fitness under positive selection in both ecological niches. The most documented positive selection force driving SARS-CoV-2 mutations is the host-specific immune response. However, after electrostatic interactions with lipid rafts, the first contact between the virus and host proteins is the viral spike-cellular receptor binding. Therefore, it is likely that the first level of selection pressure impacting viral fitness relates to the virus's affinity for its receptor, the angiotensin I converting enzyme 2 (ACE2). Although sufficiently conserved in a huge number of species to support binding of the viral spike with enough affinity to initiate fusion, ACE2 is highly polymorphic both among species and within a species. Here, we provide evidence suggesting that when the viral spike-ACE2 receptor interaction is not optimal, due to host-switching, mutations can be selected to improve the affinity of the spike for the ACE2 expressed by the new host. Notably, SARS-CoV-2 is mutation-prone in the spike receptor binding domain (RBD), allowing a better fit for ACE2 orthologs in animals. It is possibly that this may also be true for rare human alleles of ACE2 when the virus is spreading to billions of people. In this study, we present evidence that human subjects expressing the rare E329G allele of ACE2 with higher allele frequencies in European populations exhibit a improved affinity for the SARS-CoV-2 spike N501Y variant of the virus. This may suggest that this viral N501Y variant emerged in the human population after SARS-CoV-2 had infected a human carrying the rare E329G allele of ACE2. In addition, this viral evolution could impact viral replication as well as the ability of the adaptive humoral response to control infection with RBD-specific neutralizing antibodies. In a shifting landscape, this ACE2-driven genetic drift of SARS-CoV-2 which we have named the 'boomerang effect', could complicate the challenge of preventing COVID with a SARS-CoV-2 spike-derived vaccine.
Collapse
Affiliation(s)
- Christian A. Devaux
- Laboratory Microbes Evolution Phylogeny and Infection (MEPHI), Aix-Marseille Université, IRD, APHM, MEPHI, IHU–Méditerranée Infection, Marseille, France
- Centre National de la Recherche Scientifique (CNRS-SNC5039), Marseille, France
| | - Jacques Fantini
- INSERM UMR_S1072, Marseille, France, Aix-Marseille Université, Marseille, France
| |
Collapse
|
28
|
Payen SH, Gorzalski A, Siao DD, Pandori M, Verma SC, Rossetto CC. Analysis of SARS-CoV-2 variants from patient specimens in Nevada from October 2020 to August 2021. INFECTION, GENETICS AND EVOLUTION : JOURNAL OF MOLECULAR EPIDEMIOLOGY AND EVOLUTIONARY GENETICS IN INFECTIOUS DISEASES 2023; 111:105434. [PMID: 37059256 PMCID: PMC10098042 DOI: 10.1016/j.meegid.2023.105434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 04/16/2023]
Abstract
In early 2020, the emergence and spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the human population quickly developed into a global pandemic. SARS-CoV-2 is the etiological agent of coronavirus disease 2019 (COVID-19) which has a broad range of respiratory illnesses. As the virus circulates, it acquires nucleotide changes. These mutations are potentially due to the inherent differences in the selection pressures within the human population compared to the original zoonotic reservoir of SARS-CoV-2 and formerly naïve humans. The acquired mutations will most likely be neutral, but some may have implications for viral transmission, disease severity, and resistance to therapies or vaccines. This is a follow-up study from our early report (Hartley et al. J Genet Genomics. 01202021;48(1):40-51) which detected a rare variant (nsp12, RdRp P323F) circulating within Nevada in mid 2020 at high frequency. The primary goals of the current study were to determine the phylogenetic relationship of the SARS-CoV-2 genomes within Nevada and to determine if there are any unusual variants within Nevada compared to the current database of SARS-CoV-2 sequences. Whole genome sequencing and analysis of SARS-CoV-2 from 425 positively identified nasopharyngeal/nasal swab specimens were performed from October 2020 to August 2021 to determine any variants that could result in potential escape from current therapeutics. Our analysis focused on nucleotide mutations that generated amino acid variations in the viral Spike (S) protein, Receptor binding domain (RBD), and the RNA-dependent RNA-polymerase (RdRp) complex. The data indicate that SARS-CoV-2 sequences from Nevada did not contain any unusual variants that had not been previously reported. Additionally, we did not detect the previously identified the RdRp P323F variant in any of the samples. This suggests that the rare variant we detected before was only able to circulate because of the stay-at-home orders and semi-isolation experience during the early months of the pandemic. IMPORTANCE: SARS-COV-2 continues to circulate in the human population. In this study, SARS-CoV-2 positive nasopharyngeal/nasal swab samples were used for whole genome sequencing to determine the phylogenetic relationship of SARS-CoV-2 sequences within Nevada from October 2020 to August 2021. The resulting data is being added to a continually growing database of SARS-CoV-2 sequences that will be important for understanding the transmission and evolution of the virus as it spreads around the globe.
Collapse
Affiliation(s)
- Shannon Harger Payen
- Department of Microbiology & Immunology, University of Nevada, Reno, 1664 North Virginia St. MS 320 Reno, NV 89557, USA; School of Medicine, University of Nevada, Reno, 1664 North Virginia St., Reno, NV 89557, USA
| | - Andrew Gorzalski
- Nevada State Public Health Laboratory, 1664 North Virginia St., Reno, NV 89557, USA
| | - Danielle Denise Siao
- Nevada State Public Health Laboratory, 1664 North Virginia St., Reno, NV 89557, USA
| | - Mark Pandori
- School of Medicine, University of Nevada, Reno, 1664 North Virginia St., Reno, NV 89557, USA; Nevada State Public Health Laboratory, 1664 North Virginia St., Reno, NV 89557, USA
| | - Subhash C Verma
- Department of Microbiology & Immunology, University of Nevada, Reno, 1664 North Virginia St. MS 320 Reno, NV 89557, USA; School of Medicine, University of Nevada, Reno, 1664 North Virginia St., Reno, NV 89557, USA
| | - Cyprian C Rossetto
- Department of Microbiology & Immunology, University of Nevada, Reno, 1664 North Virginia St. MS 320 Reno, NV 89557, USA; School of Medicine, University of Nevada, Reno, 1664 North Virginia St., Reno, NV 89557, USA.
| |
Collapse
|
29
|
Shoaei P, Ranjbar MM, Tokhanbigli S, Ataei B, Alibakhshi A, Haghjooy Javanmard S, Ahangarzadeh S. Comparative Analysis and Identification of Spike Mutations in Iranian COVID-19 Samples from the First Three Waves of Disease. Adv Biomed Res 2023; 12:153. [PMID: 37564431 PMCID: PMC10410413 DOI: 10.4103/abr.abr_171_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: 05/24/2022] [Revised: 11/16/2022] [Accepted: 12/04/2022] [Indexed: 08/12/2023] Open
Abstract
Background The spike surface glycoprotein of SARS-CoV-2 is the essential protein in virus attachment to the target cell and cell entrance. As this protein contains immunodominant epitopes and is the main target for immune recognition, it is the critical target for vaccine and therapeutics development. In the current research, we analyzed the variability and mutations of the spike glycoprotein isolated from 72 COVID-19-positive patients from Iran's first three waves of disease. Materials and Methods The RNA was extracted from nasopharyngeal samples of confirmed COVID-19 cases and served as a template for cDNA synthesis and reverse transcriptase polymerase chain reaction. The reverse transcriptase polymerase chain reaction products of each sample were assembled and sequenced. Results After analysis of 72 sequences, we obtained 46 single nucleotide polymorphisms, including 23 that produce amino acid changes. Our analysis showed that the most frequent mutation was the D614G (in the samples of the second and third waves). Conclusions Our findings suggest that developing effective vaccines requires identifying the predominant variants of SARS-CoV-2 in each community.
Collapse
Affiliation(s)
- Parisa Shoaei
- Nosocomial Infection Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad M. Ranjbar
- Razi Vaccine and Serum Research Institute, Agricultural Research, Education, and Extension Organization (AREEO), Karaj, Iran
| | - Samaneh Tokhanbigli
- Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Behrouz Ataei
- Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Abbas Alibakhshi
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shaghayegh Haghjooy Javanmard
- Department of Physiology, Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahrzad Ahangarzadeh
- Infectious Diseases and Tropical Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
30
|
Pandit R, Matthews QL. A SARS-CoV-2: Companion Animal Transmission and Variants Classification. Pathogens 2023; 12:775. [PMID: 37375465 DOI: 10.3390/pathogens12060775] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/19/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
The continuous emergence of novel viruses and their diseases are a threat to global public health as there have been three outbreaks of coronaviruses that are highly pathogenic to humans in the span of the last two decades, severe acute respiratory syndrome (SARS)-CoV in 2002, Middle East respiratory syndrome (MERS)-CoV in 2012, and novel SARS-CoV-2 which emerged in 2019. The unprecedented spread of SARS-CoV-2 worldwide has given rise to multiple SARS-CoV-2 variants that have either altered transmissibility, infectivity, or immune escaping ability, causing diseases in a broad range of animals including human and non-human hosts such as companion, farm, zoo, or wild animals. In this review, we have discussed the recent SARS-CoV-2 outbreak, potential animal reservoirs, and natural infections in companion and farm animals, with a particular focus on SARS-CoV-2 variants. The expeditious development of COVID-19 vaccines and the advancements in antiviral therapeutics have contained the COVID-19 pandemic to some extent; however, extensive research and surveillance concerning viral epidemiology, animal transmission, variants, or seroprevalence in diverse hosts are essential for the future eradication of COVID-19.
Collapse
Affiliation(s)
- Rachana Pandit
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA
| | - Qiana L Matthews
- Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA
- Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA
| |
Collapse
|
31
|
van der Klaauw AA, Horner EC, Pereyra-Gerber P, Agrawal U, Foster WS, Spencer S, Vergese B, Smith M, Henning E, Ramsay ID, Smith JA, Guillaume SM, Sharpe HJ, Hay IM, Thompson S, Innocentin S, Booth LH, Robertson C, McCowan C, Kerr S, Mulroney TE, O'Reilly MJ, Gurugama TP, Gurugama LP, Rust MA, Ferreira A, Ebrahimi S, Ceron-Gutierrez L, Scotucci J, Kronsteiner B, Dunachie SJ, Klenerman P, Park AJ, Rubino F, Lamikanra AA, Stark H, Kingston N, Estcourt L, Harvala H, Roberts DJ, Doffinger R, Linterman MA, Matheson NJ, Sheikh A, Farooqi IS, Thaventhiran JED. Accelerated waning of the humoral response to COVID-19 vaccines in obesity. Nat Med 2023; 29:1146-1154. [PMID: 37169862 PMCID: PMC10202802 DOI: 10.1038/s41591-023-02343-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 04/07/2023] [Indexed: 05/13/2023]
Abstract
Obesity is associated with an increased risk of severe Coronavirus Disease 2019 (COVID-19) infection and mortality. COVID-19 vaccines reduce the risk of serious COVID-19 outcomes; however, their effectiveness in people with obesity is incompletely understood. We studied the relationship among body mass index (BMI), hospitalization and mortality due to COVID-19 among 3.6 million people in Scotland using the Early Pandemic Evaluation and Enhanced Surveillance of COVID-19 (EAVE II) surveillance platform. We found that vaccinated individuals with severe obesity (BMI > 40 kg/m2) were 76% more likely to experience hospitalization or death from COVID-19 (adjusted rate ratio of 1.76 (95% confidence interval (CI), 1.60-1.94). We also conducted a prospective longitudinal study of a cohort of 28 individuals with severe obesity compared to 41 control individuals with normal BMI (BMI 18.5-24.9 kg/m2). We found that 55% of individuals with severe obesity had unquantifiable titers of neutralizing antibody against authentic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus compared to 12% of individuals with normal BMI (P = 0.0003) 6 months after their second vaccine dose. Furthermore, we observed that, for individuals with severe obesity, at any given anti-spike and anti-receptor-binding domain (RBD) antibody level, neutralizing capacity was lower than that of individuals with a normal BMI. Neutralizing capacity was restored by a third dose of vaccine but again declined more rapidly in people with severe obesity. We demonstrate that waning of COVID-19 vaccine-induced humoral immunity is accelerated in individuals with severe obesity. As obesity is associated with increased hospitalization and mortality from breakthrough infections, our findings have implications for vaccine prioritization policies.
Collapse
Affiliation(s)
- Agatha A van der Klaauw
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Emily C Horner
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Pehuén Pereyra-Gerber
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Utkarsh Agrawal
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | | | - Sarah Spencer
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Bensi Vergese
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, UK
- NIHR Cambridge Clinical Research Facility, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Miriam Smith
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Elana Henning
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Isobel D Ramsay
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Jack A Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | | | - Iain M Hay
- Babraham Institute, Babraham Research Campus, Cambridge, UK
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
| | - Sam Thompson
- Babraham Institute, Babraham Research Campus, Cambridge, UK
| | | | - Lucy H Booth
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Chris Robertson
- Department of Mathematics and Statistics, University of Strathclyde, Glasgow, UK
| | - Colin McCowan
- Nuffield Department of Primary Care Health Sciences, University of Oxford, Oxford, UK
| | - Steven Kerr
- Usher Institute, University of Edinburgh, Edinburgh, UK
| | | | | | | | | | - Maria A Rust
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Alex Ferreira
- MRC Toxicology Unit, University of Cambridge, Cambridge, UK
| | - Soraya Ebrahimi
- Immunology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Clinical Biochemistry, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Lourdes Ceron-Gutierrez
- Immunology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Clinical Biochemistry, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Jacopo Scotucci
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, UK
| | - Barbara Kronsteiner
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Susanna J Dunachie
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- NDM Centre for Global Health Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
- Mahidol-Oxford Tropical Medicine Research Unit, Bangkok, Thailand
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
- NDM Centre for Global Health Research, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Adrian J Park
- Clinical Biochemistry, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Francesco Rubino
- Department of Diabetes, King's College London and King's College Hospital NHS Foundation Trust, London, UK
| | - Abigail A Lamikanra
- NHS Blood and Transplant, Oxford, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Hannah Stark
- NIHR BioResource, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Nathalie Kingston
- NIHR BioResource, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Lise Estcourt
- NHS Blood and Transplant, Oxford, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | | | - David J Roberts
- NHS Blood and Transplant, Oxford, UK
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Rainer Doffinger
- Immunology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Clinical Biochemistry, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Nicholas J Matheson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Infectious Diseases, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Aziz Sheikh
- Usher Institute, University of Edinburgh, Edinburgh, UK.
| | - I Sadaf Farooqi
- University of Cambridge Metabolic Research Laboratories and NIHR Cambridge Biomedical Research Centre, Wellcome-Medical Research Council (MRC) Institute of Metabolic Science, University of Cambridge, Cambridge, UK.
| | | |
Collapse
|
32
|
Radić L, Sliepen K, Yin V, Brinkkemper M, Capella-Pujol J, Schriek AI, Torres JL, Bangaru S, Burger JA, Poniman M, Bontjer I, Bouhuijs JH, Gideonse D, Eggink D, Ward AB, Heck AJ, Van Gils MJ, Sanders RW, Schinkel J. Bispecific antibodies combine breadth, potency, and avidity of parental antibodies to neutralize sarbecoviruses. iScience 2023; 26:106540. [PMID: 37063468 PMCID: PMC10065043 DOI: 10.1016/j.isci.2023.106540] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 02/07/2023] [Accepted: 03/24/2023] [Indexed: 04/03/2023] Open
Abstract
SARS-CoV-2 variants evade current monoclonal antibody therapies. Bispecific antibodies (bsAbs) combine the specificities of two distinct antibodies taking advantage of the avidity and synergy provided by targeting different epitopes. Here we used controlled Fab-arm exchange to produce bsAbs that neutralize SARS-CoV and SARS-CoV-2 variants, including Omicron and its subvariants, by combining potent SARS-CoV-2-specific neutralizing antibodies with broader antibodies that also neutralize SARS-CoV. We demonstrated that the parental antibodies rely on avidity for neutralization using bsAbs containing one irrelevant Fab arm. Using mass photometry to measure the formation of antibody:spike complexes, we determined that bsAbs increase binding stoichiometry compared to corresponding cocktails, without a loss of binding affinity. The heterogeneous binding pattern of bsAbs to spike, observed by negative-stain electron microscopy and mass photometry provided evidence for both intra- and inter-spike crosslinking. This study highlights the utility of cross-neutralizing antibodies for designing bivalent agents to combat circulating and future SARS-like coronaviruses.
Collapse
Affiliation(s)
- Laura Radić
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Kwinten Sliepen
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Victor Yin
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
- Netherlands Proteomics Center, 3584 CH Utrecht, the Netherlands
| | - Mitch Brinkkemper
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Joan Capella-Pujol
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Angela I. Schriek
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Jonathan L. Torres
- Department of Structural Biology and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Sandhya Bangaru
- Department of Structural Biology and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Judith A. Burger
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Meliawati Poniman
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Ilja Bontjer
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Joey H. Bouhuijs
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - David Gideonse
- Center for Infectious Disease Control, WHO COVID-19 reference laboratory, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands
| | - Dirk Eggink
- Center for Infectious Disease Control, WHO COVID-19 reference laboratory, National Institute for Public Health and the Environment (RIVM), 3721 MA Bilthoven, the Netherlands
| | - Andrew B. Ward
- Department of Structural Biology and Computational Biology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Albert J.R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 CH Utrecht, the Netherlands
- Netherlands Proteomics Center, 3584 CH Utrecht, the Netherlands
| | - Marit J. Van Gils
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Rogier W. Sanders
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| | - Janke Schinkel
- Amsterdam UMC location University of Amsterdam, Department of Medical Microbiology and Infection prevention, Laboratory of Experimental Virology, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
- Amsterdam institute for Infection and Immunity, Infectious diseases, Amsterdam, the Netherlands
| |
Collapse
|
33
|
Pascall DJ, Vink E, Blacow R, Bulteel N, Campbell A, Campbell R, Clifford S, Davis C, da Silva Filipe A, El Sakka N, Fjodorova L, Forrest R, Goldstein E, Gunson R, Haughney J, Holden MTG, Honour P, Hughes J, James E, Lewis T, Lycett S, MacLean O, McHugh M, Mollett G, Onishi Y, Parcell B, Ray S, Robertson DL, Shabaan S, Shepherd JG, Smollett K, Templeton K, Wastnedge E, Wilkie C, Williams T, Thomson EC, The COVID-19 Genomics UK (COG-UK) Consortium. The SARS-CoV-2 Alpha variant was associated with increased clinical severity of COVID-19 in Scotland: A genomics-based retrospective cohort analysis. PLoS One 2023; 18:e0284187. [PMID: 37053201 PMCID: PMC10101505 DOI: 10.1371/journal.pone.0284187] [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: 08/12/2022] [Accepted: 03/24/2023] [Indexed: 04/14/2023] Open
Abstract
OBJECTIVES The SARS-CoV-2 Alpha variant was associated with increased transmission relative to other variants present at the time of its emergence and several studies have shown an association between Alpha variant infection and increased hospitalisation and 28-day mortality. However, none have addressed the impact on maximum severity of illness in the general population classified by the level of respiratory support required, or death. We aimed to do this. METHODS In this retrospective multi-centre clinical cohort sub-study of the COG-UK consortium, 1475 samples from Scottish hospitalised and community cases collected between 1st November 2020 and 30th January 2021 were sequenced. We matched sequence data to clinical outcomes as the Alpha variant became dominant in Scotland and modelled the association between Alpha variant infection and severe disease using a 4-point scale of maximum severity by 28 days: 1. no respiratory support, 2. supplemental oxygen, 3. ventilation and 4. death. RESULTS Our cumulative generalised linear mixed model analyses found evidence (cumulative odds ratio: 1.40, 95% CI: 1.02, 1.93) of a positive association between increased clinical severity and lineage (Alpha variant versus pre-Alpha variants). CONCLUSIONS The Alpha variant was associated with more severe clinical disease in the Scottish population than co-circulating lineages.
Collapse
Affiliation(s)
- David J. Pascall
- MRC Biostatistics Unit, University of Cambridge, Cambridge, United Kingdom
- Joint Universities Pandemic and Epidemiological Research (JUNIPER) Consortium, United Kingdom
| | - Elen Vink
- MRC–University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
- NHS Lothian, Edinburgh, United Kingdom
| | - Rachel Blacow
- MRC–University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
- NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | | | | | | | | | - Chris Davis
- MRC–University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
| | - Ana da Silva Filipe
- MRC–University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
| | | | | | | | | | - Rory Gunson
- NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - John Haughney
- NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | - Matthew T. G. Holden
- Public Health Scotland, Edinburgh, United Kingdom
- School of Medicine, University of St Andrews, St Andrews, Fife, United Kingdom
| | | | - Joseph Hughes
- MRC–University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
| | - Edward James
- NHS Borders, Melrose, Scottish Borders, United Kingdom
| | - Tim Lewis
- NHS Lothian, Edinburgh, United Kingdom
| | - Samantha Lycett
- The Roslin Institute, University of Edinburgh, Midlothian, United Kingdom
| | - Oscar MacLean
- MRC–University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
| | | | - Guy Mollett
- MRC–University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
- NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
| | | | - Ben Parcell
- School of Life Sciences, University of Dundee, Dundee, United Kingdom
| | - Surajit Ray
- School of Mathematics and Statistics, University of Glasgow, Glasgow, United Kingdom
| | - David L. Robertson
- MRC–University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
| | | | - James G. Shepherd
- MRC–University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
| | - Katherine Smollett
- MRC–University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
| | | | | | - Craig Wilkie
- School of Mathematics and Statistics, University of Glasgow, Glasgow, United Kingdom
| | - Thomas Williams
- NHS Lothian, Edinburgh, United Kingdom
- Royal Hospital for Children and Young People, University of Edinburgh, Edinburgh, United Kingdom
| | - Emma C. Thomson
- MRC–University of Glasgow Centre for Virus Research (CVR), Glasgow, United Kingdom
- NHS Greater Glasgow and Clyde, Glasgow, United Kingdom
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | |
Collapse
|
34
|
Ahmadi S, Bazargan M, Elahi R, Esmaeilzadeh A. Immune evasion of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2); molecular approaches. Mol Immunol 2023; 156:10-19. [PMID: 36857806 PMCID: PMC9684099 DOI: 10.1016/j.molimm.2022.11.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2022] [Revised: 11/04/2022] [Accepted: 11/22/2022] [Indexed: 11/25/2022]
Abstract
In December 2019, a new betacoronavirus, known as severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), caused an outbreak at the Wuhan seafood market in China. The disease was further named coronavirus disease 2019 (COVID-19). In March 2020, the World Health Organization (WHO) announced the disease to be a pandemic, as more cases were reported globally. SARS-CoV-2, like many other viruses, employs diverse strategies to elude the host immune response and/or counter immune responses. The infection outcome mainly depends on interactions between the virus and the host immune system. Inhibiting IFN production, blocking IFN signaling, enhancing IFN resistance, and hijacking the host's translation machinery to expedite the production of viral proteins are among the main immune evasion mechanisms of SARS-CoV-2. SARS-CoV-2 also downregulates the expression of MHC-I on infected cells, which is an additional immune-evasion mechanism of this virus. Moreover, antigenic modifications to the spike (S) protein, such as deletions, insertions, and also substitutions are essential for resistance to SARS-CoV-2 neutralizing antibodies. This review assesses the interaction between SARS-CoV-2 and host immune response and cellular and molecular approaches used by SARS-CoV-2 for immune evasion. Understanding the mechanisms of SARS-CoV-2 immune evasion is essential since it can improve the development of novel antiviral treatment options as well as vaccination methods.
Collapse
Affiliation(s)
- Shahrzad Ahmadi
- Virology Research Center, The National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Masih Daneshvari Hospital, Allergy and Immunology Subspecialty Lab, Tehran, Iran
| | - Mahsa Bazargan
- Virology Research Center, The National Research Institute of Tuberculosis and Lung Diseases (NRITLD), Masih Daneshvari Hospital, Allergy and Immunology Subspecialty Lab, Tehran, Iran,Department of Immunology, School of Medicine, Sahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Elahi
- M.D., School of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Abdolreza Esmaeilzadeh
- Department of Immunology, Zanjan University of Medical Sciences, Zanjan, Iran; Cancer Gene Therapy Research Center (CGRC), Zanjan University of Medical Sciences, Zanjan, Iran.
| |
Collapse
|
35
|
Wu D, Efimov GA, Bogolyubova AV, Pierce BG, Mariuzza RA. Structural insights into protection against a SARS-CoV-2 spike variant by T cell receptor diversity. J Biol Chem 2023; 299:103035. [PMID: 36806685 PMCID: PMC9934920 DOI: 10.1016/j.jbc.2023.103035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/06/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
T cells play a crucial role in combatting SARS-CoV-2 and forming long-term memory responses to this coronavirus. The emergence of SARS-CoV-2 variants that can evade T cell immunity has raised concerns about vaccine efficacy and the risk of reinfection. Some SARS-CoV-2 T cell epitopes elicit clonally restricted CD8+ T cell responses characterized by T cell receptors (TCRs) that lack structural diversity. Mutations in such epitopes can lead to loss of recognition by most T cells specific for that epitope, facilitating viral escape. Here, we studied an HLA-A2-restricted spike protein epitope (RLQ) that elicits CD8+ T cell responses in COVID-19 convalescent patients characterized by highly diverse TCRs. We previously reported the structure of an RLQ-specific TCR (RLQ3) with greatly reduced recognition of the most common natural variant of the RLQ epitope (T1006I). Opposite to RLQ3, TCR RLQ7 recognizes T1006I with even higher functional avidity than the WT epitope. To explain the ability of RLQ7, but not RLQ3, to tolerate the T1006I mutation, we determined structures of RLQ7 bound to RLQ-HLA-A2 and T1006I-HLA-A2. These complexes show that there are multiple structural solutions to recognizing RLQ and thereby generating a clonally diverse T cell response to this epitope that assures protection against viral escape and T cell clonal loss.
Collapse
Affiliation(s)
- Daichao Wu
- Laboratory of Structural Immunology, Department of Hepatopancreatobiliary Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China; W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | | | | | - Brian G Pierce
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Roy A Mariuzza
- W.M. Keck Laboratory for Structural Biology, University of Maryland Institute for Bioscience and Biotechnology Research, Rockville, Maryland, USA; Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA.
| |
Collapse
|
36
|
Zhao Y, Zhao Y, Xie L, Li Q, Zhang Y, Zang Y, Li X, Zhang L, Yang Z. Identification of Potential Lead Compounds Targeting Novel Druggable Cavity of SARS-CoV-2 Spike Trimer by Molecular Dynamics Simulations. Int J Mol Sci 2023; 24:6281. [PMID: 37047254 PMCID: PMC10094189 DOI: 10.3390/ijms24076281] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/07/2023] [Accepted: 03/20/2023] [Indexed: 03/29/2023] Open
Abstract
The global pandemic of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become an urgent public health problem. Spike (S) protein mediates the fusion between the virus and the host cell membranes, consequently emerging as an important target of drug design. The lack of comparisons of in situ full-length S homotrimer structures in different states hinders understanding the structures and revealing the function, thereby limiting the discovery and development of therapeutic agents. Here, the steady-state structures of the in situ full-length S trimer in closed and open states (Sclosed and Sopen) were modeled with the constraints of density maps, associated with the analysis of the dynamic structural differences. Subsequently, we identified various regions with structure and property differences as potential binding pockets for ligands that promote the formation of inactive trimeric protein complexes. By using virtual screening strategy and a newly defined druggable cavity, five ligands were screened with potential bioactivities. Then molecular dynamic (MD) simulations were performed on apo protein structures and ligand bound complexes to reveal the conformational changes upon ligand binding. Our simulation results revealed that sulforaphane (SFN), which has the best binding affinity, could inhibit the conformational changes of S homotrimer that would occur during the viral membrane fusion. Our results could aid in the understanding of the regulation mechanism of S trimer aggregation and the structure-activity relationship, facilitating the development of potential antiviral agents.
Collapse
Affiliation(s)
- Yizhen Zhao
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
| | - Yifan Zhao
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
| | - Linke Xie
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
| | - Qian Li
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| | - Yuze Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
| | - Yongjian Zang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
| | - Xuhua Li
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
| | - Lei Zhang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
| | - Zhiwei Yang
- MOE Key Laboratory for Nonequilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
- School of Life Science and Technology, Xi’an Jiaotong University, Xi’an 710049, China
| |
Collapse
|
37
|
Production of neutralizing antibody fragment variants in the cytoplasm of E. coli for rapid screening: SARS-CoV-2 a case study. Sci Rep 2023; 13:4408. [PMID: 36927743 PMCID: PMC10019796 DOI: 10.1038/s41598-023-31369-2] [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: 12/18/2022] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
Global health challenges such as the coronavirus pandemic warrant the urgent need for a system that allows efficient production of diagnostic and therapeutic interventions. Antibody treatments against SARS-CoV-2 were developed with an unprecedented pace and this enormous progress was achieved mainly through recombinant protein production technologies combined with expeditious screening approaches. A heterologous protein production system that allows efficient soluble production of therapeutic antibody candidates against rapidly evolving variants of deadly pathogens is an important step in preparedness towards future pandemic challenges. Here, we report cost and time-effective soluble production of SARS-CoV-2 receptor binding domain (RBD) variants as well as an array of neutralizing antibody fragments (Fabs) based on Casirivimab and Imdevimab using the CyDisCo system in the cytoplasm of E. coli. We also report variants of the two Fabs with higher binding affinity against SARS-CoV-2 RBD and suggest this cytoplasmic production of disulfide containing antigens and antibodies can be broadly applied towards addressing future global public health threats.
Collapse
|
38
|
Shi J, Zhang Y, Yang M. Recent development of microfluidics-based platforms for respiratory virus detection. BIOMICROFLUIDICS 2023; 17:024104. [PMID: 37035101 PMCID: PMC10076069 DOI: 10.1063/5.0135778] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 02/27/2023] [Indexed: 06/19/2023]
Abstract
With the global outbreak of SARS-CoV-2, the inadequacies of current detection technology for respiratory viruses have been recognized. Rapid, portable, accurate, and sensitive assays are needed to expedite diagnosis and early intervention. Conventional methods for detection of respiratory viruses include cell culture-based assays, serological tests, nucleic acid detection (e.g., RT-PCR), and direct immunoassays. However, these traditional methods are often time-consuming, labor-intensive, and require laboratory facilities, which cannot meet the testing needs, especially during pandemics of respiratory diseases, such as COVID-19. Microfluidics-based techniques can overcome these demerits and provide simple, rapid, accurate, and cost-effective analysis of intact virus, viral antigen/antibody, and viral nucleic acids. This review aims to summarize the recent development of microfluidics-based techniques for detection of respiratory viruses. Recent advances in different types of microfluidic devices for respiratory virus diagnostics are highlighted, including paper-based microfluidics, continuous-flow microfluidics, and droplet-based microfluidics. Finally, the future development of microfluidic technologies for respiratory virus diagnostics is discussed.
Collapse
Affiliation(s)
- Jingyu Shi
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon 999077, Hong Kong SAR, People's Republic of China
| | - Yu Zhang
- Department of Mechanical and Automotive Engineering, Royal Melbourne Institute of Technology, Melbourne, VIC 3000, Australia
| | - Mo Yang
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Kowloon 999077, Hong Kong SAR, People's Republic of China
| |
Collapse
|
39
|
Bayani F, Hashkavaei NS, Arjmand S, Rezaei S, Uskoković V, Alijanianzadeh M, Uversky VN, Ranaei Siadat SO, Mozaffari-Jovin S, Sefidbakht Y. An overview of the vaccine platforms to combat COVID-19 with a focus on the subunit vaccines. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 178:32-49. [PMID: 36801471 PMCID: PMC9938630 DOI: 10.1016/j.pbiomolbio.2023.02.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 02/13/2023] [Accepted: 02/16/2023] [Indexed: 02/21/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an emerging virus that has caused the recent coronavirus disease (COVID-19) global pandemic. The current approved COVID-19 vaccines have shown considerable efficiency against hospitalization and death. However, the continuation of the pandemic for more than two years and the likelihood of new strain emergence despite the global rollout of vaccination highlight the immediate need for the development and improvement of vaccines. mRNA, viral vector, and inactivated virus vaccine platforms were the first members of the worldwide approved vaccine list. Subunit vaccines. which are vaccines based on synthetic peptides or recombinant proteins, have been used in lower numbers and limited countries. The unavoidable advantages of this platform, including safety and precise immune targeting, make it a promising vaccine with wider global use in the near future. This review article summarizes the current knowledge on different vaccine platforms, focusing on the subunit vaccines and their clinical trial advancements against COVID-19.
Collapse
Affiliation(s)
- Fatemeh Bayani
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | | | - Sareh Arjmand
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | - Shokouh Rezaei
- Protein Research Center, Shahid Beheshti University, Tehran, Iran
| | - Vuk Uskoković
- Department of Mechanical Engineering, San Diego State University, San Diego, CA, 92182, USA; TardigradeNano LLC, Irvine, CA, 92604, USA
| | - Mahdi Alijanianzadeh
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, Kharazmi University, Tehran, Iran
| | - Vladimir N Uversky
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, 33612, USA.
| | | | - Sina Mozaffari-Jovin
- Department of Medical Genetics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Yahya Sefidbakht
- Protein Research Center, Shahid Beheshti University, Tehran, Iran.
| |
Collapse
|
40
|
Kaplonek P, Cizmeci D, Lee JSL, Shin SA, Fischinger S, Gobeil P, Pillet S, Charland N, Ward BJ, Alter G. Robust induction of functional humoral response by a plant-derived Coronavirus-like particle vaccine candidate for COVID-19. NPJ Vaccines 2023; 8:13. [PMID: 36781879 PMCID: PMC9924894 DOI: 10.1038/s41541-023-00612-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Accepted: 01/27/2023] [Indexed: 02/15/2023] Open
Abstract
Despite the success of existing COVID-19 vaccine platforms, the persistent limitations in global deployment of vaccines and waning immunity exhibited by many of the currently deployed vaccine platforms have led to perpetual outbreaks of SARS-CoV-2 variants of concern. Thus, there is an urgent need to develop new durable vaccine candidates, to expand the global vaccine pipeline, and provide safe and effective solutions for every country worldwide. Here we deeply profiled the functional humoral response induced by two doses of AS03-adjuvanted and non-adjuvanted plant-derived Coronavirus-like particle (CoVLP) vaccine candidate from the phase 1 clinical trial, at peak immunogenicity and six months post-vaccination. AS03-adjuvanted CoVLP induced robust and durable SARS-CoV-2 specific humoral immunity, marked by strong IgG1antibody responses, potent FcγR binding, and antibody effector function. Contrary to a decline in neutralizing antibody titers, the FcγR2A-receptor binding capacity and antibody-mediated effector functions, such as opsonophagocytosis, remained readily detectable for at least six months.
Collapse
Affiliation(s)
| | - Deniz Cizmeci
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | - Sally A Shin
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | | | | | | | | | - Brian J Ward
- Medicago Inc., Quebec City, QC, Canada.
- Research Institute of the McGill University Health Centre, Montréal, QC, Canada.
| | - Galit Alter
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA.
| |
Collapse
|
41
|
Hirunpattarasilp C, James G, Kwanthongdee J, Freitas F, Huo J, Sethi H, Kittler JT, Owens RJ, McCoy LE, Attwell D. SARS-CoV-2 triggers pericyte-mediated cerebral capillary constriction. Brain 2023; 146:727-738. [PMID: 35867861 PMCID: PMC9384509 DOI: 10.1093/brain/awac272] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/04/2022] [Accepted: 07/11/2022] [Indexed: 11/21/2022] Open
Abstract
The SARS-CoV-2 receptor, ACE2, is found on pericytes, contractile cells enwrapping capillaries that regulate brain, heart and kidney blood flow. ACE2 converts vasoconstricting angiotensin II into vasodilating angiotensin-(1-7). In brain slices from hamster, which has an ACE2 sequence similar to human ACE2, angiotensin II evoked a small pericyte-mediated capillary constriction via AT1 receptors, but evoked a large constriction when the SARS-CoV-2 receptor binding domain (RBD, original Wuhan variant) was present. A mutated non-binding RBD did not potentiate constriction. A similar RBD-potentiated capillary constriction occurred in human cortical slices, and was evoked in hamster brain slices by pseudotyped virions expressing SARS-CoV-2 spike protein. This constriction reflects an RBD-induced decrease in the conversion of angiotensin II to angiotensin-(1-7) mediated by removal of ACE2 from the cell surface membrane and was mimicked by blocking ACE2. The clinically used drug losartan inhibited the RBD-potentiated constriction. Thus, AT1 receptor blockers could be protective in COVID-19 by preventing pericyte-mediated blood flow reductions in the brain, and perhaps the heart and kidney.
Collapse
Affiliation(s)
- Chanawee Hirunpattarasilp
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Talat Bang Khen, Lak Si, Bangkok, 10210, Thailand
| | - Greg James
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
- Department of Neurosurgery, Great Ormond Street Hospital, London WC1N 3JH, UK
| | - Jaturon Kwanthongdee
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
- Princess Srisavangavadhana College of Medicine, Chulabhorn Royal Academy, Talat Bang Khen, Lak Si, Bangkok, 10210, Thailand
| | - Felipe Freitas
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Jiandong Huo
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Protein Production UK, The Research Complex at Harwell, and Rosalind Franklin Institute, Harwell Science and Innovation Campus, Didcot OX11 0GD, UK
| | - Huma Sethi
- Division of Neurosurgery, National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| | - Josef T Kittler
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| | - Raymond J Owens
- Division of Structural Biology, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7BN, UK
- Protein Production UK, The Research Complex at Harwell, and Rosalind Franklin Institute, Harwell Science and Innovation Campus, Didcot OX11 0GD, UK
| | - Laura E McCoy
- Division of Infection and Immunity, University College London, London NW3 2PP, UK
| | - David Attwell
- Department of Neuroscience, Physiology and Pharmacology, University College London, London WC1E 6BT, UK
| |
Collapse
|
42
|
Umitaibatin R, Harisna AH, Jauhar MM, Syaifie PH, Arda AG, Nugroho DW, Ramadhan D, Mardliyati E, Shalannanda W, Anshori I. Immunoinformatics Study: Multi-Epitope Based Vaccine Design from SARS-CoV-2 Spike Glycoprotein. Vaccines (Basel) 2023; 11:vaccines11020399. [PMID: 36851275 PMCID: PMC9964839 DOI: 10.3390/vaccines11020399] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/04/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
The coronavirus disease 2019 outbreak has become a huge challenge in the human sector for the past two years. The coronavirus is capable of mutating at a higher rate than other viruses. Thus, an approach for creating an effective vaccine is still needed to induce antibodies against multiple variants with lower side effects. Currently, there is a lack of research on designing a multiepitope of the COVID-19 spike protein for the Indonesian population with comprehensive immunoinformatic analysis. Therefore, this study aimed to design a multiepitope-based vaccine for the Indonesian population using an immunoinformatic approach. This study was conducted using the SARS-CoV-2 spike glycoprotein sequences from Indonesia that were retrieved from the GISAID database. Three SARS-CoV-2 sequences, with IDs of EIJK-61453, UGM0002, and B.1.1.7 were selected. The CD8+ cytotoxic T-cell lymphocyte (CTL) epitope, CD4+ helper T lymphocyte (HTL) epitope, B-cell epitope, and IFN-γ production were predicted. After modeling the vaccines, molecular docking, molecular dynamics, in silico immune simulations, and plasmid vector design were performed. The designed vaccine is antigenic, non-allergenic, non-toxic, capable of inducing IFN-γ with a population reach of 86.29% in Indonesia, and has good stability during molecular dynamics and immune simulation. Hence, this vaccine model is recommended to be investigated for further study.
Collapse
Affiliation(s)
- Ramadhita Umitaibatin
- Lab-on-Chip Group, Department of Biomedical Engineering, School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung 40132, Indonesia
| | - Azza Hanif Harisna
- Nano Center Indonesia, Jl. Raya Puspiptek, South Tangerang 15314, Indonesia
| | | | - Putri Hawa Syaifie
- Nano Center Indonesia, Jl. Raya Puspiptek, South Tangerang 15314, Indonesia
| | | | - Dwi Wahyu Nugroho
- Nano Center Indonesia, Jl. Raya Puspiptek, South Tangerang 15314, Indonesia
| | - Donny Ramadhan
- Research Center for Pharmaceutical Ingredients and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong 16911, Indonesia
| | - Etik Mardliyati
- Research Center for Vaccine and Drug, National Research and Innovation Agency (BRIN), Cibinong 16911, Indonesia
| | - Wervyan Shalannanda
- Department of Telecommunication Engineering, School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung 40132, Indonesia
| | - Isa Anshori
- Lab-on-Chip Group, Department of Biomedical Engineering, School of Electrical Engineering and Informatics, Bandung Institute of Technology, Bandung 40132, Indonesia
- Correspondence:
| |
Collapse
|
43
|
Celikgil A, Massimi AB, Nakouzi A, Herrera NG, Morano NC, Lee JH, Yoon HA, Garforth SJ, Almo SC. SARS-CoV-2 multi-antigen protein microarray for detailed characterization of antibody responses in COVID-19 patients. PLoS One 2023; 18:e0276829. [PMID: 36757919 PMCID: PMC9910743 DOI: 10.1371/journal.pone.0276829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 01/13/2023] [Indexed: 02/10/2023] Open
Abstract
Antibodies against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) target multiple epitopes on different domains of the spike protein, and other SARS-CoV-2 proteins. We developed a SARS-CoV-2 multi-antigen protein microarray with the nucleocapsid, spike and its domains (S1, S2), and variants with single (D614G, E484K, N501Y) or double substitutions (N501Y/Deletion69/70), allowing a more detailed high-throughput analysis of the antibody repertoire following infection. The assay was demonstrated to be reliable and comparable to ELISA. We analyzed antibodies from 18 COVID-19 patients and 12 recovered convalescent donors. The S IgG level was higher than N IgG in most of the COVID-19 patients, and the receptor-binding domain of S1 showed high reactivity, but no antibodies were detected against the heptad repeat domain 2 of S2. Furthermore, antibodies were detected against S variants with single and double substitutions in COVID-19 patients who were infected with SARS-CoV-2 early in the pandemic. Here we demonstrated that the SARS-CoV-2 multi-antigen protein microarray is a powerful tool for detailed characterization of antibody responses, with potential utility in understanding the disease progress and assessing current vaccines and therapies against evolving SARS-CoV-2.
Collapse
Affiliation(s)
- Alev Celikgil
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Aldo B. Massimi
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Antonio Nakouzi
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Natalia G. Herrera
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Nicholas C. Morano
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - James H. Lee
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Hyun ah Yoon
- Department of Medicine, Division of Infectious Diseases, Albert Einstein College of Medicine and Montefiore Medical Center, Bronx, New York, United States of America
| | - Scott J. Garforth
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Steven C. Almo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
44
|
Mesner D, Reuschl AK, Whelan MVX, Bronzovich T, Haider T, Thorne LG, Ragazzini R, Bonfanti P, Towers GJ, Jolly C. SARS-CoV-2 evolution influences GBP and IFITM sensitivity. Proc Natl Acad Sci U S A 2023; 120:e2212577120. [PMID: 36693093 PMCID: PMC9945951 DOI: 10.1073/pnas.2212577120] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 12/06/2022] [Indexed: 01/25/2023] Open
Abstract
SARS-CoV-2 spike requires proteolytic processing for viral entry. A polybasic furin-cleavage site (FCS) in spike, and evolution toward an optimized FCS by dominant variants of concern (VOCs), are linked to enhanced infectivity and transmission. Here we show interferon-inducible restriction factors Guanylate-binding proteins (GBP) 2 and 5 interfere with furin-mediated spike cleavage and inhibit the infectivity of early-lineage isolates Wuhan-Hu-1 and VIC. By contrast, VOCs Alpha and Delta escape restriction by GBP2/5 that we map to the spike substitution D614G present in these VOCs. Despite inhibition of spike cleavage, these viruses remained sensitive to plasma membrane IFITM1, but not endosomal IFITM2 and 3, consistent with a preference for TMPRSS2-dependent plasma membrane entry. Strikingly, we find that Omicron is unique among VOCs, being sensitive to restriction factors GBP2/5, and also IFITM1, 2, and 3. Using chimeric spike mutants, we map the Omicron phenotype and show that the S1 domain determines Omicron's sensitivity to GBP2/5, whereas the S2' domain determines its sensitivity to endosomal IFITM2/3 and preferential use of TMPRSS2-independent entry. We propose that evolution of SARS-CoV-2 for the D614G substitution has allowed for escape from GBP restriction factors, but the selective pressures on Omicron for spike changes that mediate antibody escape, and altered tropism, have come at the expense of increased sensitivity to innate immune restriction factors that target virus entry.
Collapse
Affiliation(s)
- Dejan Mesner
- Division of Infection and Immunity, University College London, WC1E 6BTLondon, UK
| | - Ann-Kathrin Reuschl
- Division of Infection and Immunity, University College London, WC1E 6BTLondon, UK
| | - Matthew V. X. Whelan
- Division of Infection and Immunity, University College London, WC1E 6BTLondon, UK
| | - Taylor Bronzovich
- Division of Infection and Immunity, University College London, WC1E 6BTLondon, UK
| | - Tafhima Haider
- Division of Infection and Immunity, University College London, WC1E 6BTLondon, UK
- Centre for Genomics and Child Health, Blizard Institute, Queen Mary University of London, E1 2ATLondon, UK
| | - Lucy G. Thorne
- Division of Infection and Immunity, University College London, WC1E 6BTLondon, UK
| | - Roberta Ragazzini
- Division of Infection and Immunity, University College London, WC1E 6BTLondon, UK
- Epithelial Stem Cell Biology and Regenerative Medicine Laboratory, The Francis Crick Institute, LondonNW1 1AT, UK
| | - Paola Bonfanti
- Division of Infection and Immunity, University College London, WC1E 6BTLondon, UK
- Epithelial Stem Cell Biology and Regenerative Medicine Laboratory, The Francis Crick Institute, LondonNW1 1AT, UK
| | - Greg J. Towers
- Division of Infection and Immunity, University College London, WC1E 6BTLondon, UK
| | - Clare Jolly
- Division of Infection and Immunity, University College London, WC1E 6BTLondon, UK
| |
Collapse
|
45
|
Touizer E, Alrubayyi A, Ford R, Hussain N, Gerber PP, Shum HL, Rees-Spear C, Muir L, Gea-Mallorquí E, Kopycinski J, Jankovic D, Jeffery-Smith A, Pinder CL, Fox TA, Williams I, Mullender C, Maan I, Waters L, Johnson M, Madge S, Youle M, Barber TJ, Burns F, Kinloch S, Rowland-Jones S, Gilson R, Matheson NJ, Morris E, Peppa D, McCoy LE. Attenuated humoral responses in HIV after SARS-CoV-2 vaccination linked to B cell defects and altered immune profiles. iScience 2023; 26:105862. [PMID: 36590902 PMCID: PMC9788849 DOI: 10.1016/j.isci.2022.105862] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/04/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
We assessed a cohort of people living with human immunodeficiency virus (PLWH) (n = 110) and HIV negative controls (n = 64) after 1, 2 or 3 SARS-CoV-2 vaccine doses. At all timepoints, PLWH had significantly lower neutralizing antibody (nAb) titers than HIV-negative controls. We also observed a delayed development of neutralization in PLWH that was underpinned by a reduced frequency of spike-specific memory B cells (MBCs). Improved neutralization breadth was seen against the Omicron variant (BA.1) after the third vaccine dose in PLWH but lower nAb responses persisted and were associated with global MBC dysfunction. In contrast, SARS-CoV-2 vaccination induced robust T cell responses that cross-recognized variants in PLWH. Strikingly, individuals with low or absent neutralization had detectable functional T cell responses. These PLWH had reduced numbers of circulating T follicular helper cells and an enriched population of CXCR3+CD127+CD8+T cells after two doses of SARS-CoV-2 vaccination.
Collapse
Affiliation(s)
- Emma Touizer
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Aljawharah Alrubayyi
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Rosemarie Ford
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Noshin Hussain
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Pehuén Pereyra Gerber
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Hiu-Long Shum
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Chloe Rees-Spear
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Luke Muir
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | | | - Jakub Kopycinski
- Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Dylan Jankovic
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Anna Jeffery-Smith
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Christopher L. Pinder
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Thomas A. Fox
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Ian Williams
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
| | - Claire Mullender
- Institute for Global Health, University College London, London, UK
| | - Irfaan Maan
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
- Institute for Global Health, University College London, London, UK
| | - Laura Waters
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
| | - Margaret Johnson
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Sara Madge
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Michael Youle
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Tristan J. Barber
- Institute for Global Health, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Fiona Burns
- Institute for Global Health, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Sabine Kinloch
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | | | - Richard Gilson
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
- Institute for Global Health, University College London, London, UK
| | - Nicholas J. Matheson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Emma Morris
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| | - Dimitra Peppa
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust, London, UK
| | - Laura E. McCoy
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, London, UK
| |
Collapse
|
46
|
Marzi R, Bassi J, Silacci-Fregni C, Bartha I, Muoio F, Culap K, Sprugasci N, Lombardo G, Saliba C, Cameroni E, Cassotta A, Low JS, Walls AC, McCallum M, Tortorici MA, Bowen JE, Dellota EA, Dillen JR, Czudnochowski N, Pertusini L, Terrot T, Lepori V, Tarkowski M, Riva A, Biggiogero M, Franzetti-Pellanda A, Garzoni C, Ferrari P, Ceschi A, Giannini O, Havenar-Daughton C, Telenti A, Arvin A, Virgin HW, Sallusto F, Veesler D, Lanzavecchia A, Corti D, Piccoli L. Maturation of SARS-CoV-2 Spike-specific memory B cells drives resilience to viral escape. iScience 2023; 26:105726. [PMID: 36507220 PMCID: PMC9721160 DOI: 10.1016/j.isci.2022.105726] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/21/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022] Open
Abstract
Memory B cells (MBCs) generate rapid antibody responses upon secondary encounter with a pathogen. Here, we investigated the kinetics, avidity, and cross-reactivity of serum antibodies and MBCs in 155 SARS-CoV-2 infected and vaccinated individuals over a 16-month time frame. SARS-CoV-2-specific MBCs and serum antibodies reached steady-state titers with comparable kinetics in infected and vaccinated individuals. Whereas MBCs of infected individuals targeted both prefusion and postfusion Spike (S), most vaccine-elicited MBCs were specific for prefusion S, consistent with the use of prefusion-stabilized S in mRNA vaccines. Furthermore, a large fraction of MBCs recognizing postfusion S cross-reacted with human betacoronaviruses. The avidity of MBC-derived and serum antibodies increased over time resulting in enhanced resilience to viral escape by SARS-CoV-2 variants, including Omicron BA.1 and BA.2 sublineages, albeit only partially for BA.4 and BA.5 sublineages. Overall, the maturation of high-affinity and broadly reactive MBCs provides the basis for effective recall responses to future SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Roberta Marzi
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Jessica Bassi
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | | | - Istvan Bartha
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Francesco Muoio
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Katja Culap
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Nicole Sprugasci
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Gloria Lombardo
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Christian Saliba
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Elisabetta Cameroni
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Jun Siong Low
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | | | - Matthew McCallum
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | - John E. Bowen
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | | | | | - Laura Pertusini
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Tatiana Terrot
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | | | - Maciej Tarkowski
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Milan, Italy
| | - Agostino Riva
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, Milan, Italy
- III Division of Infectious Diseases, ASST Fatebenefratelli Sacco, Luigi Sacco Hospital, Milan, Italy
| | - Maira Biggiogero
- Clinical Research Unit, Clinica Luganese Moncucco, Lugano, Switzerland
| | | | - Christian Garzoni
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco, Lugano, Switzerland
| | - Paolo Ferrari
- Division of Nephrology, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Alessandro Ceschi
- Clinical Trial Unit, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, Zurich, Switzerland
| | - Olivier Giannini
- Faculty of Biomedical Sciences, Università della Svizzera italiana, Lugano, Switzerland
- Department of Medicine, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | | | | | - Ann Arvin
- Vir Biotechnology, San Francisco, CA, USA
| | - Herbert W. Virgin
- Vir Biotechnology, San Francisco, CA, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, USA
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | | | - Davide Corti
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| | - Luca Piccoli
- Humabs BioMed SA, a subsidiary of Vir Biotechnology, Bellinzona, Switzerland
| |
Collapse
|
47
|
Niu X, Xu J, Liu M, Tu H, Koenig SN, Saif LJ, Jones DM, Wang Q. Isolation and characterization of a SARS-CoV-2 variant with a Q677H mutation in the spike protein. Arch Virol 2023; 168:5. [PMID: 36539656 PMCID: PMC9767398 DOI: 10.1007/s00705-022-05621-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 10/17/2022] [Indexed: 12/24/2022]
Abstract
We isolated 20 SARS-CoV-2 strains from positive clinical samples collected in Columbus, Ohio, and investigated the replication of one pair of isolates: a clade 20G strain and a variant of this strain carrying a Q677H mutation in the spike protein and six other amino acid mutations. The OSU.20G variant replicated to a higher peak infectious titer than the 20G base strain in Vero-E6 cells, but the titers were similar when both strains were grown in Calu-3 cells. These results suggest that the OSU.20G variant has increased replication fitness compared to the 20G base strain. This may have contributed to its emergence in December 2020-January 2021.
Collapse
Affiliation(s)
- Xiaoyu Niu
- grid.261331.40000 0001 2285 7943Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH USA ,grid.10698.360000000122483208Present Address: Department of Epidemiology, University of North Carolina At Chapel Hill, Chapel Hill, NC 27516 USA
| | - Jiayu Xu
- grid.261331.40000 0001 2285 7943Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH USA
| | - Mingde Liu
- grid.261331.40000 0001 2285 7943Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH USA
| | - Huolin Tu
- grid.261331.40000 0001 2285 7943James Molecular Laboratory at Polaris, The Ohio State University James Cancer Center, Columbus, OH USA
| | - Sara N. Koenig
- grid.261331.40000 0001 2285 7943Dorothy M. Davis Heart and Lung Research Institute and Frick Center for Heart Failure and Arrhythmia Research, The Ohio State University, Columbus, USA ,grid.261331.40000 0001 2285 7943Department of Physiology and Cell Biology, The Ohio State University, Columbus, USA
| | - Linda J. Saif
- grid.261331.40000 0001 2285 7943Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH USA
| | - Daniel M. Jones
- grid.261331.40000 0001 2285 7943James Molecular Laboratory at Polaris, The Ohio State University James Cancer Center, Columbus, OH USA ,grid.412332.50000 0001 1545 0811Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH USA ,grid.413944.f0000 0001 0447 4797The Ohio State University Comprehensive Cancer Center, The Ohio State University James Cancer Center, Columbus, OH USA
| | - Qiuhong Wang
- grid.261331.40000 0001 2285 7943Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH USA ,grid.261331.40000 0001 2285 7943Center for Food Animal Health, Department of Animal Sciences, College of Food, Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH USA
| |
Collapse
|
48
|
Abavisani M, Rahimian K, Mahdavi B, Tokhanbigli S, Mollapour Siasakht M, Farhadi A, Kodori M, Mahmanzar M, Meshkat Z. Mutations in SARS-CoV-2 structural proteins: a global analysis. Virol J 2022; 19:220. [PMID: 36528612 PMCID: PMC9759450 DOI: 10.1186/s12985-022-01951-7] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 12/08/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Emergence of new variants mainly variants of concerns (VOC) is caused by mutations in main structural proteins of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Therefore, we aimed to investigate the mutations among structural proteins of SARS-CoV-2 globally. METHODS We analyzed samples of amino-acid sequences (AASs) for envelope (E), membrane (M), nucleocapsid (N), and spike (S) proteins from the declaration of the coronavirus 2019 (COVID-19) as pandemic to January 2022. The presence and location of mutations were then investigated by aligning the sequences to the reference sequence and categorizing them based on frequency and continent. Finally, the related human genes with the viral structural genes were discovered, and their interactions were reported. RESULTS The results indicated that the most relative mutations among the E, M, N, and S AASs occurred in the regions of 7 to 14, 66 to 88, 164 to 205, and 508 to 635 AAs, respectively. The most frequent mutations in E, M, N, and S proteins were T9I, I82T, R203M/R203K, and D614G. D614G was the most frequent mutation in all six geographical areas. Following D614G, L18F, A222V, E484K, and N501Y, respectively, were ranked as the most frequent mutations in S protein globally. Besides, A-kinase Anchoring Protein 8 Like (AKAP8L) was shown as the linkage unit between M, E, and E cluster genes. CONCLUSION Screening the structural protein mutations can help scientists introduce better drug and vaccine development strategies.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Microbiology and Virology, School of Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Karim Rahimian
- Institute of Biochemistry and Biophysics (IBB), University of Tehran, Tehran, Iran
| | - Bahar Mahdavi
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Samaneh Tokhanbigli
- Department of Molecular and Cellular Sciences, Faculty of Advanced Sciences and Technology, Pharmaceutical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Mahsa Mollapour Siasakht
- Department of Biochemistry, Erasmus University Medical Center, P.O. Box 2040, 3000 CA, Rotterdam, The Netherlands
| | - Amin Farhadi
- Department of Biology, Payame Noor University, Tehran, Iran
| | - Mansoor Kodori
- Non Communicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Mohammadamin Mahmanzar
- Department of Bioinformatics, Kish International Campus University of Tehran, Kish, Iran.
| | - Zahra Meshkat
- Department of Microbiology and Virology, School of Medicine, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
49
|
Touizer E, Alrubbayi A, Ford R, Hussain N, Gerber PP, Shum HL, Rees-Spear C, Muir L, Gea-Mallorquí E, Kopycinski J, Jankovic D, Pinder C, Fox TA, Williams I, Mullender C, Maan I, Waters L, Johnson M, Madge S, Youle M, Barber T, Burns F, Kinloch S, Rowland-Jones S, Gilson R, Matheson NJ, Morris E, Peppa D, McCoy LE. Attenuated humoral responses in HIV infection after SARS-CoV-2 vaccination are linked to global B cell defects and cellular immune profiles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2022:2022.11.11.516111. [PMID: 36380764 PMCID: PMC9665338 DOI: 10.1101/2022.11.11.516111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
People living with HIV (PLWH) on suppressive antiretroviral therapy (ART) can have residual immune dysfunction and often display poorer responses to vaccination. We assessed in a cohort of PLWH (n=110) and HIV negative controls (n=64) the humoral and spike-specific B-cell responses following 1, 2 or 3 SARS-CoV-2 vaccine doses. PLWH had significantly lower neutralizing antibody (nAb) titers than HIV-negative controls at all studied timepoints. Moreover, their neutralization breadth was reduced with fewer individuals developing a neutralizing response against the Omicron variant (BA.1) relative to controls. We also observed a delayed development of neutralization in PLWH that was underpinned by a reduced frequency of spike-specific memory B cells (MBCs) and pronounced B cell dysfunction. Improved neutralization breadth was seen after the third vaccine dose in PLWH but lower nAb responses persisted and were associated with global, but not spike-specific, MBC dysfunction. In contrast to the inferior antibody responses, SARS-CoV-2 vaccination induced robust T cell responses that cross-recognized variants in PLWH. Strikingly, a subset of PLWH with low or absent neutralization had detectable functional T cell responses. These individuals had reduced numbers of circulating T follicular helper cells and an enriched population of CXCR3 + CD127 + CD8 + T cells after two doses of SARS-CoV-2 vaccination, which may compensate for sub-optimal serological responses in the event of infection. Therefore, normalisation of B cell homeostasis could improve serological responses to vaccines in PLWH and evaluating T cell immunity could provide a more comprehensive immune status profile in these individuals and others with B cell imbalances.
Collapse
Affiliation(s)
- Emma Touizer
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Aljawharah Alrubbayi
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
- Nuffield Department of Medicine, University of Oxford, UK
| | - Rosemarie Ford
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Noshin Hussain
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Pehuén Pereyra Gerber
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, UK
| | - Hiu-Long Shum
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Chloe Rees-Spear
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Luke Muir
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | | | | | - Dylan Jankovic
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Christopher Pinder
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Thomas A Fox
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Ian Williams
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, UK
| | | | - Irfaan Maan
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, UK
- Institute for Global Health, University College London, UK
| | - Laura Waters
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, UK
| | - Margaret Johnson
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | - Sara Madge
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | - Michael Youle
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | - Tristan Barber
- Institute for Global Health, University College London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | - Fiona Burns
- Institute for Global Health, University College London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | - Sabine Kinloch
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
- The Ian Charleson Day Centre, Royal Free Hospital NHS Foundation Trust UK
| | | | - Richard Gilson
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, UK
- Institute for Global Health, University College London, UK
| | - Nicholas J Matheson
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Department of Medicine, University of Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Emma Morris
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| | - Dimitra Peppa
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
- Mortimer Market Centre, Department of HIV, Central and North West London NHS Trust, UK
- Institute for Global Health, University College London, UK
| | - Laura E McCoy
- Institute for Immunity and Transplantation, Division of Infection and Immunity, University College London, UK
| |
Collapse
|
50
|
Yu C, Fengzhao Z, Hongmei W, Zeyuan L, Yu L, Yuhang G, Rufei S, Qingzhu J, Xiaorong S, Xia W, Caiping S, Zhi X, Chunmei L. The impact of vaccination on patients with COVID-19 during the wave of Omicron in Shanghai. Front Public Health 2022; 10:1054313. [PMID: 36438303 PMCID: PMC9682288 DOI: 10.3389/fpubh.2022.1054313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 10/26/2022] [Indexed: 11/11/2022] Open
Abstract
Background The global health has been affected by the COVID-19 pandemic persistently, of which Omicron is currently the predominant variant. However, the impact of vaccination on Omicron remained uncertain. Objective This study sought to explore the effect of vaccination on patients infected with Omicron. Methods A retrospective observational cohort was conducted in the largest Fangcang shelter hospital in Shanghai from April 1 to May 30, 2022. The demographics, length of hospital stay, clinical symptoms, the comorbidities and vaccination status were recorded. Clinical outcomes of the vaccinated and non-vaccinated groups were compared and analyzed. Results Of the 3,119 patients who fulfilled the eligibility criteria and were enrolled in the study, 2,226 (71.4%) patients had received nCoV-19 vaccine while 893 (28.6%) patients had not received it before admission. Patients in the vaccinated group had significantly shorter length of hospital stay than those in the unvaccinated group (15.48 ± 2.708 vs. 15.85 ± 3.102, p < 0.001). More asymptomatic patients were observed in the vaccinated group than the non-vaccinated (70.4 vs. 64.5%, p < 0.001). Further subgroup analysis demonstrated that the older the age, the more significant the difference was (p < 0.005). Conclusions Vaccination was associated with a significant reduction in the severity of Omicron infection compared with no vaccination. Vaccination appears to make Omicron-infected people with milder symptoms than unvaccinated people. This suggests the potential effectiveness of current vaccines against Omicron.
Collapse
Affiliation(s)
- Chen Yu
- Department of Orthopaedics, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Zhu Fengzhao
- Department of Orthopaedics, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Wu Hongmei
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Lei Zeyuan
- Department of Plastic Surgery, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Liu Yu
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Guo Yuhang
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Shen Rufei
- Department of Endocrinology, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Jia Qingzhu
- Department of Oncology, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Sun Xiaorong
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Wang Xia
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Song Caiping
- Department of Office of the Hospital, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Xu Zhi
- Department of Respiratory, Xinqiao Hospital, The Army Medical University, Chongqing, China
| | - Luo Chunmei
- Department of Orthopaedics, Xinqiao Hospital, The Army Medical University, Chongqing, China
| |
Collapse
|