1
|
Bean DJ, Liang YM, Avila F, He X, Asundi A, Sagar M. Endemic coronavirus infection is associated with SARS-CoV-2 Fc receptor-binding antibodies. J Virol 2025:e0055025. [PMID: 40387363 DOI: 10.1128/jvi.00550-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 05/20/2025] Open
Abstract
Recent documented infection with an endemic coronavirus (eCoV) is associated with less severe coronavirus disease 2019 (COVID-19), yet the immune mechanism behind this protection has not been fully explored. We measured both antibody and T-cell responses against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in SARS-CoV-2-naïve individuals, classified into two groups: those with or without presumed recent eCoV infections. There was no difference in neutralizing antibodies and T-cell responses against SARS-CoV-2 antigens between the two groups. SARS-CoV-2-naïve individuals with recent presumed eCoV infection, however, had higher and significantly correlated levels of Fc receptor (FcR)-binding antibodies against eCoV spikes (S) and SARS-CoV-2 S2. Recent eCoV infection boosts cross-reactive antibodies that can mediate Fc effector functions, and this may play a role in the observed heterotypic immune protection against severe COVID-19. IMPORTANCE With the recent emergence of SARS-CoV-2 and other pathogenic coronaviruses, it is important to understand how the immune system may protect against disease from future coronavirus outbreaks. We investigated the adaptive immune responses elicited from a "common cold" eCoV and measured the cross-reactivity against SARS-CoV-2 in individuals classified as having or not having a recent eCoV infection. Although both groups had similar cross-reactive T-cell and neutralizing antibody responses, individuals with a recent eCoV infection had higher antibody levels capable of Fc receptor binding. Antibodies with enhanced Fc receptor binding could mediate the killing of virally infected cells through mechanisms such as antibody-dependent cellular cytotoxicity, which may reduce the severity of COVID-19. Antibodies capable of mediating Fc effector functions may be critical for therapies and vaccines against future pathogenic coronavirus outbreaks.
Collapse
Affiliation(s)
- David J Bean
- Department of Virology, Immunology and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Yan Mei Liang
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Frida Avila
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Xianbao He
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Archana Asundi
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Manish Sagar
- Department of Virology, Immunology and Microbiology, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
- Department of Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, Massachusetts, USA
| |
Collapse
|
2
|
Browne DJ, Crooks P, Smith C, Doolan DL. Differential reactivity of SARS-CoV-2 S-protein T-cell epitopes in vaccinated versus naturally infected individuals. Clin Transl Immunology 2025; 14:e70031. [PMID: 40342296 PMCID: PMC12056234 DOI: 10.1002/cti2.70031] [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: 11/18/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 05/11/2025] Open
Abstract
Objectives Vaccine-induced protective immunity against SARS-CoV-2 has proved difficult to sustain. Robust T-cell responses are thought to play an important role, but T-cell responses against the SARS-CoV-2 spike protein (S-protein), the core vaccine antigen, following vaccination or natural infection are incompletely understood. Methods Herein, the reactivity of 170 putative SARS-CoV-2 S-protein CD8+ and CD4+ T-cell peptide epitopes in the same individuals prior to vaccination, after COVID-19 vaccination, and again following subsequent natural infection was assayed using a high-throughput reverse transcription-quantitative PCR (HTS-RT-qPCR) assay. Results The profile of immunoreactive SARS-CoV-2 S-protein epitopes differed between vaccination and natural infection. Vaccine-induced immunoreactive epitopes were localised primarily into two extra-domanial regions. In contrast, epitopes recognised following natural infection were spread across the antigen. Furthermore, T-cell epitopes in naïve individuals were primarily recognised in association with HLA-A, while natural infection shifted epitope associations towards HLA-B, particularly the B7 supertype. Conclusion This study provides insight into T-cell responses against the SARS-CoV-2 S-protein following vaccination and subsequent natural infection.
Collapse
Affiliation(s)
- Daniel J Browne
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQLDAustralia
| | - Pauline Crooks
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of ImmunologyQIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of ImmunologyQIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
- Faculty of MedicineThe University of QueenslandBrisbaneQLDAustralia
| | - Denise L Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health and MedicineJames Cook UniversityCairnsQLDAustralia
- Institute for Molecular BioscienceThe University of QueenslandSt LuciaQLDAustralia
| |
Collapse
|
3
|
Karl V, Hofmann M, Thimme R. Role of antiviral CD8+ T cell immunity to SARS-CoV-2 infection and vaccination. J Virol 2025; 99:e0135024. [PMID: 40029063 PMCID: PMC11998524 DOI: 10.1128/jvi.01350-24] [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] [Indexed: 03/05/2025] Open
Abstract
The COVID-19 pandemic has greatly enhanced our understanding of CD8+ T cell immunity and their role in natural infection and vaccine-induced protection. Rapid and early SARS-CoV-2-specific CD8+ T cell responses have been associated with efficient viral clearance and mild disease. Virus-specific CD8+ T cell responses can compensate for waning, morbidity-related, and iatrogenic reduction of humoral immunity. After infection or vaccination, SARS-CoV-2-specific memory CD8+ T cells are formed, which mount an efficient recall response in the event of breakthrough infection and help to protect from severe disease. Due to their breadth and ability to target mainly highly conserved epitopes, SARS-CoV-2-specific CD8+ T cells are also able to cross-recognize epitopes of viral variants, thus maintaining immunity even after the emergence of viral evolution. In some cases, however, CD8+ T cells may contribute to the pathogenesis of severe COVID-19. In particular, delayed and uncontrolled, e.g., nonspecific and hyperactivated, cytotoxic CD8+ T cell responses have been linked to poor COVID-19 outcomes. In this minireview, we summarize the tremendous knowledge about CD8+ T cell responses to SARS-CoV-2 infection and COVID-19 vaccination that has been gained over the past 5 years, while also highlighting the critical knowledge gaps that remain.
Collapse
Affiliation(s)
- Vivien Karl
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Maike Hofmann
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Robert Thimme
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology and Infectious Diseases), Freiburg University Medical Center, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
4
|
Kaushal N, Baranwal M. Computational insights in design of Crimean-Congo hemorrhagic fever virus conserved immunogenic nucleoprotein peptides containing multiple epitopes. Biotechnol Appl Biochem 2025; 72:498-512. [PMID: 39402918 DOI: 10.1002/bab.2679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 09/24/2024] [Indexed: 04/09/2025]
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) belongs to Nairoviridae family and has tripartite RNA genome. It is endemic in various countries of Asia, Africa, and Europe and is primarily transmitted by Hyalomma ticks but nosocomial transmission also been reported. Vaccines for CCHF are in early phase of clinical trial; therefore, this work is centered on identification of potential immunogenic peptide as vaccine candidates with application of different immunoinformatics approaches. Eleven conserved (>90%) peptides of CCHFV nucleoprotein were selected for CD8+ T-cell (NetMHCpan 4.1b and NetCTLpan 1.1 server) and CD4+ T-cell (NetMHCIIpan-4.0 server and Tepitool) epitope prediction. Three peptides containing multiple CD8+ and CD4+ T-cell and B-cell epitopes were identified on basis of consensus prediction approach. Peptides displayed good antigenicity score of 0.45-0.68 and predicted to bind with diverse human leukocyte antigen (HLA) alleles. Molecular docking was performed with epitopes to HLA and HLA-epitopes complex to T-cell receptor (TCR). In most of the cases, docked complex of HLA-epitope and HLA-epitopes-TCR have the binding energy close to respective natural bound peptide complex with HLA and TCR. Molecular dynamic simulation also revealed that HLA-peptide complexes have minimum fluctuation and deviation than HLA-peptide-TCR docked over 50 ns simulation run. Considering these findings, identified peptides can serve as potential vaccine candidates for CCHFV disease.
Collapse
Affiliation(s)
- Neha Kaushal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India
| | - Manoj Baranwal
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala, Punjab, India
| |
Collapse
|
5
|
Shah M, Moon SU, Shin J, Choi J, Kim D, Woo HG. Pan-Variant SARS-CoV-2 Vaccines Induce Protective Immunity by Targeting Conserved Epitopes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409919. [PMID: 40014015 PMCID: PMC12021035 DOI: 10.1002/advs.202409919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/27/2025] [Indexed: 02/28/2025]
Abstract
The development of a globally effective COVID-19 vaccine faces significant challenges, particularly in redirecting the B-cell response from immunodominant yet variable regions of viral proteins toward their conserved domains. To address this, an integrated strategy is implemented that combines classical B-cell epitope prediction with protein-antibody cluster docking and antibody titer analysis from 30 vaccinated and convalescent individuals. This approach yields stable immunodominant and immunoprevalent B-cell epitopes capable of eliciting robust antibody responses in BALB/c mice and effectively neutralizing pseudoviruses expressing the Spike protein of SARS-CoV-2 variants of concern, including Alpha, Beta, Gamma, Delta, and Omicron. To achieve a broader T-cell-based immune response, promiscuous T-cell epitopes are identified by integrating classical T-cell epitope predictions, differential scanning fluorimetry, and peptide-MHC structural analysis. Unique peptides with conserved MHC-anchoring residues are identified, enabling binding to a spectrum of MHC-I and MHC-II haplotypes. These peptides elicit strong interferon gamma responses in human peripheral blood mononuclear cells and demonstrate cross-species efficacy by activating both CD4+ and CD8+ T-cells in BALB/c mice. Collectively, these findings highlight the significance of innovative vaccine strategies targeting immunodominant/immunoprevalent B-cell and promiscuous T-cell epitopes to drive broad and robust humoral and cellular immune responses against a wide range of SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Masaud Shah
- Department of PhysiologyAjou University School of MedicineSuwon16499Republic of Korea
- Ajou Translational Omics Center (ATOC)Research Institute for Innovative MedicineAjou University Medical CenterSuwon16499Republic of Korea
| | - Sung Ung Moon
- Department of PhysiologyAjou University School of MedicineSuwon16499Republic of Korea
| | - Ji‐Yon Shin
- Department of PhysiologyAjou University School of MedicineSuwon16499Republic of Korea
- AI‐Superconvergence KIURI Translational Research CenterAjou University School of MedicineSuwon16499Republic of Korea
| | - Ji‐Hye Choi
- Department of PhysiologyAjou University School of MedicineSuwon16499Republic of Korea
- Ajou Translational Omics Center (ATOC)Research Institute for Innovative MedicineAjou University Medical CenterSuwon16499Republic of Korea
| | - Doyoon Kim
- Ajou Translational Omics Center (ATOC)Research Institute for Innovative MedicineAjou University Medical CenterSuwon16499Republic of Korea
| | - Hyun Goo Woo
- Department of PhysiologyAjou University School of MedicineSuwon16499Republic of Korea
- Ajou Translational Omics Center (ATOC)Research Institute for Innovative MedicineAjou University Medical CenterSuwon16499Republic of Korea
- Department of Biomedical ScienceGraduate SchoolAjou UniversitySuwon16499Republic of Korea
| |
Collapse
|
6
|
Barreto AS, de Franca MNF, dos Reis TLDS, Silva JABM, Dos Santos PL, de Oliveira FA, da Silva AM, Magalhaes LS, Secco DA, Andrade MAF, Porto LC, Rosa DS, Cavalcante RCM, Corrêa CB, Sidney J, Sette A, de Almeida RP, Palatnik-de-Sousa CB. Design and development of highly conserved, HLA-promiscuous T cell multiepitope vaccines against human visceral leishmaniasis. Front Immunol 2025; 16:1540537. [PMID: 40230841 PMCID: PMC11994619 DOI: 10.3389/fimmu.2025.1540537] [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: 12/06/2024] [Accepted: 02/24/2025] [Indexed: 04/16/2025] Open
Abstract
Introduction No vaccine is currently licensed against human visceral leishmaniasis (VL), a fatal CD4+ T cell immunosupressive disease against which chemotherapy is reduced to a few toxic drugs. The NH36 nucleoside hydrolase is a DNA metabolism vital enzyme present in all Leishmania species. A vaccine based on such a conserved antigen could protect against both VL and cutaneous leishmaniasis, whose epidemics geographically overlap. Increased frequencies of NH36-specific IL-2+TNF-α+IFN-γ+-producing CD4+ T cells were associated with VL immune protection. Methods the sequences of HLA-Class I and Class II T cell epitopes were predicted in the NH36 peptide sequence using the Tepitope, Propred, IEDB and NetMHCpan EL 4.1 immune informatic tools. The epitopes were synthetized and used to study their reactivity with sera samples, and to stimulate the in vitro response of PBMC of human patients cured from VL, asymptomatic individuals and healthy blood donors of a non-endemic area. Cytokine production was studied intracellularly by flow cytometry (ICS) and cytokine secretion was measured in PBMC supernatants. The HLA typing of DNA patients and the analysis of epitope conservancy in the Leishmania genus were obtained. Two recombinant multiepitope proteins were designed, cloned in E. coli, expressed, purified and used for in vitro stimulation of PBMC of VL cured and asymptomatic patients. Results We identified in silico fifteen NH36 conserved epitopes that correspond to promiscuous binders of HLA-DR, -DQ, -DP class II molecules, as well as HLA-A, B and C class I molecules. Collectively, these epitopes provide high worldwide population coverage of both class I and II alleles, and bound to alleles associated with VL susceptibility and resistance. VL asymptomatic individuals showed maximal frequencies of CD4+ and CD8+ multifunctional IL-2+TNF-α+IFN-γ+-producing T lymphocytes in response to these epitopes, with secretion of TNF-α, IL-1β and IL-6. Two recombinant multiepitope vaccines were designed using these epitopes linked by AAA or GPGPG spacers. Both proteins promoted CD4+ and CD8+ T cell responses in PBMC of VL cured and asymptomatic individuals. Discussion Both MultiAAA and MultiGPGPG proteins could be potentially used for universal human vaccination against leishmaniasis.
Collapse
Affiliation(s)
- Aline Silva Barreto
- Molecular Biology Laboratory, University Hospital, Department of Medicine Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Postgraduate Program in Health Science, Federal University of Sergipe, Aracaju, SE, Brazil
| | - Mariana Nobre Farias de Franca
- Postgraduate Program in Health Science, Federal University of Sergipe, Aracaju, SE, Brazil
- Department of Morphology, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Tatiana Leão dos Santos dos Reis
- Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Postgraduate Program in Plant Biotechnology and Bioprocesses, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | | | - Priscila Lima Dos Santos
- Molecular Biology Laboratory, University Hospital, Department of Medicine Federal University of Sergipe, Aracaju, Sergipe, Brazil
- Postgraduate Program in Health Science, Federal University of Sergipe, Aracaju, SE, Brazil
| | - Fabrícia Alvisi de Oliveira
- Molecular Biology Laboratory, University Hospital, Department of Medicine Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Angela Maria da Silva
- Postgraduate Program in Health Science, Federal University of Sergipe, Aracaju, SE, Brazil
- Department of Medicine, Federal University of Sergipe, Aracaju, Brazil
| | - Lucas Sousa Magalhaes
- Molecular Biology Laboratory, University Hospital, Department of Medicine Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Danielle Angst Secco
- TIXUS Technologic Core for Tissue Repair and Histocompatibility, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Maria Aiza Fontes Andrade
- Department of Pharmacy and Graduate program in Applied Heath Science, Federal University of Sergipe, Lagarto, Sergipe, Brazil
| | - Luís Cristóvão Porto
- TIXUS Technologic Core for Tissue Repair and Histocompatibility, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Daniela Santoro Rosa
- Department of Microbiology, Immunology and Parasitology, Federal University of Sao Paulo, São Paulo, Brazil
| | - Rafael Ciro Marques Cavalcante
- Department of Pharmacy and Graduate program in Applied Heath Science, Federal University of Sergipe, Lagarto, Sergipe, Brazil
| | - Cristiane Bani Corrêa
- Postgraduate Program in Health Science, Federal University of Sergipe, Aracaju, SE, Brazil
- Department of Morphology, Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - John Sidney
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
| | - Alessandro Sette
- Division of Vaccine Discovery, La Jolla Institute for Immunology, La Jolla, CA, United States
- Division of Infectious Diseases and Global Public Health, Department of Medicine, University of California, San Diego, La Jolla, CA, United States
| | - Roque Pacheco de Almeida
- Molecular Biology Laboratory, University Hospital, Department of Medicine Federal University of Sergipe, Aracaju, Sergipe, Brazil
| | - Clarisa Beatriz Palatnik-de-Sousa
- Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
- Postgraduate Program in Plant Biotechnology and Bioprocesses, Center for Health Sciences, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| |
Collapse
|
7
|
Florian DM, Bauer M, Popovitsch A, Fae I, Springer DN, Graninger M, Traugott M, Weseslindtner L, Aberle SW, Fischer G, Kundi M, Stiasny K, Zoufaly A, Landry SJ, Aberle JH. Enhanced and long-lasting SARS-CoV-2 immune memory in individuals with common cold coronavirus cross-reactive T cell immunity. Front Immunol 2025; 16:1501704. [PMID: 40191213 PMCID: PMC11968687 DOI: 10.3389/fimmu.2025.1501704] [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] [Received: 09/25/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
With the continuous emergence of novel SARS-CoV-2 variants, long-lasting and broadly reactive cellular and humoral immunity is critical for durable protection from COVID-19. We investigated SARS-CoV-2-specific T cell immunity in relation to antibodies, infection outcome and disease severity and assessed its durability in a longitudinal cohort over a three-year time course. We identified pre-existing T cells reactive to the seasonal coronavirus (CoV) OC43 that cross-react with the conserved SARS-CoV-2 spike S813-829 peptide. These cross-reactive T cells increased in frequency following SARS-CoV-2 infection or vaccination and correlated with enhanced spike-specific T cell responses and significantly reduced viral loads. Furthermore, our data revealed that CoV-cross-reactive T cells were maintained as part of the long-lasting memory response, contributing to increased T cell frequencies against omicron variants. These findings suggest a functional role of CoV-cross-reactive T cells that extends beyond the initial SARS-CoV-2 exposure, contributing to enhanced immunity against highly mutated SARS-CoV-2 variants.
Collapse
Affiliation(s)
- David M. Florian
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Michael Bauer
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | | | - Ingrid Fae
- Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | - Gottfried Fischer
- Department of Transfusion Medicine and Cell Therapy, Medical University of Vienna, Vienna, Austria
| | - Michael Kundi
- Center for Public Health, Department for Environmental Health, Medical University of Vienna, Vienna, Austria
| | - Karin Stiasny
- Center for Virology, Medical University of Vienna, Vienna, Austria
| | - Alexander Zoufaly
- Department of Medicine IV, Klinik Favoriten, Vienna, Austria
- Faculty of Medicine, Sigmund Freud University, Vienna, Austria
| | - Samuel J. Landry
- Department of Biochemistry and Molecular Biology, Tulane University School of Medicine, New Orleans, LA, United States
| | - Judith H. Aberle
- Center for Virology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Afeyan AB, Wu CJ, Oliveira G. Rapid parallel reconstruction and specificity screening of hundreds of T cell receptors. Nat Protoc 2025; 20:539-586. [PMID: 39516267 PMCID: PMC11896752 DOI: 10.1038/s41596-024-01061-4] [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: 03/05/2024] [Accepted: 08/06/2024] [Indexed: 11/16/2024]
Abstract
The ability to screen the reactivity of T cell receptors (TCRs) is essential to understanding how antigen-specific T cells drive productive or dysfunctional immune responses during infections, cancer and autoimmune diseases. Methods to profile large numbers of TCRs are critical for characterizing immune responses sustained by diverse T cell clones. Here we provide a medium-throughput approach to reconstruct dozens to hundreds of TCRs in parallel, which can be simultaneously screened against primary human tissues and broad curated panels of antigenic targets. Using Gibson assembly and miniaturized lentiviral transduction, individual TCRs are rapidly cloned and expressed in T cells; before screening, TCR cell lines undergo combinatorial labeling with dilutions of three fluorescent dyes, which allows retrieval of the identity of individual T cell effectors when they are organized and tested in pools using flow cytometry. Upon incubation with target cells, we measure the upregulation of CD137 on T cells as a readout of TCR activation. This approach is scalable and simultaneously captures the reactivity of pooled TCR cell lines, whose activation can be deconvoluted in real time, thus providing a path for screening the reactivity of dozens of TCRs against broad panels of synthetic antigens or against cellular targets, such as human tumor cells. We applied this pipeline to systematically deconvolute the antitumoral and antiviral reactivity and antigenic specificity of TCRs from human tumor-infiltrating lymphocytes. This protocol takes ~2 months, from experimental design to data analysis, and requires standard expertise in cloning, cell culture and flow cytometry.
Collapse
Affiliation(s)
- Alexander B Afeyan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Catherine J Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA.
| | - Giacomo Oliveira
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| |
Collapse
|
9
|
Liu J, Wu Y, Gao GF. A Structural Voyage Toward the Landscape of Humoral and Cellular Immune Escapes of SARS-CoV-2. Immunol Rev 2025; 330:e70000. [PMID: 39907512 DOI: 10.1111/imr.70000] [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: 12/15/2024] [Accepted: 01/08/2025] [Indexed: 02/06/2025]
Abstract
The genome-based surveillance of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in the past nearly 5 years since its emergence has refreshed our understanding of virus evolution, especially on convergent co-evolution with the host. SARS-CoV-2 evolution has been characterized by the emergence of sets of mutations that affect the functional properties of the virus by altering its infectivity, virulence, transmissibility, and interactions with host immunity. This poses a huge challenge to global prevention and control measures based on drug treatment and vaccine application. As one of the key evasion strategies in response to the immune profile of the human population, there are overwhelming amounts of evidence for the reduced antibody neutralization of SARS-CoV-2 variants. Additionally, data also suggest that the levels of CD4+ and CD8+ T-cell responses against variants or sub-variants decrease in the populations, although non-negligible cross-T-cell responses are maintained. Herein, from the perspectives of structural immunology, we outline the characteristics and mechanisms of the T cell and antibody responses to SARS-CoV and its variants/sub-variants. The molecular bases for the impact of the immune escaping variants on the interaction of the epitopes with the key receptors in adaptive immunity, that is, major histocompatibility complex (MHC), T-cell receptor (TCR), and antibody are summarized and discussed, the knowledge of which will widen our understanding of this pandemic-threatening virus and assist the preparedness for Pathogen X in the future.
Collapse
Affiliation(s)
- Jun Liu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Wu
- Department of Pathogen Microbiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - George F Gao
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
- The D. H. Chen School of Universal Health, Zhejiang University, Hangzhou, China
| |
Collapse
|
10
|
van den Dijssel J, Konijn VAL, Duurland MC, de Jongh R, Koets L, Veldhuisen B, Raaphorst H, Turksma AW, Freen‐van Heeren JJ, Steenhuis M, Rispens T, van der Schoot CE, van Ham SM, van Lier RAW, van Gisbergen KPJM, ten Brinke A, van de Sandt CE. Age and Latent Cytomegalovirus Infection Do Not Affect the Magnitude of De Novo SARS-CoV-2-Specific CD8 + T Cell Responses. Eur J Immunol 2025; 55:e202451565. [PMID: 40071711 PMCID: PMC11898545 DOI: 10.1002/eji.202451565] [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: 10/10/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 03/15/2025]
Abstract
Immunosenescence, age-related immune dysregulation, reduces immunity upon vaccinations and infections. Cytomegalovirus (CMV) infection results in declining naïve (Tnaïve) and increasing terminally differentiated (Temra) T cell populations, further aggravating immune aging. Both immunosenescence and CMV have been speculated to hamper the formation of protective T-cell immunity against novel or emerging pathogens. The SARS-CoV-2 pandemic presented a unique opportunity to examine the impact of age and/or CMV on the generation of de novo SARS-CoV-2-specific CD8+ T cell responses in 40 younger (22-40 years) and 37 older (50-66 years) convalescent individuals. Heterotetramer combinatorial coding combined with phenotypic markers were used to study 35 SARS-CoV-2 epitope-specific CD8+ T cell populations directly ex vivo. Neither age nor CMV affected SARS-CoV-2-specific CD8+ T cell frequencies, despite reduced total CD8+ Tnaïve cells in older CMV- and CMV+ individuals. Robust SARS-CoV-2-specific central memory CD8+ T (Tcm) responses were detected in younger and older adults regardless of CMV status. Our data demonstrate that immune aging and CMV status did not impact the SARS-CoV-2-specific CD8+ T cell response. However, SARS-CoV-2-specific CD8+ T cells of older CMV- individuals displayed the lowest stem cell memory (Tscm), highest Temra and PD1+ populations, suggesting that age, not CMV, may impact long-term SARS-CoV-2 immunity.
Collapse
Affiliation(s)
- Jet van den Dijssel
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Veronique A. L. Konijn
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Mariël C Duurland
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Rivka de Jongh
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Lianne Koets
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- National Screening Laboratory of SanquinResearch and Laboratory ServicesAmsterdamThe Netherlands
| | - Barbera Veldhuisen
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Department of Immunohematology DiagnosticsSanquin Diagnostic ServicesAmsterdamThe Netherlands
| | | | | | | | - Maurice Steenhuis
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - Theo Rispens
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Amsterdam UMC location Vrije Universiteit AmsterdamMolecular Cell Biology and ImmunologyAmsterdamThe Netherlands
| | - C Ellen van der Schoot
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
| | - S. Marieke van Ham
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Swammerdam Institute for Life SciencesUniversity of AmsterdamAmsterdamThe Netherlands
| | | | - Klaas P. J. M. van Gisbergen
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Physiology and Cancer Programme, Champalimaud ResearchChampalimaud FoundationLisboaPortugal
| | - Anja ten Brinke
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
| | - Carolien E. van de Sandt
- Sanquin Research and Landsteiner Laboratory, Amsterdam UMCUniversity of AmsterdamAmsterdamThe Netherlands
- Amsterdam Institute for Immunology and Infectious DiseasesAmsterdamThe Netherlands
- Department of Microbiology and ImmunologyUniversity of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneVictoriaAustralia
| |
Collapse
|
11
|
Fahnøe U, Feng S, Underwood AP, Jacobsen K, Ameri A, Blicher TH, Sølund CS, Rosenberg BR, Brix L, Weis N, Bukh J. T cell receptor usage and epitope specificity amongst CD8 + and CD4 + SARS-CoV-2-specific T cells. Front Immunol 2025; 16:1510436. [PMID: 40092978 PMCID: PMC11906682 DOI: 10.3389/fimmu.2025.1510436] [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] [Received: 10/12/2024] [Accepted: 01/02/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction The coronavirus disease 2019 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has highlighted the critical importance of understanding protective long-lasting immune responses. This study investigates the epitope specificity, T cell receptor (TCR) usage, and phenotypic changes in SARS-CoV-2-specfic CD8+ and CD4+ T cells over time in convalescent individuals with COVID-19. Methods Peripheral blood mononuclear cells (PBMCs) were collected from 28 unvaccinated individuals with primary SARS-CoV-2 infection (6 identified as the D614G variant, clade 20C) and analyzed up to 12 months post-symptom onset. Antigen-specific CD8+ and CD4+ T cells were analyzed using flow cytometry and single-cell RNA sequencing (scRNAseq) using specific dextramer and antibody reagents. TCR clonotypes and activation markers were characterized to explore T cell dynamics. Results SARS-CoV-2-specific CD8+ T cells exhibited waning frequencies long-term, transitioning from memory-like to a naïve-like state. scRNAseq revealed specificity against both spike and non-spike antigens with increased CD95 and CD127 expression over time, indicating that naïve-like T cells may represent stem cell memory T cells, which are multipotent and self-renewing, likely important for long-lived immunity. TCR clonal expansion was observed mainly in memory T cells, with overlapping TCR beta chain (TRB)-complementary determining region 3 (CDR3) sequences between participants, suggesting shared public TCR epitope-specific repertoires against SARS-CoV-2. Further, unique spike-specific CD4+ T cells with high CD95 and CD127 expression were identified, which may play a crucial role in long-term protection. Discussion This study highlights epitope-specificity heterogeneity, with some immunodominant responses, and suggests a potential role for long-lived SARS-CoV-2-specific T cell immunity. Shared TCR repertoires offers insights into cross-reactive and protective T cell clones, providing valuable information for optimizing vaccine strategies against emerging SARS-CoV-2 variants. The findings underscore the critical role of cellular immunity in long-term protection against SARS-CoV-2 and emphasizes the importance of understanding T cell dynamics.
Collapse
Affiliation(s)
- Ulrik Fahnøe
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Shan Feng
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Alexander P. Underwood
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | | | | | | | - Christina S. Sølund
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| | - Brad R. Rosenberg
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | | | - Nina Weis
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens Bukh
- Copenhagen Hepatitis C Program (CO-HEP), Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
- Copenhagen Hepatitis C Program (CO-HEP), Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Infectious Diseases, Copenhagen University Hospital, Hvidovre, Denmark
| |
Collapse
|
12
|
Meza J, Glass E, Sandhu AK, Li Y, Karanika S, Fessler K, Hui Y, Schill C, Wang T, Zhang J, Bates RE, Taylor AD, Kapoor AR, Ayeh SK, Karakousis PC, Markham RB, Gordy JT. Novel Vaccines Targeting the Highly Conserved SARS-CoV-2 ORF3a Ectodomain Elicit Immunogenicity in Mouse Models. Vaccines (Basel) 2025; 13:220. [PMID: 40266087 PMCID: PMC11946519 DOI: 10.3390/vaccines13030220] [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: 01/04/2025] [Revised: 02/16/2025] [Accepted: 02/20/2025] [Indexed: 04/24/2025] Open
Abstract
BACKGROUND The majority of antigen-based SARS-CoV-2 (SCV2) vaccines utilized in the clinic have had the Spike protein or domains thereof as the immunogen. While the Spike protein is highly immunogenic, it is also subject to genetic drift over time, which has led to a series of variants of concern that continue to evolve, requiring yearly updates to the vaccine formulations. In this study, we investigate the potential of the N-terminal ectodomain of the ORF3a protein encoded by the orf3a gene of SCV2 to be an evolution-resistant vaccine antigen. This domain is highly conserved over time, and, unlike many other SCV2 conserved proteins, it is present on the exterior of the virion, making it accessible to antibodies. ORF3a is also important for eliciting robust anti-SARS-CoV-2 T-cell responses. METHODS We designed a DNA vaccine by fusing the N-terminal ectodomain of orf3a to macrophage-inflammatory protein 3α (MIP3α), which is a chemokine utilized in our laboratory that enhances vaccine immunogenicity by targeting an antigen to its receptor CCR6 present on immature dendritic cells. The DNA vaccine was tested in mouse immunogenicity studies, vaccinating by intramuscular (IM) electroporation and by intranasal (IN) with CpG adjuvant administrations. We also tested a peptide vaccine fusing amino acids 15-28 of the ectodomain to immunogenic carrier protein KLH, adjuvanted with Addavax. RESULTS The DNA IM route was able to induce 3a-specific splenic T-cell responses, showing proof of principle that the region can be immunogenic. The DNA IN route further showed that we could induce ORF3a-specific T-cell responses in the lung, which are critical for potential disease mitigation. The peptide vaccine elicited a robust anti-ORF3a antibody response systemically, as well as in the mucosa of the lungs and sinus cavity. CONCLUSIONS These studies collectively show that this evolutionarily stable region can be targeted by vaccination strategies, and future work will test if these vaccines, alone or in combination, can result in reduced disease burden in animal challenge models.
Collapse
Affiliation(s)
- Jacob Meza
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - Elizabeth Glass
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - Avinaash K. Sandhu
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - Yangchen Li
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - Styliani Karanika
- Center for Tuberculosis Research, Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (S.K.); (A.R.K.); (S.K.A.)
| | - Kaitlyn Fessler
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - Yinan Hui
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - Courtney Schill
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - Tianyin Wang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - Jiaqi Zhang
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - Rowan E. Bates
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - Alannah D. Taylor
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - Aakanksha R. Kapoor
- Center for Tuberculosis Research, Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (S.K.); (A.R.K.); (S.K.A.)
| | - Samuel K. Ayeh
- Center for Tuberculosis Research, Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (S.K.); (A.R.K.); (S.K.A.)
| | - Petros C. Karakousis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
- Center for Tuberculosis Research, Division of Infectious Diseases, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (S.K.); (A.R.K.); (S.K.A.)
| | - Richard B. Markham
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| | - James T. Gordy
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA; (J.M.); (A.K.S.); (Y.L.); (K.F.); (Y.H.); (C.S.); (T.W.); (J.Z.); (R.E.B.); (A.D.T.); (P.C.K.)
| |
Collapse
|
13
|
Ding Y, Yan J, Huang L, Yu J, Wu Y, Shen C, Fang A. Large-scale screening of HIV-1 T-cell epitopes restricted by 12 prevalent HLA-A allotypes in Northeast Asia and universal detection of HIV-1-specific CD8 + T cells. Front Microbiol 2025; 16:1529721. [PMID: 40008047 PMCID: PMC11850406 DOI: 10.3389/fmicb.2025.1529721] [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: 11/17/2024] [Accepted: 01/24/2025] [Indexed: 02/27/2025] Open
Abstract
Background Although the immune response of host T cells to human immunodeficiency virus (HIV) significantly influences the progression of the infection, the development of T-cell-based vaccines and therapies, as well as the clinical evaluation of specific T-cell functions, is currently markedly hindered by the absence of broad-spectrum, functionally validated HIV T-cell epitopes that account for the polymorphisms of the human leukocyte antigen (HLA) within an indicated geographic population. This study aimed to identify T-cell epitopes derived from the GP160, GAG, and POL proteins of the HIV-1 strain, specifically linked to 12 prevalent HLA-A allotypes, that collectively represent approximately 91% of the total gene frequency in Northeast Asian populations. Methods A total of 134 epitopes were predicted in silico and selected as potential candidates for further validation. Subsequently, peripheral blood mononuclear cells (PBMCs) were collected from 96 individuals with HIV-1 and cocultured ex vivo with each epitope candidate peptide, followed by the detection of activated CD8+ T cells. Peripheral blood mononuclear cells (PBMCs) were collected from 96 individuals with HIV-1 and cocultured ex vivo with each candidate peptide epitope, followed by the detection of activated CD8+ T cells. A total of 69 epitopes were validated as real-world HIV T-cell epitopes presented by 12 dominant HLA-A allotypes. Furthermore, the HLA-A cross-restriction for each epitope candidate was identified through peptide competitive binding assays using 12 transfected HMy2.CIR cell lines. Results A total of 45 epitopes demonstrated high affinity, while 31 epitopes displayed intermediate affinity. A broad-spectrum CD8+ T-cell epitope library containing 141 validated epitope peptides was used to universally detect HIV-1-specific CD8+ T cells via peptide-PBMC ex vivo coculture and intracellular IFN-γ staining. In 52 people with HIV-1, the number of reactive HIV-1 specific CD8+ T cells was significantly higher in the CD4+ T-cell-high patient group compared to the CD4+ T-cell-low patient group, and it correlated with the CD4+ T-cell-low patient group (<200/μL). Conclusion This study provides a broad-spectrum CD8+ T-cell epitope library aimed at developing a T-cell-directed HIV vaccine that offers high population coverage in Northeast Asia. In addition, it establishes a universal detection method for the clinical assessment of HIV-1-specific CD8+ T-cell responses.
Collapse
Affiliation(s)
- Yan Ding
- Department of Clinical Laboratory, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jialai Yan
- School of Medical Technology, Anhui Medical College, Hefei, Anhui, China
| | - Ling Huang
- Department of Clinical Laboratory, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jinhong Yu
- Department of Clinical Laboratory, The Second Hospital of Nanjing, Affiliated to Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Yandan Wu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Chuanlai Shen
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, Jiangsu, China
| | - Anning Fang
- School of Basic Medicine, Anhui Medical College, Hefei, Anhui, China
| |
Collapse
|
14
|
Zhou M, Xiao H, Yang X, Cheng T, Yuan L, Xia N. Novel vaccine strategies to induce respiratory mucosal immunity: advances and implications. MedComm (Beijing) 2025; 6:e70056. [PMID: 39830020 PMCID: PMC11739453 DOI: 10.1002/mco2.70056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 10/31/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025] Open
Abstract
Rapid advances in vaccine technology are becoming increasingly important in tackling global health crises caused by respiratory virus infections. While traditional vaccines, primarily administered by intramuscular injection, have proven effective, they often fail to provide the broad upper respiratory tract mucosal immunity, which is urgently needed for first-line control of respiratory viral infections. Furthermore, traditional intramuscular vaccines may not adequately address the immune escape of emerging virus variants. In contrast, respiratory mucosal vaccines developed using the body's mucosal immune response mechanism can simultaneously establish both systemic and mucosal immunity. This dual action effectively allows the respiratory mucosal immune system to function as the first line of defense, preventing infections at the entry points. This review highlights the efficacy of respiratory mucosal vaccines, including innovative delivery methods such as nasal and oral formulations, in enhancing local and systemic immune barriers. Notably, respiratory mucosal vaccines offer potential advantages in protecting against emerging virus variants and maintaining long-term and multidimensional immune memory in the upper respiratory tract. In addition, a combination of intramuscular and respiratory mucosal delivery of vaccines largely improves their coverage and effectiveness, providing valuable insights for future vaccine development and public inoculation strategies.
Collapse
Affiliation(s)
- Ming Zhou
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Haiqin Xiao
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Xinyi Yang
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Tong Cheng
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Lunzhi Yuan
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| | - Ningshao Xia
- State Key Laboratory of Vaccines for Infectious DiseasesNational Institute of Diagnostics and Vaccine Development in Infectious DiseasesXiang An Biomedicine LaboratorySchool of Life Sciences & School of Public HealthXiamen UniversityXiamenFujianChina
| |
Collapse
|
15
|
He R, Meng L, Sun Y, Wang J, Wang S, Liu Y, Fei L, Wang Z, Zhang Q, Wu Y, Chen Y, Diao B. Screening for immunodominant epitopes of SARS-CoV-2 based on CD8 + T cell responses from individuals with HLA-A homozygous alleles. Mol Immunol 2025; 178:52-63. [PMID: 39864283 DOI: 10.1016/j.molimm.2025.01.010] [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: 12/09/2024] [Revised: 01/15/2025] [Accepted: 01/19/2025] [Indexed: 01/28/2025]
Abstract
PURPOSE SARS-CoV-2-specific CD8+ cytotoxic T lymphocytes (CTLs) are crucial in viral clearance, disease progression, and reinfection control. However, numerous SARS-CoV-2 immunodominant CTL epitopes theoretically are still unidentified due to the genetic polymorphism of human leukocyte antigen class I (HLA-I) molecules. METHODS The CTL epitopes of SARS-CoV-2 were predicted by the epitope affinity and immunogenicity prediction platforms: the NetMHCpan and the PromPPD. Individuals with HLA-A homozygous alleles were screened from 252 COVID-19 vaccinees, including the Ad5-nCoV vaccine (CanSino, n = 183) and the CoronaVac inactivated vaccine (Sinovac, n = 69) using MiSeqDx™ generation sequencing, and their PBMCs were further stimulated by the predicted peptides to screen the immunodominant epitopes according to the secretion of IFN-γ from CD8+ T cells. Peptide-MHC tetramers were constructed and used to detect the frequency of antigen specific CTLs in vivo. RESULTS Individuals with HLA-A homozygous alleles including HLA-A*01 (n = 1), -A*02 (n = 9), - A*03 and -A*11 (n = 12), and -A*24 (n = 7) supertypes were selected. Twelve immunodominant CTL epitopes for these HLA-A allotypes were finally screened based on the frequency of IFN-γ+CD8+ T cells in homozygous individuals. The SARS-CoV-2 specific CTLs from Omicron variant infected patients were successfully evaluated by these novel peptide-HLA tetramers. CONCLUSION A set of immunodominant CTL epitopes of SARS-CoV-2 was identified, and the antigen-specific CD8+ T cells in viral infected patients or COVID-19 vaccinees could be rapidly detected by a mixture of the peptide-MHC tetramers.
Collapse
Affiliation(s)
- Rui He
- School of Medicine, Chongqing University, Chongqing 400030, PR China; Institute of Immunology, PLA, The Army Medical University, Chongqing 400038, PR China
| | - Lingxin Meng
- Institute of Immunology, PLA, The Army Medical University, Chongqing 400038, PR China
| | - Yuting Sun
- Institute of Immunology, PLA, The Army Medical University, Chongqing 400038, PR China
| | - Jingsong Wang
- Institute of Immunology, PLA, The Army Medical University, Chongqing 400038, PR China
| | - Shufeng Wang
- Institute of Immunology, PLA, The Army Medical University, Chongqing 400038, PR China
| | - Yueping Liu
- Institute of Immunology, PLA, The Army Medical University, Chongqing 400038, PR China; Department of Medical Laboratory Center, General Hospital of Central Theater Command, Wuhan, Hubei 430015, PR China
| | - Lei Fei
- Institute of Immunology, PLA, The Army Medical University, Chongqing 400038, PR China
| | - Zhongfang Wang
- State Key Laboratory of Respiratory Disease & National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, Guangdong 511436, PR China
| | - Qinggao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, PR China
| | - Yuzhang Wu
- Institute of Immunology, PLA, The Army Medical University, Chongqing 400038, PR China
| | - Yongwen Chen
- Institute of Immunology, PLA, The Army Medical University, Chongqing 400038, PR China; Chronic Disease Research Center, Medical College, Dalian University, Dalian, Liaoning 116622, PR China.
| | - Bo Diao
- Department of Medical Laboratory Center, General Hospital of Central Theater Command, Wuhan, Hubei 430015, PR China.
| |
Collapse
|
16
|
Zhao Y, He C, Peng M, Li M, Liu X, Han X, Fu Q, Wu Y, Yue F, Yan C, Zhao G, Shen C. Large-Scale Screening of CD4 + T-Cell Epitopes From SARS-CoV-2 Proteins and the Universal Detection of SARS-CoV-2 Specific T Cells for Northeast Asian Population. J Med Virol 2025; 97:e70241. [PMID: 39977358 DOI: 10.1002/jmv.70241] [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: 10/10/2024] [Revised: 12/09/2024] [Accepted: 01/25/2025] [Indexed: 02/22/2025]
Abstract
The polymorphism of human leukocyte antigens in the Northeast Asian populations and the lack of broad-spectrum T-cell epitopes covering this cohort markedly limited the development of T cell-directed vaccines against SARS-CoV-2 infection, and also hampered the universal detection of SARS-CoV-2 specific T cells. In this study, 93 CD4+ T-cell epitopes restricted by 12 prevalent HLA-DRB1 allotypes, which covering over 80% Chinese and Northeast Asian populations, were identified from the S, E, M, N and RdRp proteins of SARS-CoV-2 by in silico prediction, DC-peptide-PBL coculture experiment, and immunization in HLA-A2/DR1 transgenic mice. Furthermore, by using validated 215 CD8+ T cell epitope peptides and 123 CD4+ T-cell epitope peptides covering Northeast Asian cohort, the universal ELISpot detection systems of SARS-CoV-2 specific CD8+ T cells and CD4+ T cells were established, for the first time, and followed by the tests for 50 unexposed and 100 convalescent samples. The median of spot-forming units for CD8+ T cells and CD4+ T cells were 68 and 15, respectively, in the unexposed donors, but were 137 and 52 in the convalescent donors 6 months after recovery while 128 and 47 in the convalescent donors 18 months after recovery. This work initially provided the broad-spectrum CD4+ T-cell epitope library of SARS-CoV-2 for the design of T cell-directed vaccines and the universal T cell detection tool tailoring to Northeast Asian population, and confirmed the long-term memory T cell immunity after SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Chengtao He
- Nanjing Red Cross Blood Center, Nanjing, China
| | - Min Peng
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Min Li
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Xiaotao Liu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Xuelian Han
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Qiang Fu
- Nanjing Red Cross Blood Center, Nanjing, China
| | - Yandan Wu
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Fangping Yue
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Chunguang Yan
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| | - Guangyu Zhao
- State Key Laboratory of Pathogen and Biosecurity, Academy of Military Medical Sciences, Beijing, China
- Laboratory of Advanced Biotechnology, Academy of Military Medical Sciences, Beijing, China
| | - Chuanlai Shen
- Department of Microbiology and Immunology, Medical School of Southeast University, Nanjing, China
| |
Collapse
|
17
|
Ramirez SI, Lopez PG, Faraji F, Parikh UM, Heaps A, Ritz J, Moser C, Eron JJ, Wohl D, Currier J, Daar ES, Greninger A, Klekotka P, Grifoni A, Weiskopf D, Sette A, Peters B, Hughes MD, Chew KW, Smith DM, Crotty S. Early antiviral CD4+ and CD8+ T cells are associated with upper airway clearance of SARS-CoV-2. JCI Insight 2024; 9:e186078. [PMID: 39704169 DOI: 10.1172/jci.insight.186078] [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: 08/19/2024] [Accepted: 10/25/2024] [Indexed: 12/21/2024] Open
Abstract
T cells are involved in protective immunity against numerous viral infections. Data regarding functional roles of human T cells in SARS-CoV-2 (SARS2) viral clearance in primary COVID-19 are limited. To address this knowledge gap, we assessed samples for associations between SARS2 upper respiratory tract viral RNA levels and early virus-specific adaptive immune responses for 95 unvaccinated clinical trial participants with acute primary COVID-19 aged 18-86 years old, approximately half of whom were considered at high risk for progression to severe COVID-19. Functionality and magnitude of acute SARS2-specific CD4+ and CD8+ T cell responses were evaluated, in addition to antibody responses. Most individuals with acute COVID-19 developed SARS2-specific T cell responses within 6 days of COVID-19 symptom onset. Early CD4+ T cell and CD8+ T cell responses were polyfunctional, and both strongly associated with reduced upper respiratory tract SARS2 viral RNA, independent of neutralizing antibody titers. Overall, these findings provide evidence for protective roles for circulating SARS2-specific CD4+ and CD8+ T cells during acute COVID-19.
Collapse
Affiliation(s)
- Sydney I Ramirez
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| | - Paul G Lopez
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Farhoud Faraji
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Otolaryngology-Head and Neck Surgery, UCSD, La Jolla, California, USA
| | - Urvi M Parikh
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Amy Heaps
- Division of Infectious Diseases, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Justin Ritz
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Carlee Moser
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Joseph J Eron
- Department of Medicine, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - David Wohl
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Judith Currier
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Eric S Daar
- Lundquist Institute at Harbor-UCLA Medical Center, Torrance, California, USA
| | - Alex Greninger
- Department of Laboratory Medicine and Pathology, University of Washington Medical Center, Seattle, Washington, USA
| | | | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Daniela Weiskopf
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| | - Bjoern Peters
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| | - Michael D Hughes
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Kara W Chew
- Division of Infectious Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Davey M Smith
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| | - Shane Crotty
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, California, USA
- Division of Infectious Diseases and Global Public Health, Department of Medicine, and
| |
Collapse
|
18
|
Pinto PBA, Timis J, Chuensirikulchai K, Li QH, Lu HH, Maule E, Nguyen M, Alves RPDS, Verma SK, Ana-Sosa-Batiz F, Valentine K, Landeras-Bueno S, Kim K, Hastie K, Saphire EO, Alves A, Elong Ngono A, Shresta S. Co-immunization with spike and nucleocapsid based DNA vaccines for long-term protective immunity against SARS-CoV-2 Omicron. NPJ Vaccines 2024; 9:252. [PMID: 39702529 PMCID: PMC11659323 DOI: 10.1038/s41541-024-01043-3] [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: 03/06/2024] [Accepted: 12/01/2024] [Indexed: 12/21/2024] Open
Abstract
The continuing evolution of SARS-CoV-2 variants challenges the durability of existing spike (S)-based COVID-19 vaccines. We hypothesized that vaccines composed of both S and nucleocapsid (N) antigens would increase the durability of protection by strengthening and broadening cellular immunity compared with S-based vaccines. To test this, we examined the immunogenicity and efficacy of wild-type SARS-CoV-2 S- and N-based DNA vaccines administered individually or together to K18-hACE2 mice. S, N, and S + N vaccines all elicited polyfunctional CD4+ and CD8+ T cell responses and provided short-term cross-protection against Beta and Omicron BA.2 variants, but only co-immunization with S + N vaccines provided long-term protection against Omicron BA.2. Depletion of CD4+ and CD8+ T cells reduced the long-term efficacy, demonstrating a crucial role for T cells in the durability of protection. These findings underscore the potential to enhance long-lived protection against SARS-CoV-2 variants by combining S and N antigens in next-generation COVID-19 vaccines.
Collapse
Affiliation(s)
- Paolla Beatriz Almeida Pinto
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Laboratory of Biotechnology and Physiology of Viral Infections, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Julia Timis
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Kantinan Chuensirikulchai
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Qin Hui Li
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Hsueh Han Lu
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Erin Maule
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Michael Nguyen
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | | | | | | | - Kristen Valentine
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Sara Landeras-Bueno
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- University Cardenal Herrera-CEU, CEU Universities, Valencia, 46113, Spain
| | - Kenneth Kim
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Microscopy and Histology Core Facility, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Kathryn Hastie
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
| | - Erica Ollmann Saphire
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, 92093, USA
| | - Ada Alves
- Laboratory of Biotechnology and Physiology of Viral Infections, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, 21040-900, Brazil
| | - Annie Elong Ngono
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA.
| | - Sujan Shresta
- Center for Vaccine Innovation, La Jolla Institute for Immunology, La Jolla, 92037, USA.
- Department of Pediatrics, Division of Host-Microbe Systems and Therapeutics, University of California San Diego, La Jolla, 92093, USA.
| |
Collapse
|
19
|
Genova SN, Pencheva MM, Abadjieva TI, Atanasov NG. Cellular and immune response in fatal COVID-19 pneumonia. Pan Afr Med J 2024; 49:130. [PMID: 40190436 PMCID: PMC11971930 DOI: 10.11604/pamj.2024.49.130.45739] [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] [Received: 10/27/2024] [Accepted: 12/03/2024] [Indexed: 04/09/2025] Open
Abstract
Introduction the severity of COVID-19, causing fatal pneumonia, acute respiratory distress syndrome (ARDS), and thrombotic complications, is linked to intense inflammation. Elevated CD4+ and CD8+ cells in the lungs indicate harmful inflammation in severe cases. This study investigates immune responses in lung tissues of deceased patients across different stages of COVID-19 pneumonia. Methods lung tissues from 160 fatal COVID-19 cases, diagnosed via Real-Time RT-PCR, were histologically analyzed to identify pneumonia stages. Inflammatory cell counts were assessed immunohistochemically. Non-parametric tests analyzed categorical variables, while regression analysis evaluated relationships between continuous variables. Results the average patient age was 68.1 years (± 12.6). Microscopic analysis identified four pneumonia stages. CD4+, CD68 (macrophages), and IgG4 levels peaked by day 14, with notable elevation within seven days of symptom onset. CD4+ levels were significantly lower in DAD pneumonia (49.4% ± 15.7%) compared to ARDS (66.4% ± 19.3%) and thrombosis (70.2% ± 28.9%) (p < 0.05). Male patients had higher CD4+ values (68.5% ± 21.1%) than females (56.9% ± 22.4%) (p < 0.05). B cells (CD20) and NK cells were depleted across all stages. IgG4 expression reached 80-90% in acute phases but was nearly absent during organization and fibrosis stages. Conclusion a sharp decline in CD4+ and CD8+ during acute pneumonia and sepsis reflects immune exhaustion, while their elevation in ARDS and thrombosis likely triggers cytokine storms, causing severe lung damage. Elevated IgG4 levels in acute lung tissue correlate with fatal outcomes in severe COVID-19.
Collapse
Affiliation(s)
- Sylvia Nikolaeva Genova
- Department of General and Clinical Pathology, Medical Faculty, Medical University Plovdiv, Plovdiv, Bulgaria
- St George University Hospital, Plovdiv, Bulgaria
| | - Mina Miroslavova Pencheva
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Tsvetana Ivanova Abadjieva
- St George University Hospital, Plovdiv, Bulgaria
- Department of Dermatology and Venereology, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Nikolay Georgiev Atanasov
- Department of Health Management and Health Economics, Faculty of Public Health, Medical University Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
20
|
López D, Zumárraga J. Bioinformatic Tools for Studying the Cellular Immune Response to SARS-CoV-2, Vaccine Efficacy, and Future Pandemics at the Global Population Level. Int J Mol Sci 2024; 25:13477. [PMID: 39769240 PMCID: PMC11678114 DOI: 10.3390/ijms252413477] [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: 11/20/2024] [Revised: 12/13/2024] [Accepted: 12/15/2024] [Indexed: 01/11/2025] Open
Abstract
Antigen recognition by human leukocyte antigen (HLA) restriction is critical for an adequate antiviral response in both natural infection and vaccination. However, the overwhelming polymorphism of HLA, with nearly 40,000 alleles identified, is an important limitation for the global analysis of cellular immune responses and vaccine efficacy. In this narrative review, we included several immunoinformatics studies performed in our laboratory to circumvent this limitation. These analyses focused on studying the cellular immune responses restricted by the most common HLA alleles, and their role in vaccine efficacy. Computational studies validated experimentally, such as our laboratory has carried out, represent a useful, rapid, and cost-effective strategy to combat future pandemics.
Collapse
Affiliation(s)
- Daniel López
- Centro Nacional de Microbiología, Instituto de Salud Carlos III, 28220 Majadahonda, Madrid, Spain;
| | | |
Collapse
|
21
|
Lee Y, Tarke A, Grifoni A. In-depth characterization of T cell responses with a combined Activation-Induced Marker (AIM) and Intracellular Cytokine Staining (ICS) assay. OXFORD OPEN IMMUNOLOGY 2024; 5:iqae014. [PMID: 39713046 PMCID: PMC11661976 DOI: 10.1093/oxfimm/iqae014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024] Open
Abstract
Since T cells are key mediators in the adaptive immune system, evaluating antigen-specific T cell immune responses is pivotal to understanding immune function. Commonly used methods for measuring T cell responses include Activation-Induced Marker (AIM) assays and Intracellular Cytokine Staining (ICS). However, combining these approaches has rarely been reported. This study describes a combined AIM + ICS assay and the effect of collecting the supernatant. Peripheral blood mononuclear cells (PBMCs) from seven healthy donors were stimulated with DMSO (negative control), Epstein-Barr virus (EBV) peptide pools, and PHA (positive control). The AIM markers OX40 + CD137+ were used for CD4+ T cells and CD69 + CD137+ and CD107a + CD137+ for CD8+ T cells. Cytokine-secreting cells were identified as CD40L+ cytokine+ for CD4+ and CD69+ or CD107 + cytokine+ for CD8+ T cells. Half of the supernatant was collected before adding the BFA/Monensin/CD137 antibody solution to assess the impact on T cell responses. The CD107a + CD137+ AIM markers combination had a lower background than CD69 + CD137+, making CD107a+ a more sensitive marker for CD8+ AIM markers. Collecting half of the supernatant did not significantly affect the immune responses. Our AIM + ICS combined protocol enables the simultaneous assessment of activation and cytokine release reducing the sample volume for testing T cell responses. We also show that collecting half of the supernatant does not significantly interfere with immune responses detection.
Collapse
Affiliation(s)
- Yeji Lee
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, United States
| | - Alison Tarke
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, United States
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, United States
| |
Collapse
|
22
|
Hurst JR, Naghibosadat M, Budowski P, Liu J, Samaan P, Budiman F, Kurtesi A, Qi F, Menon H, Krishnan R, Abioye J, Gingras AC, Ostrowski M, Orozco NM, Kozak RA. Comparison of a SARS-CoV-2 mRNA booster immunization containing additional antigens to a spike-based mRNA vaccine against Omicron BA.5 infection in hACE2 mice. PLoS One 2024; 19:e0314061. [PMID: 39625929 PMCID: PMC11614295 DOI: 10.1371/journal.pone.0314061] [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: 05/03/2024] [Accepted: 11/04/2024] [Indexed: 12/06/2024] Open
Abstract
The emergence of SARS-CoV-2 variants presents challenges to vaccine effectiveness, underlining the necessity for next-generation vaccines with multiple antigens beyond the spike protein. Here, we investigated a multiantigenic booster containing spike and a chimeric construct composed of nucleoprotein (N) and membrane (M) proteins, comparing its efficacy to a spike-only booster against Omicron BA.5 in K18-hACE2 mice. Initially, mice were primed and boosted with Beta (B.1.351) spike-only mRNA, showing strong spike-specific T cell responses and neutralizing antibodies, albeit with limited cross-neutralization to Omicron variants. Subsequently, a spike-NM multiantigenic vaccine was then examined as a second booster dose for protection in hACE2-transgenic mice. Mice receiving either homologous spike-only or heterologous spike-NM booster had nearly complete inhibition of infectious virus shedding in oral swabs and reduced viral burdens in both lung and nasal tissues following BA.5 challenge. Examination of lung pathology further revealed that both spike-only and spike-NM boosters provided comparable protection against inflammatory infiltrates and fibrosis. Moreover, the spike-NM booster demonstrated neutralization efficacy in a pseudovirus assay against Wuhan-Hu-1, Beta, and Omicron variants akin to the spike-only booster. These findings indicate that supplementing spike with additional SARS-CoV-2 targets in a booster immunization confers equivalent immunity and protection against Omicron BA.5. This work highlights a promising strategy for individuals previously vaccinated with spike-only vaccines, potentially offering enhanced protection against emerging coronaviruses.
Collapse
Affiliation(s)
- Jacklyn R. Hurst
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Maedeh Naghibosadat
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Patrick Budowski
- Institute of Medical Sciences, University of Toronto, Ontario, Canada
| | - Jun Liu
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | - Philip Samaan
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | - Frans Budiman
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Alexandra Kurtesi
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Fredo Qi
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
| | - Haritha Menon
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | | | - Jumai Abioye
- Providence Therapeutics Holdings, Inc., Calgary, AB, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Sinai Health System, Toronto, ON, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Mario Ostrowski
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | | | - Robert A. Kozak
- Biological Sciences Platform, Sunnybrook Research Institute at Sunnybrook Health Sciences Centre, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
- Division of Microbiology, Sunnybrook Health Sciences Centre, Department of Laboratory Medicine and Molecular Diagnostics, Toronto, ON, Canada
| |
Collapse
|
23
|
Abt ER, Lam AK, Noguchi M, Rashid K, McLaughlin J, Teng PL, Tran W, Cheng D, Nesterenko PA, Mao Z, Creech AL, Burton Sojo G, Jeyachandran AV, Tam YK, Henley JE, Comai L, Pardi N, Arumugaswami V, Witte ON, Radu CG, Wu TT. Staggered immunization with mRNA vaccines encoding SARS-CoV-2 polymerase or spike antigens broadens the T cell epitope repertoire. Proc Natl Acad Sci U S A 2024; 121:e2406332121. [PMID: 39589869 PMCID: PMC11626164 DOI: 10.1073/pnas.2406332121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 10/16/2024] [Indexed: 11/28/2024] Open
Abstract
Combining a T cell-targeting mRNA vaccine encoding the conserved SARS-CoV-2 RNA-dependent RNA polymerase, RdRp, with a Spike-encoding mRNA vaccine may offer an additional pathway toward COVID-19 protection. Here, we show that a nucleoside-modified RdRp mRNA vaccine raises robust and durable CD8+ T cell responses in mice. Immunization drives a CD8+ T cell response enriched toward a specific RdRp epitope. Unexpectedly, coadministration of mRNA vaccines encoding RdRp or the Spike Receptor Binding Domain (RBD) dampens RBD-specific immune responses. Contralateral administration reduces the suppression of RBD-specific T cell responses while type I interferon signaling blockade restores RBD-specific antibodies. A staggered immunization strategy maintains both RBD vaccine-mediated antibody and T cell responses as well as protection against lethal SARS-CoV-2 challenge in human ACE2 transgenic mice. In HLA-A2.1 transgenic mice, the RdRp vaccine elicits CD8+ T cell responses against HLA-A*02:01-restricted epitopes recognized by human donor T cells. These results highlight RdRp as a candidate antigen for COVID-19 vaccines. The findings also offer insights into crafting effective multivalent mRNA vaccines to broaden CD8+ T cell responses against SARS-CoV-2 and potentially other viruses with pandemic potential.
Collapse
Affiliation(s)
- Evan R. Abt
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Alex K. Lam
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Miyako Noguchi
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Khalid Rashid
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Jami McLaughlin
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Pu-Lin Teng
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Wendy Tran
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Donghui Cheng
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Pavlo A. Nesterenko
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Zhiyuan Mao
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Amanda L. Creech
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Giselle Burton Sojo
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
| | - Arjit Vijey Jeyachandran
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
| | - Ying K. Tam
- Acuitas Therapeutics, Vancouver, BCV6T 1Z3, Canada
| | - Jill E. Henley
- Department of Molecular Microbiology and Immunology, The Hastings and Wright Laboratories, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
| | - Lucio Comai
- Department of Molecular Microbiology and Immunology, The Hastings and Wright Laboratories, Keck School of Medicine, University of Southern California, Los Angeles, CA90089
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA19104
| | | | - Owen N. Witte
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Department of Microbiology, Immunology, and Molecular Genetics, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
- Molecular Biology Institute, University of California Los Angeles, Los Angeles, CA90095
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA90095
- Parker Institute for Cancer Immunotherapy, University of California Los Angeles, Los Angeles, CA90095
| | - Caius G. Radu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
| | - Ting-Ting Wu
- Department of Molecular and Medical Pharmacology, University of California Los Angeles, Los Angeles, CA90095
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA90095
- AIDS Institute, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA90095
| |
Collapse
|
24
|
Zhou D, Luo Y, Ma Q, Xu Y, Yao X. The characteristics of TCR CDR3 repertoire in COVID-19 patients and SARS-CoV-2 vaccine recipients. Virulence 2024; 15:2421987. [PMID: 39468707 PMCID: PMC11540089 DOI: 10.1080/21505594.2024.2421987] [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: 08/08/2024] [Revised: 09/28/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024] Open
Abstract
The COVID-19 pandemic and large-scale administration of multiple SARS-CoV-2 vaccines have attracted global attention to the short-term and long-term effects on the human immune system. An analysis of the "traces" left by the body's T-cell immune response is needed, especially for the prevention and treatment of breakthrough infections and long COVID-19 and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variant infections. T-cell receptor complementarity determining region 3 (TCR CDR3) repertoire serves as a target molecule for monitoring the effects, mechanisms, and memory of the T-cell response. Furthermore, it has been extensively applied in the elucidation of the infectious mechanism and vaccine refinement of hepatitis B virus (HBV), influenza virus, human immunodeficiency virus (HIV), and SARS-CoV. Laboratories worldwide have utilized high-throughput sequencing (HTS) and scTCR-seq to characterize, share, and apply the TCR CDR3 repertoire in COVID-19 patients and SARS-CoV-2 vaccine recipients. This article focuses on the comparative analysis of the diversity, clonality, V&J gene usage and pairing, CDR3 length, shared CDR3 sequences or motifs, and other characteristics of TCR CDR3 repertoire. These findings provide molecular targets for evaluating T-cell response effects and short-term and long-term impacts on the adaptive immune system following SARS-CoV-2 infection or vaccination and establish a comparative archive of T-cell response "traces."
Collapse
Affiliation(s)
- Dewei Zhou
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
- Department of Clinical Laboratory, The First People’s Hospital of Zunyi (The Third Affiliated Hospital of Zunyi Medical University), Zunyi, China
| | - Yan Luo
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Qingqing Ma
- Department of Central Laboratory, Guizhou Aerospace Hospital, Zunyi, China
| | - Yuanyuan Xu
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| | - Xinsheng Yao
- Department of Immunology, Center of Immunomolecular Engineering, Innovation & Practice Base for Graduate Students Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
25
|
Lobaina Y, Musacchio A, Ai P, Chen R, Suzarte E, Chinea G, Zhang M, Zhou Z, Lan Y, Silva R, Guillén G, Yang K, Li W, Perera Y, Hermida L. Obtaining HBV core protein VLPs carrying SARS-CoV-2 nucleocapsid conserved fragments as vaccine candidates. Virol J 2024; 21:310. [PMID: 39609857 PMCID: PMC11606075 DOI: 10.1186/s12985-024-02583-9] [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: 07/15/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024] Open
Abstract
The Hepatitis B core antigen (HBcAg) has been used as a carrier of several heterologous protein fragments based on its capacity to form virus-like particles (VLPs) and to activate innate and adaptive immune responses. In the present work, two chimeric proteins were designed as potential pancorona vaccine candidates, comprising the N- or C- terminal domain of SARS-CoV-2 nucleocapsid (N) protein fused to HBcAg. The recombinant proteins, obtained in E. coli, were named CN-1 and CND-1, respectively. The final protein preparations were able to form 10-25 nm particles, visualized by TEM. Both proteins were recognized by sera from COVID-19 convalescent donors; however, the antigenicity of CND-1 tends to be higher. The immunogenicity of both proteins was studied in Balb/C mice by intranasal route without adjuvant. After three doses, only CND-1 elicited a positive immune response, systemic and mucosal, against SARS-CoV-2 N protein. CND-1 was evaluated in a second experiment mixed with the CpG ODN-39 M as nasal adjuvant. The induced anti-N immunity was significantly enhanced, and the antibodies generated were cross-reactive with N protein from Omicron variant, and SARS-CoV-1. Also, an anti-N broad cellular immune response was detected in spleen, by IFN-γ ELISpot. The nasal formulation composed by CND-1 and ODN-39 M constitutes an attractive component for a second generation coronavirus vaccine, increasing the scope of S protein-based vaccines, by inducing mucosal immunity and systemic broad humoral and cellular responses against Sarbecovirus N protein.
Collapse
Affiliation(s)
- Yadira Lobaina
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC) Lengshuitan District, Yongzhou City, 425000, Hunan, China
- R&D Department, Yongzhou Zhong Gu Biotechnology Co., Ltd., Yangjiaqiao Street, Lengshuitan District, Yongzhou City, 425000, Hunan, China
| | - Alexis Musacchio
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC) Lengshuitan District, Yongzhou City, 425000, Hunan, China
- R&D Department, Yongzhou Zhong Gu Biotechnology Co., Ltd., Yangjiaqiao Street, Lengshuitan District, Yongzhou City, 425000, Hunan, China
- Research Department, Center for Genetic Engineering and Biotechnology (CIGB), 10600, Havana, Cuba
| | - Panchao Ai
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC) Lengshuitan District, Yongzhou City, 425000, Hunan, China
- Yongzhou Development and Construction Investment Co. Ltd. (YDCI), Yongzhou Economic and Technological Development Zone, Changfeng Industry Park, No. 1 Liebao Road, Lengshuitan District, Yongzhou City, Hunan Province, China
| | - Rong Chen
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC) Lengshuitan District, Yongzhou City, 425000, Hunan, China
- Yongzhou Development and Construction Investment Co. Ltd. (YDCI), Yongzhou Economic and Technological Development Zone, Changfeng Industry Park, No. 1 Liebao Road, Lengshuitan District, Yongzhou City, Hunan Province, China
| | - Edith Suzarte
- Research Department, Center for Genetic Engineering and Biotechnology (CIGB), 10600, Havana, Cuba
| | - Glay Chinea
- Research Department, Center for Genetic Engineering and Biotechnology (CIGB), 10600, Havana, Cuba
| | - Miaohong Zhang
- Hunan PRIMA Drug Research Center Co., Ltd., National Liuyang Economic and Technological Development Zone, 123 Kangtian Road, Changsha City, Hunan, China
| | - Zhiqiang Zhou
- Hunan PRIMA Drug Research Center Co., Ltd., National Liuyang Economic and Technological Development Zone, 123 Kangtian Road, Changsha City, Hunan, China
| | - Yaqin Lan
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC) Lengshuitan District, Yongzhou City, 425000, Hunan, China
- Yongzhou Development and Construction Investment Co. Ltd. (YDCI), Yongzhou Economic and Technological Development Zone, Changfeng Industry Park, No. 1 Liebao Road, Lengshuitan District, Yongzhou City, Hunan Province, China
| | - Ricardo Silva
- Science and Innovation Directorate, BioCubaFarma, Independence Avenue, No. 8126, Corner 100 Street, 10800, Havana, Cuba
| | - Gerardo Guillén
- Research Department, Center for Genetic Engineering and Biotechnology (CIGB), 10600, Havana, Cuba
| | - Ke Yang
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC) Lengshuitan District, Yongzhou City, 425000, Hunan, China
- Yongzhou Development and Construction Investment Co. Ltd. (YDCI), Yongzhou Economic and Technological Development Zone, Changfeng Industry Park, No. 1 Liebao Road, Lengshuitan District, Yongzhou City, Hunan Province, China
| | - Wen Li
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC) Lengshuitan District, Yongzhou City, 425000, Hunan, China.
- Yongzhou Development and Construction Investment Co. Ltd. (YDCI), Yongzhou Economic and Technological Development Zone, Changfeng Industry Park, No. 1 Liebao Road, Lengshuitan District, Yongzhou City, Hunan Province, China.
| | - Yasser Perera
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC) Lengshuitan District, Yongzhou City, 425000, Hunan, China.
- R&D Department, Yongzhou Zhong Gu Biotechnology Co., Ltd., Yangjiaqiao Street, Lengshuitan District, Yongzhou City, 425000, Hunan, China.
- Research Department, Center for Genetic Engineering and Biotechnology (CIGB), 10600, Havana, Cuba.
| | - Lisset Hermida
- Research Department, China-Cuba Biotechnology Joint Innovation Center (CCBJIC) Lengshuitan District, Yongzhou City, 425000, Hunan, China.
- Yongzhou Development and Construction Investment Co. Ltd. (YDCI), Yongzhou Economic and Technological Development Zone, Changfeng Industry Park, No. 1 Liebao Road, Lengshuitan District, Yongzhou City, Hunan Province, China.
- Science and Innovation Directorate, BioCubaFarma, Independence Avenue, No. 8126, Corner 100 Street, 10800, Havana, Cuba.
| |
Collapse
|
26
|
Zornikova K, Dianov D, Ivanova N, Davydova V, Nenasheva T, Fefelova E, Bogolyubova A. Features of Highly Homologous T-Cell Receptor Repertoire in the Immune Response to Mutations in Immunogenic Epitopes. Int J Mol Sci 2024; 25:12591. [PMID: 39684303 DOI: 10.3390/ijms252312591] [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: 10/18/2024] [Revised: 11/21/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
CD8+ T-cell immunity, mediated through interactions between human leukocyte antigen (HLA) and the T-cell receptor (TCR), plays a pivotal role in conferring immune memory and protection against viral infections. The emergence of SARS-CoV-2 variants presents a significant challenge to the existing population immunity. While numerous SARS-CoV-2 mutations have been associated with immune evasion from CD8+ T cells, the molecular effects of most mutations on epitope-specific TCR recognition remain largely unexplored, particularly for epitope-specific repertoires characterized by common TCRs. In this study, we investigated an HLA-A*24-restricted NYN epitope (Spike448-456) that elicits broad and highly homologous CD8+ T cell responses in COVID-19 patients. Eleven naturally occurring mutations in the NYN epitope, all of which retained cell surface presentation by HLA, were tested against four transgenic Jurkat reporter cell lines. Our findings demonstrate that, with the exception of L452R and the combined mutation L452Q + Y453F, these mutations have minimal impact on the avidity of recognition by NYN peptide-specific TCRs. Additionally, we observed that a similar TCR responded differently to mutant epitopes and demonstrated cross-reactivity to the unrelated VYF epitope (ORF3a112-120). The results contradict the idea that immune responses with limited receptor diversity are insufficient to provide protection against emerging variants.
Collapse
Affiliation(s)
- Ksenia Zornikova
- National Medical Research Center for Hematology, Moscow 125167, Russia
| | - Dmitry Dianov
- National Medical Research Center for Hematology, Moscow 125167, Russia
| | - Natalia Ivanova
- National Medical Research Center for Hematology, Moscow 125167, Russia
| | - Vassa Davydova
- National Medical Research Center for Hematology, Moscow 125167, Russia
| | - Tatiana Nenasheva
- National Medical Research Center for Hematology, Moscow 125167, Russia
| | | | | |
Collapse
|
27
|
Sharma S, Roy D, Cherian S. In-silico evaluation of the T-cell based immune response against SARS-CoV-2 omicron variants. Sci Rep 2024; 14:25413. [PMID: 39455652 PMCID: PMC11511884 DOI: 10.1038/s41598-024-75658-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
During of COVID-19 pandemic, Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has continuously evolved, resulting in the emergence of several new variants of concerns (VOCs) with numerous mutations. These VOCs dominate in various regions due to increased transmissibility and antibody evasion, potentially reducing vaccine effectiveness. Nonetheless, it remains uncertain whether the recent SARS-CoV-2 VOCs have the ability to circumvent the T cell immunity elicited by either COVID-19 vaccination or natural infection. To address this, we conducted in-silico analysis to examine the impact of VOC-specific mutations at the epitope level and T cell cross-reactivity with the ancestral SARS-CoV-2. According to the in-silico investigation, T cell responses triggered by immunization or prior infections still recognize the variants in spite of mutations. These variants are expected to either maintain their dominant epitope HLA patterns or bind with new HLAs, unlike the epitopes of the ancestral strain. Our findings indicate that a significant proportion of immuno-dominant CD8 + and CD4 + epitopes are conserved across all the variants, implying that existing vaccines might maintain efficacy against new variations. However, further in-vitro and in-vivo studies are needed to validate the in-silico results and fully elucidate immune sensitivities to VOCs.
Collapse
Affiliation(s)
- Shivangi Sharma
- Bioinformatics and Data Management Group, ICMR-National Institute of Virology, Pune, Maharashtra, 411001, India
| | - Diya Roy
- Bioinformatics and Data Management Group, ICMR-National Institute of Virology, Pune, Maharashtra, 411001, India
| | - Sarah Cherian
- Bioinformatics and Data Management Group, ICMR-National Institute of Virology, Pune, Maharashtra, 411001, India.
| |
Collapse
|
28
|
Bozkus CC, Brown M, Velazquez L, Thomas M, Wilson EA, O’Donnell T, Ruchnewitz D, Geertz D, Bykov Y, Kodysh J, Oguntuyo KY, Roudko V, Hoyos D, Srivastava KD, Kleiner G, Alshammary H, Karekar N, McClain C, Gopal R, Nie K, Del Valle D, Delbeau-Zagelbaum D, Rodriguez D, Setal J, The Mount Sinai COVID-19 Biobank Team, Carroll E, Wiesendanger M, Gulko PS, Charney A, Merad M, Kim-Schulze S, Lee B, Wajnberg A, Simon V, Greenbaum BD, Chowell D, Vabret N, Luksza M, Bhardwaj N. T cell epitope mapping reveals immunodominance of evolutionarily conserved regions within SARS-CoV-2 proteome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619918. [PMID: 39484455 PMCID: PMC11527131 DOI: 10.1101/2024.10.23.619918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
As SARS-CoV-2 variants continue to emerge capable of evading neutralizing antibodies, it has become increasingly important to fully understand the breadth and functional profile of T cell responses to determine their impact on the immune surveillance of variant strains. Here, sampling healthy individuals, we profiled the kinetics and polyfunctionality of T cell immunity elicited by mRNA vaccination. Modeling of anti-spike T cell responses against ancestral and variant strains of SARS-CoV-2 suggested that epitope immunodominance and cross-reactivity are major predictive determinants of T cell immunity. To identify immunodominant epitopes across the viral proteome, we generated a comprehensive map of CD4+ and CD8+ T cell epitopes within non-spike proteins that induced polyfunctional T cell responses in convalescent patients. We found that immunodominant epitopes mainly resided within regions that were minimally disrupted by mutations in emerging variants. Conservation analysis across historical human coronaviruses combined with in silico alanine scanning mutagenesis of non-spike proteins underscored the functional importance of mutationally-constrained immunodominant regions. Collectively, these findings identify immunodominant T cell epitopes across the mutationally-constrained SARS-CoV-2 proteome, potentially providing immune surveillance against emerging variants, and inform the design of next-generation vaccines targeting antigens throughout SARS-CoV-2 proteome for broader and more durable protection.
Collapse
Affiliation(s)
- Cansu Cimen Bozkus
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Parker Institute of Cancer Immunotherapy, San Francisco, CA, USA
| | - Matthew Brown
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Parker Institute of Cancer Immunotherapy, San Francisco, CA, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leandra Velazquez
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marcus Thomas
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric A. Wilson
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Timothy O’Donnell
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Denis Ruchnewitz
- Institute for Biological Physics, University of Cologne, 50937 Cologne, Germany
| | - Douglas Geertz
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yonina Bykov
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julia Kodysh
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kasopefoluwa Y. Oguntuyo
- The Department of Medicine, The Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vladimir Roudko
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Hoyos
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Komal D. Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulio Kleiner
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hala Alshammary
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Neha Karekar
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher McClain
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ramya Gopal
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kai Nie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diane Del Valle
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Denise Rodriguez
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Setal
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Emily Carroll
- The Department of Medicine, The Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Margrit Wiesendanger
- The Department of Medicine, The Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Percio S. Gulko
- The Department of Medicine, The Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Charney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ania Wajnberg
- The Department of Medicine, The Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin D Greenbaum
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Physiology, Biophysics & Systems Biology, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Diego Chowell
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicolas Vabret
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marta Luksza
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Parker Institute of Cancer Immunotherapy, San Francisco, CA, USA
| |
Collapse
|
29
|
Bean DJ, Liang YM, Sagar M. Recent Endemic Coronavirus Infection Associates With Higher SARS-CoV-2 Cross-Reactive Fc Receptor Binding Antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619886. [PMID: 39484477 PMCID: PMC11527020 DOI: 10.1101/2024.10.23.619886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Recent documented infection with an endemic coronavirus (eCoV) associates with less severe coronavirus disease 2019 (COVID-19), yet the immune mechanism behind this protection has not been fully explored. We measured both antibody and T cell responses against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in SARS-CoV-2 naïve individuals classified into two groups, either with or without presumed recent eCoV infections. There was no difference in neutralizing antibodies and T cell responses against SARS-CoV-2 antigens between the two groups. SARS-CoV-2 naïve individuals with recent presumed eCoV infection, however, had higher levels of Fc receptor (FcR) binding antibodies against eCoV spikes (S) and SARS-CoV-2 S2. There was also a significant correlation between eCoV and SARS-CoV-2 FcR binding antibodies. Recent eCoV infection boosts cross-reactive antibodies that can mediate Fc effector functions, and this may play a role in the observed heterotypic immune protection against severe COVID-19.
Collapse
Affiliation(s)
- David J. Bean
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine; Boston, MA
| | - Yan Mei Liang
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine; Boston, MA
| | - Manish Sagar
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine; Boston, MA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine; Boston, MA
| |
Collapse
|
30
|
Deng S, Xu Z, Hu J, Yang Y, Zhu F, Liu Z, Zhang H, Wu S, Jin T. The molecular mechanisms of CD8 + T cell responses to SARS-CoV-2 infection mediated by TCR-pMHC interactions. Front Immunol 2024; 15:1468456. [PMID: 39450171 PMCID: PMC11499136 DOI: 10.3389/fimmu.2024.1468456] [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: 07/30/2024] [Accepted: 09/16/2024] [Indexed: 10/26/2024] Open
Abstract
Cytotoxic CD8+ T lymphocytes (CTLs) have been implicated in the severity of COVID-19. The TCR-pMHC ternary complex, formed by the T cell receptor (TCR) and peptide-MHC (major histocompatibility complex), constitutes the molecular basis of CTL responses against SARS-CoV-2. While numerous studies have been conducted on T cell immunity, the molecular mechanisms underlying CTL-mediated immunity against SARS-CoV-2 infection have not been well elaborated. In this review, we described the association between HLA variants and different immune responses to SARS-CoV-2 infection, which may lead to varying COVID-19 outcomes. We also summarized the specific TCR repertoires triggered by certain SARS-CoV-2 CTL epitopes, which might explain the variations in disease outcomes among different patients. Importantly, we have highlighted the primary strategies used by SARS-CoV-2 variants to evade T-cell killing: disrupting peptide-MHC binding, TCR recognition, and antigen processing. This review provides valuable insights into the molecule mechanism of CTL responses during SARS-CoV-2 infection, aiding efforts to control the pandemic and prepare for future challenges.
Collapse
Affiliation(s)
- Shasha Deng
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Zhihao Xu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jing Hu
- Laboratory of Structural Immunology, the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yunru Yang
- Laboratory of Structural Immunology, the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Fang Zhu
- Laboratory of Structural Immunology, the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Zhuan Liu
- Laboratory of Structural Immunology, the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hongliang Zhang
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Songquan Wu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
| | - Tengchuan Jin
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, China
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
- Laboratory of Structural Immunology, the Chinese Academy of Sciences (CAS) Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science & Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, China
| |
Collapse
|
31
|
Vránová L, Poláková I, Vaníková Š, Saláková M, Musil J, Vaníčková M, Vencálek O, Holub M, Bohoněk M, Řezáč D, Dresler J, Tachezy R, Šmahel M. Multiparametric analysis of the specific immune response against SARS-CoV-2. Infect Dis (Lond) 2024; 56:851-869. [PMID: 38805304 DOI: 10.1080/23744235.2024.2358379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 04/24/2024] [Accepted: 05/17/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND SARS-CoV-2, which causes COVID-19, has killed more than 7 million people worldwide. Understanding the development of postinfectious and postvaccination immune responses is necessary for effective treatment and the introduction of appropriate antipandemic measures. OBJECTIVES We analysed humoral and cell-mediated anti-SARS-CoV-2 immune responses to spike (S), nucleocapsid (N), membrane (M), and open reading frame (O) proteins in individuals collected up to 1.5 years after COVID-19 onset and evaluated immune memory. METHODS Peripheral blood mononuclear cells and serum were collected from patients after COVID-19. Sampling was performed in two rounds: 3-6 months after infection and after another year. Most of the patients were vaccinated between samplings. SARS-CoV-2-seronegative donors served as controls. ELISpot assays were used to detect SARS-CoV-2-specific T and B cells using peptide pools (S, NMO) or recombinant proteins (rS, rN), respectively. A CEF peptide pool consisting of selected viral epitopes was applied to assess the antiviral T-cell response. SARS-CoV-2-specific antibodies were detected via ELISA and a surrogate virus neutralisation assay. RESULTS We confirmed that SARS-CoV-2 infection induces the establishment of long-term memory IgG+ B cells and memory T cells. We also found that vaccination enhanced the levels of anti-S memory B and T cells. Multivariate comparison also revealed the benefit of repeated vaccination. Interestingly, the T-cell response to CEF was lower in patients than in controls. CONCLUSION This study supports the importance of repeated vaccination for enhancing immunity and suggests a possible long-term perturbation of the overall antiviral immune response caused by SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Lucie Vránová
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Ingrid Poláková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Šárka Vaníková
- Department of Immunomonitoring and Flow Cytometry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Martina Saláková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Jan Musil
- Department of Immunomonitoring and Flow Cytometry, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Marie Vaníčková
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Ondřej Vencálek
- Department of Mathematical Analysis and Applications of Mathematics, Faculty of Science, Palacky University in Olomouc, Olomouc, Czech Republic
| | - Michal Holub
- Department of Infectious Diseases, First Faculty of Medicine, Military University Hospital Prague and Charles University, Prague, Czech Republic
| | - Miloš Bohoněk
- Department of Hematology and Blood Transfusion, Military University Hospital Prague, Prague, Czech Republic
- Faculty of Biomedical Engineering, Czech Technical University, Prague, Czech Republic
| | - David Řezáč
- Department of Infectious Diseases, First Faculty of Medicine, Military University Hospital Prague and Charles University, Prague, Czech Republic
| | - Jiří Dresler
- Military Health Institute, Military Medical Agency, Prague, Czech Republic
| | - Ruth Tachezy
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| | - Michal Šmahel
- Department of Genetics and Microbiology, Faculty of Science, Charles University, BIOCEV, Vestec, Czech Republic
| |
Collapse
|
32
|
Notarbartolo S. T-Cell Immune Responses to SARS-CoV-2 Infection and Vaccination. Vaccines (Basel) 2024; 12:1126. [PMID: 39460293 PMCID: PMC11511197 DOI: 10.3390/vaccines12101126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/25/2024] [Accepted: 09/27/2024] [Indexed: 10/28/2024] Open
Abstract
The innate and adaptive immune systems collaborate to detect SARS-CoV-2 infection, minimize the viral spread, and kill infected cells, ultimately leading to the resolution of the infection. The adaptive immune system develops a memory of previous encounters with the virus, providing enhanced responses when rechallenged by the same pathogen. Such immunological memory is the basis of vaccine function. Here, we review the current knowledge on the immune response to SARS-CoV-2 infection and vaccination, focusing on the pivotal role of T cells in establishing protective immunity against the virus. After providing an overview of the immune response to SARS-CoV-2 infection, we describe the main features of SARS-CoV-2-specific CD4+ and CD8+ T cells, including cross-reactive T cells, generated in patients with different degrees of COVID-19 severity, and of Spike-specific CD4+ and CD8+ T cells induced by vaccines. Finally, we discuss T-cell responses to SARS-CoV-2 variants and hybrid immunity and conclude by highlighting possible strategies to improve the efficacy of COVID-19 vaccination.
Collapse
Affiliation(s)
- Samuele Notarbartolo
- Infectious Diseases Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|
33
|
Macchia I, La Sorsa V, Ciervo A, Ruspantini I, Negri D, Borghi M, De Angelis ML, Luciani F, Martina A, Taglieri S, Durastanti V, Altavista MC, Urbani F, Mancini F. T Cell Peptide Prediction, Immune Response, and Host-Pathogen Relationship in Vaccinated and Recovered from Mild COVID-19 Subjects. Biomolecules 2024; 14:1217. [PMID: 39456150 PMCID: PMC11505848 DOI: 10.3390/biom14101217] [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: 07/29/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/28/2024] Open
Abstract
COVID-19 remains a significant threat, particularly to vulnerable populations. The emergence of new variants necessitates the development of treatments and vaccines that induce both humoral and cellular immunity. This study aimed to identify potentially immunogenic SARS-CoV-2 peptides and to explore the intricate host-pathogen interactions involving peripheral immune responses, memory profiles, and various demographic, clinical, and lifestyle factors. Using in silico and experimental methods, we identified several CD8-restricted SARS-CoV-2 peptides that are either poorly studied or have previously unreported immunogenicity: fifteen from the Spike and three each from non-structural proteins Nsp1-2-3-16. A Spike peptide, LA-9, demonstrated a 57% response rate in ELISpot assays using PBMCs from 14 HLA-A*02:01 positive, vaccinated, and mild-COVID-19 recovered subjects, indicating its potential for diagnostics, research, and multi-epitope vaccine platforms. We also found that younger individuals, with fewer vaccine doses and longer intervals since infection, showed lower anti-Spike (ELISA) and anti-Wuhan neutralizing antibodies (pseudovirus assay), higher naïve T cells, and lower central memory, effector memory, and CD4hiCD8low T cells (flow cytometry) compared to older subjects. In our cohort, a higher prevalence of Vδ2-γδ and DN T cells, and fewer naïve CD8 T cells, seemed to correlate with strong cellular and lower anti-NP antibody responses and to associate with Omicron infection, absence of confusional state, and habitual sporting activity.
Collapse
Affiliation(s)
- Iole Macchia
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina La Sorsa
- Research Promotion and Coordination Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Alessandra Ciervo
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Irene Ruspantini
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Donatella Negri
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Martina Borghi
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| | - Maria Laura De Angelis
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Francesca Luciani
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Antonio Martina
- National Center for the Control and Evaluation of Medicines, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.L.); (A.M.)
| | - Silvia Taglieri
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Valentina Durastanti
- Neurology Unit, San Filippo Neri Hospital, ASL RM1, 00135 Rome, Italy; (V.D.); (M.C.A.)
| | | | - Francesca Urbani
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, 00161 Rome, Italy; (I.M.); (M.L.D.A.); (S.T.)
| | - Fabiola Mancini
- Department of Infectious Diseases, Istituto Superiore di Sanità, 00161 Rome, Italy; (A.C.); (D.N.); (M.B.); (F.M.)
| |
Collapse
|
34
|
Lee YR, Liou CW, Liu IH, Chang JM. A nonadjuvanted HLA-restricted peptide vaccine induced both T and B cell immunity against SARS-CoV-2 spike protein. Sci Rep 2024; 14:20579. [PMID: 39242614 PMCID: PMC11379847 DOI: 10.1038/s41598-024-71663-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024] Open
Abstract
During COVID-19 pandemic, cases of postvaccination infections and restored SARS-CoV-2 virus have increased after full vaccination, which might be contributed to by immune surveillance escape or virus rebound. Here, artificial linear 9-mer human leucocyte antigen (HLA)-restricted UC peptides were designed based on the well-conserved S2 region of the SARS-CoV-2 spike protein regardless of rapid mutation and glycosylation hindrance. The UC peptides were characterized for its effect on immune molecules and cells by HLA-tetramer refolding assay for HLA-binding ability, by HLA-tetramer specific T cell assay for engaged cytotoxic T lymphocytes (CTLs) involvement, by HLA-dextramer T cell assay for B cell activation, by intracellular cytokine release assay for polarization of immune response, Th1 or Th2. The specific lysis activity assay of T cells was performed for direct activation of cytotoxic T lymphocytes by UC peptides. Mice were immunized for immunogenicity of UC peptides in vivo and immunized sera was assay for complement cytotoxicity assay. Results appeared that through the engagement of UC peptides and immune molecules, HLA-I and II, that CTLs elicited cytotoxic activity by recognizing SARS-CoV-2 spike-bearing cells and preferably secreting Th1 cytokines. The UC peptides also showed immunogenicity and generated a specific antibody in mice by both intramuscular injection and oral delivery without adjuvant formulation. In conclusion, a T-cell vaccine could provide long-lasting protection against SARS-CoV-2 either during reinfection or during SARS-CoV-2 rebound. Due to its ability to eradicate SARS-CoV-2 virus-infected cells, a COVID-19 T-cell vaccine might provide a solution to lower COVID-19 severity and long COVID-19.
Collapse
Affiliation(s)
- Yi-Ru Lee
- Vacino Biotech Co., Ltd., 4F, No. 99, Lane 130, Sec 1, Academia Rd., Nangang District, Taipei, 11571, Taiwan, ROC
| | - Chiung-Wen Liou
- Vacino Biotech Co., Ltd., 4F, No. 99, Lane 130, Sec 1, Academia Rd., Nangang District, Taipei, 11571, Taiwan, ROC
| | - I-Hua Liu
- Vacino Biotech Co., Ltd., 4F, No. 99, Lane 130, Sec 1, Academia Rd., Nangang District, Taipei, 11571, Taiwan, ROC
| | - Jia-Ming Chang
- Vacino Biotech Co., Ltd., 4F, No. 99, Lane 130, Sec 1, Academia Rd., Nangang District, Taipei, 11571, Taiwan, ROC.
| |
Collapse
|
35
|
Gangaev A, van Sleen Y, Brandhorst N, Hoefakker K, Prajapati B, Singh A, Boerma A, van der Heiden M, Oosting SF, van der Veldt AAM, Hiltermann TJN, GeurtsvanKessel CH, Dingemans AMC, Smit EF, de Vries EGE, Haanen JBAG, Kvistborg P, van Baarle D. mRNA-1273 vaccination induces polyfunctional memory CD4 and CD8 T cell responses in patients with solid cancers undergoing immunotherapy or/and chemotherapy. Front Immunol 2024; 15:1447555. [PMID: 39257577 PMCID: PMC11385311 DOI: 10.3389/fimmu.2024.1447555] [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: 06/11/2024] [Accepted: 08/08/2024] [Indexed: 09/12/2024] Open
Abstract
Introduction Research has confirmed the safety and comparable seroconversion rates following SARS-CoV-2 vaccination in patients with solid cancers. However, the impact of cancer treatment on vaccine-induced T cell responses remains poorly understood. Methods In this study, we expand on previous findings within the VOICE trial by evaluating the functional and phenotypic composition of mRNA-1273-induced T cell responses in patients with solid tumors undergoing immunotherapy, chemotherapy, or both, compared to individuals without cancer. We conducted an ELISpot analysis on 386 participants to assess spike-specific T cell responses 28 days after full vaccination. Further in-depth characterization of using flow cytometry was performed on a subset of 63 participants to analyze the functional phenotype and differentiation state of spike-specific T cell responses. Results ELISpot analysis showed robust induction of spike-specific T cell responses across all treatment groups, with response rates ranging from 75% to 80%. Flow cytometry analysis revealed a distinctive cytokine production pattern across cohorts, with CD4 T cells producing IFNγ, TNF, and IL-2, and CD8 T cells producing IFNγ, TNF, and CCL4. Variations were observed in the proportion of monofunctional CD4 T cells producing TNF, particularly higher in individuals without cancer and patients treated with chemotherapy alone, while those treated with immunotherapy or chemoimmunotherapy predominantly produced IFNγ. Despite these differences, polyfunctional spike-specific memory CD4 and CD8 T cell responses were comparable across cohorts. Notably, immunotherapy-treated patients exhibited an expansion of spike-specific CD4 T cells with a terminally differentiated effector memory phenotype. Discussion These findings demonstrate that systemic treatment in patients with solid tumors does not compromise the quality of polyfunctional mRNA-1273-induced T cell responses. This underscores the importance of COVID-19 vaccination in patients with solid cancers undergoing systemic treatment.
Collapse
Affiliation(s)
- Anastasia Gangaev
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Yannick van Sleen
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
| | - Nicole Brandhorst
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Kelly Hoefakker
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Bimal Prajapati
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
| | - Amrita Singh
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
| | - Annemarie Boerma
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
| | - Marieke van der Heiden
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
| | - Sjoukje F. Oosting
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Astrid A. M. van der Veldt
- Department of Medical Oncology and Radiology & Nuclear Medicine, Erasmus Medical Center (MC)-Cancer Institute, Rotterdam, Netherlands
| | - T. Jeroen N. Hiltermann
- Department of Pulmonary Diseases, University Medical Centre Groningen, Groningen, Netherlands
| | - Corine H. GeurtsvanKessel
- Department of Viroscience, Erasmus Medical Center (MC) Cancer Institute, University Medical Centre, Rotterdam, Netherlands
| | | | - Egbert F. Smit
- Department of Thoracic Oncology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Elisabeth G. E. de Vries
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - John B. A. G. Haanen
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Pia Kvistborg
- Division of Molecular Oncology and Immunology, The Netherlands Cancer Institute, Amsterdam, Netherlands
| | - Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University Medical Centre Groningen, Groningen, Netherlands
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Utrecht, Netherlands
| |
Collapse
|
36
|
Benhamouda N, Besbes A, Bauer R, Mabrouk N, Gadouas G, Desaint C, Chevrier L, Lefebvre M, Radenne A, Roelens M, Parfait B, Weiskopf D, Sette A, Gruel N, Courbebaisse M, Appay V, Paul S, Gorochov G, Ropers J, Lebbah S, Lelievre JD, Johannes L, Ulmer J, Lebeaux D, Friedlander G, De Lamballerie X, Ravel P, Kieny MP, Batteux F, Durier C, Launay O, Tartour E. Cytokine profile of anti-spike CD4 +T cells predicts humoral and CD8 +T cell responses after anti-SARS-CoV-2 mRNA vaccination. iScience 2024; 27:110441. [PMID: 39104410 PMCID: PMC11298648 DOI: 10.1016/j.isci.2024.110441] [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: 04/08/2024] [Revised: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Coordinating immune responses - humoral and cellular - is vital for protection against severe Covid-19. Our study evaluates a multicytokine CD4+T cell signature's predictive for post-vaccinal serological and CD8+T cell responses. A cytokine signature composed of four cytokines (IL-2, TNF-α, IP10, IL-9) excluding IFN-γ, and generated through machine learning, effectively predicted the CD8+T cell response following mRNA-1273 or BNT162b2 vaccine administration. Its applicability extends to murine vaccination models, encompassing diverse immunization routes (such as intranasal) and vaccine platforms (including adjuvanted proteins). Notably, we found correlation between CD4+T lymphocyte-produced IL-21 and the humoral response. Consequently, we propose a test that offers a rapid overview of integrated immune responses. This approach holds particular relevance for scenarios involving immunocompromised patients because they often have low cell counts (lymphopenia) or pandemics. This study also underscores the pivotal role of CD4+T cells during a vaccine response and highlights their value in vaccine immunomonitoring.
Collapse
Affiliation(s)
- Nadine Benhamouda
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Anissa Besbes
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | | | - Nesrine Mabrouk
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Gauthier Gadouas
- Bioinformatics and Cancer System Biology Team, IRCM-INSERM U1194, Institut de Recherche en Cancerologie de Montpellier, Montpellier, France
| | - Corinne Desaint
- INSERM SC10-US019, Villejuif, France
- Université Paris Cité, INSERM, CIC 1417, F-CRIN, Innovative Clinical Research Network in Vaccinology (I-REIVAC), APHP, CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Lucie Chevrier
- Université Paris Cité, INSERM U1016 Insitut Cochin, Hôpital Cochin, APHP, Centre Service d’immunologie Biologique, Paris, France
| | - Maeva Lefebvre
- Service de maladies infectieuses et tropicales, Centre de prévention des maladies infectieuses et transmissibles CHU de Nantes, Nantes, France
| | - Anne Radenne
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière-Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, APHP, Paris, France
| | - Marie Roelens
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Béatrice Parfait
- Centre de ressources Biologiques, Hôpital Cochin, APHP, Paris, France
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, School of Medicine in Health Sciences, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Nadège Gruel
- INSERM U830, Équipe Labellisée Ligue Nationale Contre le Cancer, Diversity and Plasticity of Childhood Tumors Lab, Centre de Recherche, Institut Curie, Université PSL, Paris, France
- Department of Translational Research, Centre de Recherche, Institut Curie, Université PSL, Paris, France
| | - Marie Courbebaisse
- Faculté de Médecine, Université Paris Cité, Paris, France
- Explorations fonctionnelles rénales, Physiologie, Hôpital Européen Georges-Pompidou, APHP, Paris, France
| | - Victor Appay
- Université de Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
- International Research Center of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Stephane Paul
- Centre International de Recherche en Infectiologie, Team GIMAP, Université Jean Monnet, Université Claude Bernard Lyon, INSERM, CIC 1408 INSERM Vaccinology, Immunology Department, iBiothera Reference Center, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Guy Gorochov
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses, APHP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jacques Ropers
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière –Hôpitaux Universitaires Pitié Salpêtrière- Charles Foix, APHP, Paris, France
| | - Said Lebbah
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière –Hôpitaux Universitaires Pitié Salpêtrière- Charles Foix, APHP, Paris, France
| | - Jean-Daniel Lelievre
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
- Groupe Henri-Mondor Albert-Chenevier, APHP, Créteil, France
| | - Ludger Johannes
- Cellular and Chemical Biology Unit, U1143 INSERM, UMR3666 CNRS, Institut Curie, Centre de Recherche, Université PSL, Paris, France
| | - Jonathan Ulmer
- Cellular and Chemical Biology Unit, U1143 INSERM, UMR3666 CNRS, Institut Curie, Centre de Recherche, Université PSL, Paris, France
| | - David Lebeaux
- Université Paris Cité, Service de maladies infectieuses Hôpital Saint Louis/Lariboisère APHP, INSERM, Paris, France
| | - Gerard Friedlander
- Department of « Croissance et Signalisation », Institut Necker Enfants Malades, INSERM U1151, CNRS UMR 8253, Université de Paris Cité, Paris, France
| | - Xavier De Lamballerie
- Unité des Virus Émergents, UVE: Aix-Marseille Université, IRD 190, INSERM 1207 Marseille, France
| | - Patrice Ravel
- Bioinformatics and Cancer System Biology Team, IRCM-INSERM U1194, Institut de Recherche en Cancerologie de Montpellier, Montpellier, France
| | - Marie Paule Kieny
- Institut National de la Santé et de la Recherche Médicale, INSERM, Paris, France
| | - Fréderic Batteux
- Université Paris Cité, INSERM U1016 Insitut Cochin, Hôpital Cochin, APHP, Centre Service d’immunologie Biologique, Paris, France
| | | | - Odile Launay
- Université Paris Cité, INSERM, CIC 1417, F-CRIN, Innovative Clinical Research Network in Vaccinology (I-REIVAC), APHP, CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Eric Tartour
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| |
Collapse
|
37
|
Rastogi A, Gautam S, Kumar M. Bioinformatic elucidation of conserved epitopes to design a potential vaccine candidate against existing and emerging SARS-CoV-2 variants of concern. Heliyon 2024; 10:e35129. [PMID: 39157328 PMCID: PMC11328099 DOI: 10.1016/j.heliyon.2024.e35129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Abstract
The COVID-19 pandemic caused by SARS-CoV-2 poses a significant adverse effects on health and economy globally. Due to mutations in genome, COVID-19 vaccine efficacy decreases. We used immuno-informatics to design a Multi epitope vaccine (MEV) candidate for SARS-CoV-2 variants of concern (VOCs). Hence, we predicted binders/epitopes MHC-I, CD8+, MHC-II, CD4+, and CTLs from spike, membrane and envelope proteins of VOCs. In addition, we assessed the conservation of these binders and epitopes across different VOCs. Subsequently, we designed MEV by combining the predicted CTL and CD4+ epitopes from spike protein, peptide linkers, and an adjuvant. Further, we evaluated the binding of MEV candidate against immune receptors namely HLA class I histocompatibility antigen, HLA class II histocompatibility antigen, and TLR4, achieving binding scores of -1265.3, -1330.7, and -1337.9. Molecular dynamics and normal mode analysis revealed stable docking complexes. Moreover, immune simulation suggested MEV candidate elicits both innate and adaptive immune response. We anticipate that this conserved MEV candidate will provide protection from VOCs and emerging strains.
Collapse
Affiliation(s)
- Amber Rastogi
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39A, Chandigarh, 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Sakshi Gautam
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39A, Chandigarh, 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Manoj Kumar
- Virology Unit and Bioinformatics Centre, Institute of Microbial Technology, Council of Scientific and Industrial Research (CSIR), Sector 39A, Chandigarh, 160036, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| |
Collapse
|
38
|
Hendricks GG, Grigoryan L, Navarro MJ, Catanzaro NJ, Hubbard ML, Powers JM, Mattocks M, Treichel C, Walls AC, Lee J, Ellis D, Wang JY(J, Cheng S, Miranda MC, Valdez A, Chao CW, Chan S, Men C, Johnson MR, Hui H, Wu SY, Lujan V, Muramatsu H, Lin PJ, Sung MM, Tam YK, Leaf EM, Pardi N, Baric RS, Pulendran B, Veesler D, Schäfer A, King NP. Computationally designed mRNA-launched protein nanoparticle vaccines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.22.604655. [PMID: 39091730 PMCID: PMC11291046 DOI: 10.1101/2024.07.22.604655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/04/2024]
Abstract
Both protein nanoparticle and mRNA vaccines were clinically de-risked during the COVID-19 pandemic1-6. These vaccine modalities have complementary strengths: antigen display on protein nanoparticles can enhance the magnitude, quality, and durability of antibody responses7-10, while mRNA vaccines can be rapidly manufactured11 and elicit antigen-specific CD4 and CD8 T cells12,13. Here we leverage a computationally designed icosahedral protein nanoparticle that was redesigned for optimal secretion from eukaryotic cells14 to develop an mRNA-launched nanoparticle vaccine for SARS-CoV-2. The nanoparticle, which displays 60 copies of a stabilized variant of the Wuhan-Hu-1 Spike receptor binding domain (RBD)15, formed monodisperse, antigenically intact assemblies upon secretion from transfected cells. An mRNA vaccine encoding the secreted RBD nanoparticle elicited 5- to 28-fold higher levels of neutralizing antibodies than an mRNA vaccine encoding membrane-anchored Spike, induced higher levels of CD8 T cells than the same immunogen when delivered as an adjuvanted protein nanoparticle, and protected mice from vaccine-matched and -mismatched SARS-CoV-2 challenge. Our data establish that delivering protein nanoparticle immunogens via mRNA vaccines can combine the benefits of each modality and, more broadly, highlight the utility of computational protein design in genetic immunization strategies.
Collapse
Affiliation(s)
- Grace G. Hendricks
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Lilit Grigoryan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Mary Jane Navarro
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Nicholas J. Catanzaro
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Miranda L. Hubbard
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - John M. Powers
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Melissa Mattocks
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Catherine Treichel
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Alexandra C. Walls
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, Seattle, WA, USA
| | - Jimin Lee
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Daniel Ellis
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Jing Yang (John) Wang
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Suna Cheng
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Marcos C. Miranda
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Adian Valdez
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Cara W. Chao
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Graduate Program in Molecular and Cellular Biology, University of Washington, Seattle, WA, USA
| | - Sidney Chan
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Christine Men
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Max R. Johnson
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Harold Hui
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Sheng-Yang Wu
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Victor Lujan
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Hiromi Muramatsu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - Elizabeth M. Leaf
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ralph S. Baric
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - David Veesler
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alexandra Schäfer
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Neil P. King
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Lead contact
| |
Collapse
|
39
|
Valiate BVS, Castro JTD, Marçal TG, Andrade LAF, Oliveira LID, Maia GBF, Faustino LP, Hojo-Souza NS, Reis MAAD, Bagno FF, Salazar N, Teixeira SR, Almeida GG, Gazzinelli RT. Evaluation of an RBD-nucleocapsid fusion protein as a booster candidate for COVID-19 vaccine. iScience 2024; 27:110177. [PMID: 38993669 PMCID: PMC11238127 DOI: 10.1016/j.isci.2024.110177] [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: 08/17/2023] [Revised: 04/30/2024] [Accepted: 05/31/2024] [Indexed: 07/13/2024] Open
Abstract
Despite successful vaccines and updates, constant mutations of SARS-CoV-2 makes necessary the search for new vaccines. We generated a chimeric protein that comprises the receptor-binding domain from spike and the nucleocapsid antigens (SpiN) from SARS-CoV-2. Once SpiN elicits a protective immune response in rodents, here we show that convalescent and previously vaccinated individuals respond to SpiN. CD4+ and CD8+ T cells from these individuals produced greater amounts of IFN-γ when stimulated with SpiN, compared to SARS-CoV-2 antigens. Also, B cells from these individuals were able to secrete antibodies that recognize SpiN. When administered as a boost dose in mice previously immunized with CoronaVac, ChAdOx1-S or BNT162b2, SpiN was able to induce a greater or equivalent immune response to homologous prime/boost. Our data reveal the ability of SpiN to induce cellular and humoral responses in vaccinated human donors, rendering it a promising candidate.
Collapse
Affiliation(s)
- Bruno Vinicius Santos Valiate
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Julia Teixeira de Castro
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | | | - Luis Adan Flores Andrade
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Livia Isabela de Oliveira
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte 31.630-901, MG, Brazil
| | | | | | - Natalia S Hojo-Souza
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | | | - Flávia Fonseca Bagno
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Natalia Salazar
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Santuza R Teixeira
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Gregório Guilherme Almeida
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| | - Ricardo Tostes Gazzinelli
- Fundação Oswaldo Cruz-Minas, Belo Horizonte 30.190-002, MG, Brazil
- Centro de Tecnologia de Vacinas, Universidade Federal de Minas Gerais, Parque Tecnológico de Belo Horizonte, Belo Horizonte 31.310-260, MG, Brazil
| |
Collapse
|
40
|
Carretero D, Giménez E, Albert E, Colomer E, Montomoli M, Hernani R, Piñana JL, Górriz JL, Solano C, Navarro D. Performance of the Roche Elecsys® IGRA SARS-CoV-2 test for the detection and quantification of virus-reactive T cells in COVID-19-vaccinated immunosuppressed patients and healthy subjects. Eur J Clin Microbiol Infect Dis 2024; 43:1427-1436. [PMID: 38777919 PMCID: PMC11271317 DOI: 10.1007/s10096-024-04852-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 05/16/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE Comparing the performance of commercially available SARS-CoV-2 T-cell immunoassay responses may provide useful information for future observational or intervention studies as well as to their potential customers. METHOD Whole blood was collected from a total of 183 subjects fully vaccinated against COVID-19: 55 healthy controls (Group 1), 50 hematological patients (Group 2), 50 chronic kidney disease patients (Group 3), and 28 elderly nursing home residents (Group 4). Samples were tested with the Roche Elecsys® IGRA (Interferon-gamma release assay) SARS-CoV-2 test (Roche Diagnostics, Rotkreuz, Switzerland), the Euroimmun SARS-CoV-2 test (Euroimmun, Lubeck, Germany), the SARS-CoV-2 T Cell Analysis Kit (Miltenyi Biotec, Bergisch Gladbach, Germany), and a flow-cytometry for intracellular cytokine (IFN-γ) staining-based immunoassay (FC-ICS). RESULTS Overall, the Roche Elecsys® assay returned the highest number of positive results (151/179; 84.3%), followed by the Euroimmun test (127/183; 69%), and the FC-ICS (135/179; 75%). The Kappa coefficient of agreement was best between IGRAs (0.64). Most discordant results across assays involved patients from Group 2. Overall, IFN-γ concentrations measured by both IGRAs correlated strongly (rho = 0.78; 95% CI 0.71-0.84; P < 0.001) irrespective of the study group. The frequencies of SARS-CoV-2-reactive IFN-γ T cells and IFN-γ concentrations measured by the IGRAs correlated moderately for CD4+ T cells, however, weakly for CD8+ T cells. SARS-CoV-2-experienced participants displayed stronger responses than SARS-CoV-2-naïve when IGRAs, rather than FC-ICS, were used. CONCLUSION The SARS-CoV-2 immunoassays evaluated in the present study did not return interchangeable qualitative or quantitative results either in seemingly healthy individuals or in immunosuppressed patients.
Collapse
Affiliation(s)
- Diego Carretero
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Estela Giménez
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Eliseo Albert
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Ester Colomer
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Marco Montomoli
- Nephrology Service, Hospital Clínico Universitario de Valencia, INCLIVA Health Research Institute, Valencia, Spain
| | - Rafael Hernani
- Hematology Service, Hospital Clínico Universitario, INCLIVA Health Research Institute, Valencia, Spain
| | - José Luis Piñana
- Hematology Service, Hospital Clínico Universitario, INCLIVA Health Research Institute, Valencia, Spain
| | - José Luis Górriz
- Nephrology Service, Hospital Clínico Universitario de Valencia, INCLIVA Health Research Institute, Valencia, Spain
- Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain
| | - Carlos Solano
- Hematology Service, Hospital Clínico Universitario, INCLIVA Health Research Institute, Valencia, Spain
- Department of Medicine, School of Medicine, University of Valencia, Valencia, Spain
| | - David Navarro
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain.
- Department of Microbiology, School of Medicine, University of Valencia, Valencia, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain.
- Microbiology Service, Hospital Clínico Universitario, Instituto de Investigación INCLIVA, Av. Blasco Ibáñez 17, 46010, Valencia, Spain.
| |
Collapse
|
41
|
Kakugawa T, Mimura Y, Mimura-Kimura Y, Doi K, Ohteru Y, Kakugawa H, Oishi K, Kakugawa M, Hirano T, Matsunaga K. Kinetics of pro- and anti-inflammatory spike-specific cellular immune responses in long-term care facility residents after COVID-19 mRNA primary and booster vaccination: a prospective longitudinal study in Japan. Immun Ageing 2024; 21:41. [PMID: 38909235 PMCID: PMC11193299 DOI: 10.1186/s12979-024-00444-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/05/2024] [Indexed: 06/24/2024]
Abstract
BACKGROUND The magnitude and durability of cell-mediated immunity in older and severely frail individuals following coronavirus disease 2019 (COVID-19) vaccination remain unclear. A controlled immune response could be the key to preventing severe COVID-19; however, it is uncertain whether vaccination induces an anti-inflammatory cellular immune response. To address these issues, a 48-week-long prospective longitudinal study was conducted. A total of 106 infection-naive participants (57 long-term care facility [LTCF] residents [median age; 89.0 years], 28 outpatients [median age; 72.0 years], and 21 healthcare workers [median age; 51.0 years]) provided peripheral blood mononuclear cell (PBMC) samples for the assessment of spike-specific PBMC responses before primary vaccination, 24 weeks after primary vaccination, and three months after booster vaccination. Cellular immune responses to severe acute respiratory syndrome coronavirus 2 spike protein were examined by measuring interferon (IFN)-γ, tumor necrosis factor (TNF), interleukin (IL)-2, IL-4, IL-6, and IL-10 levels secreted from the spike protein peptide-stimulated PBMCs of participants. RESULTS LTCF residents exhibited significantly lower IFN-γ, TNF, IL-2, and IL-6 levels than healthcare workers after the primary vaccination. Booster vaccination increased IL-2 and IL-6 levels in LTCF residents comparable to those in healthcare workers, whereas IFN-γ and TNF levels in LTCF residents remained significantly lower than those in healthcare workers. IL-10 levels were not significantly different from the initial values after primary vaccination but increased significantly after booster vaccination in all subgroups. Multivariate analysis showed that age was negatively associated with IFN-γ, TNF, IL-2, and IL-6 levels but not with IL-10 levels. The levels of pro-inflammatory cytokines, including IFN-γ, TNF, IL-2, and IL-6, were positively correlated with humoral immune responses, whereas IL-10 levels were not. CONCLUSIONS Older and severely frail individuals may exhibit diminished spike-specific PBMC responses following COVID-19 vaccination compared to the general population. A single booster vaccination may not adequately enhance cell-mediated immunity in older and severely frail individuals to a level comparable to that in the general population. Furthermore, booster vaccination may induce not only a pro-inflammatory cellular immune response but also an anti-inflammatory cellular immune response, potentially mitigating detrimental hyperinflammation.
Collapse
Affiliation(s)
- Tomoyuki Kakugawa
- Department of Pulmonology and Gerontology, Graduate School of Medicine, Yamaguchi University, Ube, Japan.
- Medical Corporation WADOKAI, Hofu Rehabilitation Hospital, Hofu, Japan.
- Department of Respiratory Medicine, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan.
| | - Yusuke Mimura
- The Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan
| | - Yuka Mimura-Kimura
- The Department of Clinical Research, National Hospital Organization Yamaguchi Ube Medical Center, Ube, Japan
| | - Keiko Doi
- Department of Pulmonology and Gerontology, Graduate School of Medicine, Yamaguchi University, Ube, Japan
- Department of Respiratory Medicine and Infectious Disease, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Yuichi Ohteru
- Medical Corporation WADOKAI, Hofu Rehabilitation Hospital, Hofu, Japan
- Department of Respiratory Medicine and Infectious Disease, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Hiroyuki Kakugawa
- Medical Corporation WADOKAI, Hofu Rehabilitation Hospital, Hofu, Japan
| | - Keiji Oishi
- Department of Respiratory Medicine and Infectious Disease, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Masahiro Kakugawa
- Medical Corporation WADOKAI, Hofu Rehabilitation Hospital, Hofu, Japan
| | - Tsunahiko Hirano
- Department of Respiratory Medicine and Infectious Disease, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| | - Kazuto Matsunaga
- Department of Respiratory Medicine and Infectious Disease, Graduate School of Medicine, Yamaguchi University, Ube, Japan
| |
Collapse
|
42
|
Fan X, Song JW, Cao WJ, Zhou MJ, Yang T, Wang J, Meng FP, Shi M, Zhang C, Wang FS. T-Cell Epitope Mapping of SARS-CoV-2 Reveals Coordinated IFN-γ Production and Clonal Expansion of T Cells Facilitates Recovery from COVID-19. Viruses 2024; 16:1006. [PMID: 39066169 PMCID: PMC11281491 DOI: 10.3390/v16071006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/01/2024] [Accepted: 06/14/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND T-cell responses can be protective or detrimental during severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection; however, the underlying mechanism is poorly understood. METHODS In this study, we screened 144 15-mer peptides spanning the SARS-CoV-2 spike, nucleocapsid (NP), M, ORF8, ORF10, and ORF3a proteins and 39 reported SARS-CoV-1 peptides in peripheral blood mononuclear cells (PBMCs) from nine laboratory-confirmed coronavirus disease 2019 (COVID-19) patients (five moderate and four severe cases) and nine healthy donors (HDs) collected before the COVID-19 pandemic. T-cell responses were monitored by IFN-γ and IL-17A production using ELISA, and the positive samples were sequenced for the T cell receptor (TCR) β chain. The positive T-cell responses to individual SARS-CoV-2 peptides were validated by flow cytometry. RESULTS COVID-19 patients with moderate disease produced more IFN-γ than HDs and patients with severe disease (moderate vs. HDs, p < 0.0001; moderate vs. severe, p < 0.0001) but less IL-17A than those with severe disease (p < 0.0001). A positive correlation was observed between IFN-γ production and T-cell clonal expansion in patients with moderate COVID-19 (r = 0.3370, p = 0.0214) but not in those with severe COVID-19 (r = -0.1700, p = 0.2480). Using flow cytometry, we identified that a conserved peptide of the M protein (Peptide-120, P120) was a dominant epitope recognized by CD8+ T cells in patients with moderate disease. CONCLUSION Coordinated IFN-γ production and clonal expansion of SARS-CoV-2-specific T cells are associated with disease resolution in COVID-19. Our findings contribute to a better understanding of T-cell-mediated immunity in COVID-19 and may inform future strategies for managing and preventing severe outcomes of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Xing Fan
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Jin-Wen Song
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
- Medical School of Chinese PLA, Beijing 100853, China
| | - Wen-Jing Cao
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Ming-Ju Zhou
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Tao Yang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Jing Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Fan-Ping Meng
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Ming Shi
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Chao Zhang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
| | - Fu-Sheng Wang
- Senior Department of Infectious Diseases, The Fifth Medical Center of PLA General Hospital, National Clinical Research Center for Infectious Diseases, Beijing 100039, China; (X.F.); (J.-W.S.); (W.-J.C.); (M.-J.Z.); (T.Y.); (J.W.); (F.-P.M.); (M.S.)
- Medical School of Chinese PLA, Beijing 100853, China
| |
Collapse
|
43
|
Richards KA, Changrob S, Thomas PG, Wilson PC, Sant AJ. Lack of memory recall in human CD4 T cells elicited by the first encounter with SARS-CoV-2. iScience 2024; 27:109992. [PMID: 38868209 PMCID: PMC11166706 DOI: 10.1016/j.isci.2024.109992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 04/11/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024] Open
Abstract
The studies reported here focus on the impact of pre-existing CD4 T cell immunity on the first encounter with SARS-CoV-2. They leverage PBMC samples from plasma donors collected after a first SARS-CoV-2 infection, prior to vaccine availability and compared to samples collected prior to the emergence of SARS-CoV-2. Analysis of CD4 T cell specificity across the entire SARS-CoV-2 proteome revealed that the recognition of SARS-CoV-2-derived epitopes by CD4 memory cells prior to the pandemic are enriched for reactivity toward non-structural proteins conserved across endemic CoV strains. However, CD4 T cells after primary infection with SARS-CoV-2 focus on epitopes from structural proteins. We observed little evidence for preferential recall to epitopes conserved between SARS-CoV-2 and seasonal CoV, a finding confirmed through use of selectively curated conserved and SARS-unique peptides. Our data suggest that SARS-CoV-2 CD4 T cells elicited by the first infection are primarily established from the naive CD4 T cell pool.
Collapse
Affiliation(s)
- Katherine A. Richards
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Siriruk Changrob
- Drukier Institute for Children’s Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Patrick C. Wilson
- Drukier Institute for Children’s Health, Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, USA
| | - Andrea J. Sant
- David H. Smith Center for Vaccine Biology and Immunology, University of Rochester Medical Center, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
44
|
Gupta S, Su H, Agrawal S, Demirdag Y, Tran M, Gollapudi S. Adaptive Cellular Responses following SARS-CoV-2 Vaccination in Primary Antibody Deficiency Patients. Pathogens 2024; 13:514. [PMID: 38921811 PMCID: PMC11206773 DOI: 10.3390/pathogens13060514] [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: 04/25/2024] [Revised: 05/31/2024] [Accepted: 06/15/2024] [Indexed: 06/27/2024] Open
Abstract
Since the start of the COVID-19 pandemic, in a short span of 3 years, vaccination against SARS-CoV-2 has resulted in the end of the pandemic. Patients with inborn errors of immunity (IEI) are at an increased risk for SARS-CoV-2 infection; however, serious illnesses and mortality, especially in primary antibody deficiencies (PADs), have been lower than expected and lower than other high-risk groups. This suggests that PAD patients may mount a reasonable effective response to the SARS-CoV-2 vaccine. Several studies have been published regarding antibody responses, with contradictory reports. The current study is, perhaps, the most comprehensive study of phenotypically defined various lymphocyte populations in PAD patients following the SARS-CoV-2 vaccine. In this study, we examined, following two vaccinations and, in a few cases, prior to and following the 1st and 2nd vaccinations, subsets of CD4 and CD8 T cells (Naïve, TCM, TEM, TEMRA), T follicular helper cells (TFH1, TFH2, TFH17, TFH1/17), B cells (naïve, transitional, marginal zone, germinal center, IgM memory, switched memory, plasmablasts, CD21low), regulatory lymphocytes (CD4Treg, CD8Treg, TFR, Breg), and SARS-CoV-2-specific activation of CD4 T cells and CD8 T cells (CD69, CD137), SARS-CoV-2 tetramer-positive CD8 T cells, and CD8 CTL. Our data show significant alterations in various B cell subsets including Breg, whereas only a few subsets of various T cells revealed alterations. These data suggest that large proportions of PAD patients may mount significant responses to the vaccine.
Collapse
Affiliation(s)
- Sudhir Gupta
- Program in Primary Immunodeficiencies, Division of Basic and Clinical Immunology, University of California at Irvine, Irvine, CA 92697, USA; (H.S.); (S.A.); (Y.D.); (M.T.); (S.G.)
| | | | | | | | | | | |
Collapse
|
45
|
Bean DJ, Monroe J, Liang YM, Borberg E, Senussi Y, Swank Z, Chalise S, Walt D, Weinberg J, Sagar M. Heterotypic immunity from prior SARS-CoV-2 infection but not COVID-19 vaccination associates with lower endemic coronavirus incidence. Sci Transl Med 2024; 16:eado7588. [PMID: 38865483 PMCID: PMC11565543 DOI: 10.1126/scitranslmed.ado7588] [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: 02/19/2024] [Accepted: 05/07/2024] [Indexed: 06/14/2024]
Abstract
Immune responses from prior severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and COVID-19 vaccination mitigate disease severity, but they do not fully prevent subsequent infections, especially from genetically divergent strains. We examined the incidence of and immune differences against human endemic coronaviruses (eCoVs) as a proxy for response against future genetically heterologous coronaviruses (CoVs). We assessed differences in symptomatic eCoV and non-CoV respiratory disease incidence among those with known prior SARS-CoV-2 infection or previous COVID-19 vaccination but no documented SARS-CoV-2 infection or neither exposure. Retrospective cohort analyses suggest that prior SARS-CoV-2 infection, but not previous COVID-19 vaccination alone, associates with a lower incidence of subsequent symptomatic eCoV infection. There was no difference in non-CoV incidence, implying that the observed difference was eCoV specific. In a second cohort where both cellular and humoral immunity were measured, those with prior SARS-CoV-2 spike protein exposure had lower eCoV-directed neutralizing antibodies, suggesting that neutralization is not responsible for the observed decreased eCoV disease. The three groups had similar cellular responses against the eCoV spike protein and nucleocapsid antigens. However, CD8+ T cell responses to the nonstructural eCoV proteins nsp12 and nsp13 were higher in individuals with previous SARS-CoV-2 infection as compared with the other groups. This association between prior SARS-CoV-2 infection and decreased incidence of eCoV disease may therefore be due to a boost in CD8+ T cell responses against eCoV nsp12 and nsp13, suggesting that incorporation of nonstructural viral antigens in a future pan-CoV vaccine may improve vaccine efficacy.
Collapse
Affiliation(s)
- David J. Bean
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Janet Monroe
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Yan Mei Liang
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Ella Borberg
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Yasmeen Senussi
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Zoe Swank
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Sujata Chalise
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - David Walt
- Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02215, USA
| | - Janice Weinberg
- Department of Biostatistics, Boston University School of Public Health, Boston, MA 02118, USA
| | - Manish Sagar
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
46
|
Kozłowski P, Leszczyńska A, Ciepiela O. Long COVID Definition, Symptoms, Risk Factors, Epidemiology and Autoimmunity: A Narrative Review. AMERICAN JOURNAL OF MEDICINE OPEN 2024; 11:100068. [PMID: 39034937 PMCID: PMC11256271 DOI: 10.1016/j.ajmo.2024.100068] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 01/29/2024] [Accepted: 02/01/2024] [Indexed: 07/23/2024]
Abstract
The virus called SARS-CoV-2 emerged in 2019 and quickly spread worldwide, causing COVID-19. It has greatly impacted on everyday life, healthcare systems, and the global economy. In order to save as many lives as possible, precautions such as social distancing, quarantine, and testing policies were implemented, and effective vaccines were developed. A growing amount of data collected worldwide allowed the characterization of this new disease, which turned out to be more complex than other common respiratory tract infections. An increasing number of convalescents presented with a variety of nonspecific symptoms emerging after the acute infection. This possible new global health problem was identified and labelled as long COVID. Since then, a great effort has been made by clinicians and the scientific community to understand the underlying mechanisms and to develop preventive measures and effective treatment. The role of autoimmunity induced by SARS-CoV-2 infection in the development of long COVID is discussed in this review. We aim to deliver a description of several conditions with an autoimmune background observed in COVID-19 convalescents, including Guillain-Barré syndrome, antiphospholipid syndrome and related thrombosis, and Kawasaki disease highlighting a relationship between SARS-CoV-2 infection and the development of autoimmunity. However, further studies are required to determine its true clinical significance.
Collapse
Affiliation(s)
- Paweł Kozłowski
- Central Laboratory, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
| | - Aleksandra Leszczyńska
- Central Laboratory, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
| | - Olga Ciepiela
- Central Laboratory, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
- Department of Laboratory Medicine, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
47
|
Zhang J, Yue C, Lin Y, Tian J, Guo Y, Zhang D, Guo Y, Ye B, Chai Y, Qi J, Zhao Y, Gao GF, Sun Z, Liu J. Uncommon P1 Anchor-featured Viral T Cell Epitope Preference within HLA-A*2601 and HLA-A*0101 Individuals. Immunohorizons 2024; 8:415-430. [PMID: 38885041 PMCID: PMC11220742 DOI: 10.4049/immunohorizons.2400026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/09/2024] [Indexed: 06/20/2024] Open
Abstract
The individual HLA-related susceptibility to emerging viral diseases such as COVID-19 underscores the importance of understanding how HLA polymorphism influences peptide presentation and T cell recognition. Similar to HLA-A*0101, which is one of the earliest identified HLA alleles among the human population, HLA-A*2601 possesses a similar characteristic for the binding peptide and acts as a prevalent allomorph in HLA-I. In this study, we found that, compared with HLA-A*0101, HLA-A*2601 individuals exhibit distinctive features for the T cell responses to SARS-CoV-2 and influenza virus after infection and/or vaccination. The heterogeneous T cell responses can be attributed to the distinct preference of HLA-A*2601 and HLA-A*0101 to T cell epitope motifs with negative-charged residues at the P1 and P3 positions, respectively. Furthermore, we determined the crystal structures of the HLA-A*2601 complexed to four peptides derived from SARS-CoV-2 and human papillomavirus, with one structure of HLA-A*0101 for comparison. The shallow pocket C of HLA-A*2601 results in the promiscuous presentation of peptides with "switchable" bulged conformations because of the secondary anchor in the median portion. Notably, the hydrogen bond network formed between the negative-charged P1 anchors and the HLA-A*2601-specific residues lead to a "closed" conformation and solid placement for the P1 secondary anchor accommodation in pocket A. This insight sheds light on the intricate relationship between HLA I allelic allomorphs, peptide binding, and the immune response and provides valuable implications for understanding disease susceptibility and potential vaccine design.
Collapse
Affiliation(s)
- Jianing Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Can Yue
- CAS Key Laboratory of Infection and Immunity, National Laboratory of Macromolecules, Institute of Biophysics, Chinese Academy of Sciences (CAS), Beijing, China
| | - Yin Lin
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Jinmin Tian
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yuanyuan Guo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, Shandong Province, China
| | - Danni Zhang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yaxin Guo
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Beiwei Ye
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Yan Chai
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Jianxun Qi
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Yingze Zhao
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| | - George F. Gao
- CAS Key Laboratory of Pathogen Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences (CAS), Beijing, China
| | - Zeyu Sun
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Jun Liu
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases (NITFID), National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
- NHC Key Laboratory of Biosafety, Research Unit of Adaptive Evolution and Control of Emerging Viruses, Chinese Academy of Medical Sciences, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
48
|
Huang Y, Chen J, Chen S, Huang C, Li B, Li J, Jin Z, Zhang Q, Pan P, Du W, Liu L, Liu Z. Molecular characterization of SARS-CoV-2 nucleocapsid protein. Front Cell Infect Microbiol 2024; 14:1415885. [PMID: 38846351 PMCID: PMC11153676 DOI: 10.3389/fcimb.2024.1415885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/07/2024] [Indexed: 06/09/2024] Open
Abstract
Corona Virus Disease 2019 (COVID-19) is a highly prevalent and potent infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Until now, the world is still endeavoring to develop new ways to diagnose and treat COVID-19. At present, the clinical prevention and treatment of COVID-19 mainly targets the spike protein on the surface of SRAS-CoV-2. However, with the continuous emergence of SARS-CoV-2 Variants of concern (VOC), targeting the spike protein therapy shows a high degree of limitation. The Nucleocapsid Protein (N protein) of SARS-CoV-2 is highly conserved in virus evolution and is involved in the key process of viral infection and assembly. It is the most expressed viral structural protein after SARS-CoV-2 infection in humans and has high immunogenicity. Therefore, N protein as the key factor of virus infection and replication in basic research and clinical application has great potential research value. This article reviews the research progress on the structure and biological function of SARS-CoV-2 N protein, the diagnosis and drug research of targeting N protein, in order to promote researchers' further understanding of SARS-CoV-2 N protein, and lay a theoretical foundation for the possible outbreak of new and sudden coronavirus infectious diseases in the future.
Collapse
Affiliation(s)
- Yanping Huang
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Junkai Chen
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Siwei Chen
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Congcong Huang
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
| | - Bei Li
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Jian Li
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Zhixiong Jin
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Qiwei Zhang
- Central Laboratory, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Pan Pan
- Guangdong Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Weixing Du
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
| | - Long Liu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| | - Zhixin Liu
- Department of Infectious Diseases, Renmin Hospital, School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, China
- Shiyan Key Laboratory of Virology, Hubei University of Medicine, Shiyan, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, China
| |
Collapse
|
49
|
Zareein A, Mahmoudi M, Jadhav SS, Wilmore J, Wu Y. Biomaterial engineering strategies for B cell immunity modulations. Biomater Sci 2024; 12:1981-2006. [PMID: 38456305 PMCID: PMC11019864 DOI: 10.1039/d3bm01841e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
B cell immunity has a penetrating effect on human health and diseases. Therapeutics aiming to modulate B cell immunity have achieved remarkable success in combating infections, autoimmunity, and malignancies. However, current treatments still face significant limitations in generating effective long-lasting therapeutic B cell responses for many conditions. As the understanding of B cell biology has deepened in recent years, clearer regulation networks for B cell differentiation and antibody production have emerged, presenting opportunities to overcome current difficulties and realize the full therapeutic potential of B cell immunity. Biomaterial platforms have been developed to leverage these emerging concepts to augment therapeutic humoral immunity by facilitating immunogenic reagent trafficking, regulating T cell responses, and modulating the immune microenvironment. Moreover, biomaterial engineering tools have also advanced our understanding of B cell biology, further expediting the development of novel therapeutics. In this review, we will introduce the general concept of B cell immunobiology and highlight key biomaterial engineering strategies in the areas including B cell targeted antigen delivery, sustained B cell antigen delivery, antigen engineering, T cell help optimization, and B cell suppression. We will also discuss our perspective on future biomaterial engineering opportunities to leverage humoral immunity for therapeutics.
Collapse
Affiliation(s)
- Ali Zareein
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Mina Mahmoudi
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Shruti Sunil Jadhav
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
| | - Joel Wilmore
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
50
|
Kristensen NP, Dionisio E, Bentzen AK, Tamhane T, Kemming JS, Nos G, Voss LF, Hansen UK, Lauer GM, Hadrup SR. Simultaneous analysis of pMHC binding and reactivity unveils virus-specific CD8 T cell immunity to a concise epitope set. SCIENCE ADVANCES 2024; 10:eadm8951. [PMID: 38608022 PMCID: PMC11014448 DOI: 10.1126/sciadv.adm8951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 03/12/2024] [Indexed: 04/14/2024]
Abstract
CD8 T cells provide immunity to virus infection through recognition of epitopes presented by peptide major histocompatibility complexes (pMHCs). To establish a concise panel of widely recognized T cell epitopes from common viruses, we combined analysis of TCR down-regulation upon stimulation with epitope-specific enumeration based on barcode-labeled pMHC multimers. We assess CD8 T cell binding and reactivity for 929 previously reported epitopes in the context of 1 of 25 HLA alleles representing 29 viruses. The prevalence and magnitude of CD8 T cell responses were evaluated in 48 donors and reported along with 137 frequently recognized virus epitopes, many of which were underrepresented in the public domain. Eighty-four percent of epitope-specific CD8 T cell populations demonstrated reactivity to peptide stimulation, which was associated with effector and long-term memory phenotypes. Conversely, nonreactive T cell populations were associated primarily with naive phenotypes. Our analysis provides a reference map of epitopes for characterizing CD8 T cell responses toward common human virus infections.
Collapse
Affiliation(s)
- Nikolaj Pagh Kristensen
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Edoardo Dionisio
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Amalie Kai Bentzen
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Tripti Tamhane
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Janine Sophie Kemming
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Grigorii Nos
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Lasse Frank Voss
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Ulla Kring Hansen
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| | - Georg Michael Lauer
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Sine Reker Hadrup
- Section for Experimental and Translational Immunology, Department of Health Technology, Technical University of Denmark (DTU), Kongens Lyngby, Denmark
| |
Collapse
|