1
|
Taguchi A, Misumi F, Teraguchi S, Nagamatsu T, Sakakibara S, Otani T, Ichinose M, Priest D, Nakajima K, Nakamura J, Sawada R, Suzutani T, Ikeda T, Nagura Y, Iriyama T, Okuzaki D, Okazaki H, Wing JB, Hirota Y, Osuga Y. Multifaceted profiling of virus-specific CD8 T cells reveals distinct immune signatures against cytomegalovirus infection states during pregnancy. iScience 2025; 28:112416. [PMID: 40343282 PMCID: PMC12059710 DOI: 10.1016/j.isci.2025.112416] [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: 08/02/2024] [Revised: 12/12/2024] [Accepted: 04/08/2025] [Indexed: 05/11/2025] Open
Abstract
Anti-cytomegalovirus (CMV) serological testing, including the IgG avidity index (AI), is used to assess CMV infection phases during pregnancy. However, little is known about anti-CMV cellular immunity during pregnancy, particularly its relation to serological diagnosis. Herein, using MHC-dextramer single-cell RNA sequencing and flow cytometry, we characterized IE1 and pp65 CMV-antigen specific CD8 T cells from pregnant women with different anti-CMV serological patterns, including IgG+IgM+/AI-low, IgG+IgM+/AI-high, and IgG+IgM-. In IgG+IgM+/AI-low and IgG+IgM+/AI-high specimens, CMV-specific T cells consisted largely of effectors, with a minor but characteristic proportion of memory T cells, including HLA-DR-positive memory precursors and granzyme K-high memory cells reactive to IE1. Conversely, IgG+IgM- cases had a distinctive expansion of pp65-specific terminally differentiated T effector memory with a signature of convergent clonal selection. Our findings revealed that different CMV infection phases have characteristic patterns of CD8 cell phenotype and antigen recognition, potentially offering a new approach for assessing congenital infection risk.
Collapse
Affiliation(s)
- Ayumi Taguchi
- Laboratory of Human Single Cell Immunology, WPI Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Fumi Misumi
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Shunsuke Teraguchi
- Faculty of Data Science, Shiga University, Shiga 522-8522, Japan
- Department of Genome Informatics, Research Institute for Microbial Diseases, The University of Osaka, Osaka 565-0871, Japan
| | - Takeshi Nagamatsu
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
- Department of Obstetrics and Gynecology, International University of Health and Welfare, Narita 286-8686, Japan
| | - Shuhei Sakakibara
- Laboratory of Systems Immunology, WPI Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
- Graduate School of Medical Safety Management, Jikei University of Health Care Sciences, Osaka 532-0003, Japan
| | - Tomohiro Otani
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Mari Ichinose
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - David Priest
- Laboratory of Human Single Cell Immunology, WPI Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
| | - Kazuki Nakajima
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Junko Nakamura
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Laboratory Sciences, Department of Health Sciences, School of Health and Social Service, Saitama Prefectural University, Saitama 343-0036, Japan
| | - Ryoko Sawada
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Tatsuo Suzutani
- Department of Microbiology, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Toshiyuki Ikeda
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Yutaka Nagura
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Takayuki Iriyama
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Daisuke Okuzaki
- Laboratory of Human Immunology (Single Cell Genomics), WPI Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
- Human Single Cell Immunology Team, Center for Infectious Disease Education and Research, The University of Osaka, Osaka 565-0871, Japan
| | - Hitoshi Okazaki
- Department of Transfusion Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - James B. Wing
- Laboratory of Human Single Cell Immunology, WPI Immunology Frontier Research Center, The University of Osaka, Osaka 565-0871, Japan
- Human Single Cell Immunology Team, Center for Infectious Disease Education and Research, The University of Osaka, Osaka 565-0871, Japan
| | - Yasushi Hirota
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| | - Yutaka Osuga
- Department of Obstetrics and Gynecology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8654, Japan
| |
Collapse
|
2
|
Kwon DI, Mao T, Israelow B, Santos Guedes de Sá K, Dong H, Iwasaki A. Mucosal unadjuvanted booster vaccines elicit local IgA responses by conversion of pre-existing immunity in mice. Nat Immunol 2025:10.1038/s41590-025-02156-0. [PMID: 40360777 DOI: 10.1038/s41590-025-02156-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 04/08/2025] [Indexed: 05/15/2025]
Abstract
Mucosal delivery of vaccine boosters induces robust local protective immune responses even without any adjuvants. Yet, the mechanisms by which antigen alone induces mucosal immunity in the respiratory tract remain unclear. Here we show that an intranasal booster with an unadjuvanted recombinant SARS-CoV-2 spike protein, after intramuscular immunization with 1 μg of mRNA-LNP vaccine encoding the full-length SARS-CoV-2 spike protein (Pfizer/BioNTech BNT162b2), elicits protective mucosal immunity by retooling the lymph node-resident immune cells. On intranasal boosting, peripheral lymph node-primed B cells rapidly migrated to the lung through CXCR3-CXCL9 and CXCR3-CXCL10 signaling and differentiated into antigen-specific IgA-secreting plasma cells. Memory CD4+ T cells in the lung served as a natural adjuvant for developing mucosal IgA by inducing the expression of chemokines CXCL9 and CXCL10 for memory B cell recruitment. Furthermore, CD40 and TGFβ signaling had important roles in mucosal IgA development. Repeated mucosal boosting with an unadjuvanted protein amplified anamnestic IgA responses in both the upper and the lower respiratory tracts. These findings help explain why nasal boosters do not require an adjuvant to induce robust mucosal immunity at the respiratory mucosa and can be used to design safe and effective vaccines against respiratory pathogens.
Collapse
Affiliation(s)
- Dong-Il Kwon
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Tianyang Mao
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Benjamin Israelow
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
- Section of Infectious Diseases, Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Keyla Santos Guedes de Sá
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Huiping Dong
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA
| | - Akiko Iwasaki
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA.
- Center for Infection and Immunity, Yale School of Medicine, New Haven, CT, USA.
- Howard Hughes Medical Institute, Chevy Chase, MD, USA.
| |
Collapse
|
3
|
Masuo Y, Lu D, Matsuyama J, Shin EC, Ueno H. Human immunology soars in Japan. Nat Immunol 2025; 26:653-656. [PMID: 40186069 DOI: 10.1038/s41590-025-02122-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Affiliation(s)
- Yuki Masuo
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
| | - Dongyun Lu
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan
- Kyoto University Immunomonitoring Center (KIC), Kyoto University, Kyoto, Japan
| | - Joey Matsuyama
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eui-Cheol Shin
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hideki Ueno
- Department of Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.
- Kyoto University Immunomonitoring Center (KIC), Kyoto University, Kyoto, Japan.
| |
Collapse
|
4
|
Zhao J, Xiao P, Xin A, Zhu H, Wang H, Xiao J, Gao H. Preliminary evaluation of a novel serotype O foot-and-mouth disease mRNA vaccine. Front Microbiol 2025; 16:1503191. [PMID: 40356647 PMCID: PMC12067417 DOI: 10.3389/fmicb.2025.1503191] [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: 09/28/2024] [Accepted: 04/07/2025] [Indexed: 05/15/2025] Open
Abstract
Foot-and-mouth disease virus (FMDV) is one of the most significant animal pathogens worldwide, severely impacting the health and productivity of pigs, cattle, sheep, and other ungulates. Although the traditional vaccines have played a crucial role in epidemic control, inactivated vaccines face persistent challenges concerning the potential for virus dissemination and pressures from serotype and subtype matching. However, the manufacture of attenuated vaccines is forbidden, and the efficiency of alternative vaccines for immune protection is still inadequate. Consequently, there exists an urgent need for safer and more effective innovative vaccines in animal husbandry. In this study, we aimed to develop a lipid nanoparticle mRNA vaccine based on VP1-3A-3D epitopes from serotype O FMD and to verify its specific expression within cytoplasmic and injection sites. Our findings demonstrated that mRNA transfected into primary spleen cells derived from guinea pigs induced cytokine release, promoted differentiation of both CD4+ T and CD8+ T lymphocytes, and enhanced lymphocyte proliferation rates. Following immunization of mRNA vaccine in guinea pigs, we observed increased differentiation of both CD4+ T and CD8+ T cells, alongside elevated levels of cytokine secretion. Additionally, this vaccination induced the production of specific IgG antibodies as well as neutralizing antibodies. Importantly, our vaccine provided complete protection for all six guinea pigs against a lethal challenge of 100 GPID50, with histopathological scores indicating protection equivalent to that conferred by the inactivated vaccine. The viral load results demonstrated that the vaccine group significantly reduced viral copy numbers in serum and effectively decreased the concentration of the inflammatory cytokine IL-1β. Furthermore, during the pre-immune phase following vaccination with the mRNA vaccine in pigs, heightened cytokine secretion was observed, along with the inhibition of viral replication. Simultaneously, the neutralizing antibody titer in the serum remained stable over 4 months. Immunofluorescence analysis of spleen tissues from both guinea pigs and pigs demonstrated marked activation and increased expression of CD4+ and CD8+ T lymphocytes, as well as macrophages, in the mRNA vaccine group. In summary, this study suggests that the serotype O FMD mRNA vaccine is a promising candidate for further development in the fight against FMDV.
Collapse
Affiliation(s)
- Jingang Zhao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
- Yunnan Tropical and Subtropical Animal Virus Disease Laboratory, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
| | - Peng Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Aiguo Xin
- Yunnan Tropical and Subtropical Animal Virus Disease Laboratory, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
- National Foot-and-Mouth Disease Para-Reference Laboratory (Kunming), Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
| | - Heran Zhu
- Yunnan Tropical and Subtropical Animal Virus Disease Laboratory, Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
- National Foot-and-Mouth Disease Para-Reference Laboratory (Kunming), Yunnan Academy of Animal Husbandry and Veterinary Sciences, Kunming, China
| | - Hao Wang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Jinlong Xiao
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Hong Gao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| |
Collapse
|
5
|
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
|
6
|
Wen X, Hu AK, Presnell SR, Ford ES, Koelle DM, Kwok WW. Longitudinal single cell profiling of epitope specific memory CD4+ T cell responses to recombinant zoster vaccine. Nat Commun 2025; 16:2332. [PMID: 40057520 PMCID: PMC11890790 DOI: 10.1038/s41467-025-57562-7] [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: 04/01/2024] [Accepted: 02/25/2025] [Indexed: 05/13/2025] Open
Abstract
Vaccination leads to rapid expansion of antigen-specific T cells within in the first few days. However, understanding of transcriptomic changes and fates of antigen-specific T cells upon vaccination remains limited. Here, we investigate the fate of memory CD4+ T cells upon reactivation to recombinant zoster vaccine for shingles at cellular and transcriptional levels. We show that glycoprotein E-specific memory CD4+ T cells respond strongly, their frequencies remain high, and they retain markers of cell activation one year following vaccination. Memory T cells with the most dominant TCR clonotype pre-vaccination remain prevalent at year one post-vaccination. These data implicate a major role for pre-existing memory T cells in perpetuating immune repertoires upon re-encountering cognate antigens. Differential gene expression indicates that cells post-vaccination are distinct from cells at baseline, suggesting committed memory T cells display transcriptional changes upon vaccination that could alter their responses against cognate immunogens.
Collapse
Affiliation(s)
- Xiaomin Wen
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
- AstraZeneca Pharmaceuticals, Gaithersburg, MD, USA
| | - Alex K Hu
- Center for Systems Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Scott R Presnell
- Center for Systems Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Emily S Ford
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - David M Koelle
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Fred Hutchinson Cancer Center, Seattle, WA, USA
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - William W Kwok
- Center for Translational Immunology, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA.
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA.
| |
Collapse
|
7
|
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
|
8
|
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
|
9
|
Sun Y, Sen S, Parmar R, Arakawa-Hoyt J, Cappelletti M, Rossetti M, Gjertson DW, Sigdel TK, Sarwal MM, Schaenman JM, Bunnapradist S, Lanier LL, Pickering H, Reed EF. Cytotoxic KLRG1+ IL-7R- effector CD8+ T cells distinguish kidney transplant recipients controlling cytomegalovirus reactivation. Front Immunol 2025; 16:1542531. [PMID: 40028342 PMCID: PMC11868092 DOI: 10.3389/fimmu.2025.1542531] [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: 12/10/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Cytomegalovirus (CMV) viremia remains a major contributor to clinical complications in solid organ transplant (SOT) patients, including organ injury, morbidity and mortality. Given their critical role in antiviral defense, CD8+ T cells are essential for protective immunity against CMV. Methods Using single-cell RNA sequencing, we investigated the transcriptional signatures and developmental lineages of CD8+ T cells in eight immunosuppressed kidney transplant recipients (KTRs) who received organs from CMV-seropositive donors. Results were validated in a cohort of 62 KTRs using immunophenotyping. Results Our data revealed a significant influence of CMV serostatus on transcriptional variance of CD8+ memory T cells, associating with the first principal component from a global analysis of CD8+ T cells (p =0.0406), forming a continuum with five principal differentiation trajectories driven by CMV primary infection or reactivation. Following CMV primary infection, CD8+ T cells were hallmarked by restrained effector-memory differentiation. CD8+ T cells during CMV reactivation diverged non-linearly into senescent-like cells with signatures of arrested cell cycle, diminished translational activity and downregulated ZNF683 and longitudinally expanding effector cells with robust cytotoxic potential and upregulated ZNF683, acting as a reservoir for long-lived effector cells supporting long-term protection. Notably, CD28lo KLRG1hi IL-7R (CD127)lo HLA-DRhi CD8+ T cells present prior to the detection of viremia in CMV-seropositive patients emerged as a key feature distinguishing patients who did or did not undergo CMV reactivation after prophylaxis discontinuation (p =0.0163). Frequencies of these cells were also positively correlated with CMV-stimulated secretion of IFN-γ (p =0.0494), TNF-α (p =0.0358), MIP-1α (p =0.0262), MIP-1β (p =0.0043). Discussion These results provide insights into the transcriptional regulation that influences the generation of CD8+ T cell immunity to CMV and may inform strategics for monitoring host immune response to CMV to better identify and introduce therapeutic intervention to patients at risk of developing clinically significant CMV viremia.
Collapse
Affiliation(s)
- Yumeng Sun
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Subha Sen
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rajesh Parmar
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Janice Arakawa-Hoyt
- Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States
| | - Monica Cappelletti
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maura Rossetti
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - David W. Gjertson
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tara K. Sigdel
- Department of Surgery, Division of Multi Organ Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Minnie M. Sarwal
- Department of Surgery, Division of Multi Organ Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Joanna M. Schaenman
- Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Suphamai Bunnapradist
- Division of Nephrology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
10
|
Xie J, Chen DG, Chour W, Ng RH, Zhang R, Yuan D, Choi J, McKasson M, Troisch P, Smith B, Jones L, Webster A, Rasheed Y, Li S, Edmark R, Hong S, Murray KM, Logue JK, Franko NM, Lausted CG, Piening B, Algren H, Wallick J, Magis AT, Watanabe K, Mease P, Greenberg PD, Chu H, Goldman JD, Su Y, Heath JR. APMAT analysis reveals the association between CD8 T cell receptors, cognate antigen, and T cell phenotype and persistence. Nat Commun 2025; 16:1402. [PMID: 39915487 PMCID: PMC11802929 DOI: 10.1038/s41467-025-56659-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025] Open
Abstract
Elucidating the relationships between a class I peptide antigen, a CD8 T cell receptor (TCR) specific to that antigen, and the T cell phenotype that emerges following antigen stimulation, remains a mostly unsolved problem, largely due to the lack of large data sets that can be mined to resolve such relationships. Here, we describe Antigen-TCR Pairing and Multiomic Analysis of T-cells (APMAT), an integrated experimental-computational framework designed for the high-throughput capture and analysis of CD8 T cells, with paired antigen, TCR sequence, and single-cell transcriptome. Starting with 951 putative antigens representing a comprehensive survey of the SARS-CoV-2 viral proteome, we utilize APMAT for the capture and single cell analysis of CD8 T cells from 62 HLA A*02:01 COVID-19 participants. We leverage this comprehensive dataset to integrate with peptide antigen properties, TCR CDR3 sequences, and T cell phenotypes to show that distinct physicochemical features of the antigen-TCR pairs strongly associate with both T cell phenotype and T cell persistence. This analysis suggests that CD8 T cell phenotype following antigen stimulation is at least partially deterministic, rather than the result of stochastic biological properties.
Collapse
Affiliation(s)
- Jingyi Xie
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, 98195, USA
| | - Daniel G Chen
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - William Chour
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Rachel H Ng
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Rongyu Zhang
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Dan Yuan
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Jongchan Choi
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | | | | | - Brett Smith
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Lesley Jones
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | | | - Yusuf Rasheed
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Sarah Li
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Rick Edmark
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Sunga Hong
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Kim M Murray
- Institute for Systems Biology, Seattle, WA, 98109, USA
| | - Jennifer K Logue
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | - Nicholas M Franko
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | | | - Brian Piening
- Providence Health & Services, Seattle, WA, 99109, USA
| | - Heather Algren
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Julie Wallick
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | | | - Kino Watanabe
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | - Phil Mease
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Philip D Greenberg
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
- Department of Immunology, University of Washington, Seattle, WA, 98109, USA
| | - Helen Chu
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
| | - Jason D Goldman
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA, 98109, USA
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Yapeng Su
- Institute for Systems Biology, Seattle, WA, 98109, USA
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - James R Heath
- Institute for Systems Biology, Seattle, WA, 98109, USA.
| |
Collapse
|
11
|
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
|
12
|
da Graça CG, Sheikh AA, Newman DM, Wen L, Li S, Shen J, Zhang Y, Gabriel SS, Chisanga D, Seow J, Poch A, Rausch L, Nguyen MHT, Singh J, Su CH, Cluse LA, Tsui C, Burn TN, Park SL, Von Scheidt B, Mackay LK, Vasanthakumar A, Bending D, Shi W, Cui W, Schröder J, Johnstone RW, Kallies A, Utzschneider DT. Stem-like memory and precursors of exhausted T cells share a common progenitor defined by ID3 expression. Sci Immunol 2025; 10:eadn1945. [PMID: 39888981 PMCID: PMC7617396 DOI: 10.1126/sciimmunol.adn1945] [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: 11/27/2023] [Accepted: 12/23/2024] [Indexed: 02/02/2025]
Abstract
Stem-like T cells are attractive immunotherapeutic targets in patients with cancer given their ability to proliferate and differentiate into effector progeny. Thus, identifying T cells with enhanced stemness and understanding their developmental requirements are of broad clinical and therapeutic interest. Here, we demonstrate that during acute infection, the transcriptional regulator inhibitor of DNA binding 3 (ID3) identifies stem-like T cells that are uniquely adapted to generate precursors of exhausted T (Tpex) cells in response to chronic infection or cancer. Expression of ID3 itself enables Tpex cells to sustain T cell responses in chronic infection or cancer, whereas loss of ID3 results in impaired maintenance of CD8 T cell immunity. Furthermore, we demonstrate that interleukin-1 (IL-1) family members, including IL-36β and IL-18, promote the generation of ID3+ T cells that mediate superior tumor control. Overall, we identify ID3 as a common denominator of stem-like T cells in both acute and chronic infections that is specifically required to sustain T cell responses to chronic stimulation.
Collapse
Affiliation(s)
- Catarina Gago da Graça
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Amania A. Sheikh
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Dane M. Newman
- Cancer Biology and Therapeutics, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Lifen Wen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Sining Li
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Jian Shen
- Department of Pathology, Northwestern University, Chicago, IL
| | - Yuqi Zhang
- Department of Pathology, Northwestern University, Chicago, IL
| | - Sarah S. Gabriel
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - David Chisanga
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
| | - Justine Seow
- Computational Sciences Initiative (CSI), The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Annika Poch
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Lisa Rausch
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Minh-Hanh T. Nguyen
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Jayendra Singh
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
| | - Chun-Hsi Su
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Leonie A. Cluse
- Cancer Biology and Therapeutics, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Carlson Tsui
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Thomas N. Burn
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Simone L. Park
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Bianca Von Scheidt
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Laura K. Mackay
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | | | - David Bending
- Department of Immunology and Immunotherapy, College of Medicine and Health, University of Birmingham, BirminghamB15 2TT, UK
| | - Wei Shi
- Olivia Newton-John Cancer Research Institute, Heidelberg, Australia
- School of Cancer Medicine, La Trobe University, Heidelberg, Australia
| | - Weiguo Cui
- Department of Pathology, Northwestern University, Chicago, IL
| | - Jan Schröder
- Computational Sciences Initiative (CSI), The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Ricky W. Johnstone
- Cancer Biology and Therapeutics, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Axel Kallies
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Daniel T. Utzschneider
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| |
Collapse
|
13
|
Masuda K, Iketani S, Liu L, Huang J, Qiao Y, Shah J, McNairy ML, Groso C, Ricupero C, Loffredo LF, Wang Q, Purpura L, Coelho-dos-Reis JGA, Sheng Z, Yin MT, Tsuji M. Distinct CD8 + T-cell types Associated with COVID-19 Severity in Unvaccinated HLA-A2 + Patients. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.12.632164. [PMID: 39868279 PMCID: PMC11761488 DOI: 10.1101/2025.01.12.632164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Although emerging data have revealed the critical role of memory CD8+ T cells in preventing and controlling SARS-CoV-2 infection, virus-specific CD8+ T-cell responses against SARS-CoV-2 and its memory and innate-like subsets in unvaccinated COVID-19 patients with various disease manifestations in an HLA-restricted fashion remain to be understood. Here, we show the strong association of protective cellular immunity with mild COVID-19 and unique cell types against SARS-CoV-2 virus in an HLA-A2 restricted manner. ELISpot assays reveal that SARS-CoV-2-specific CD8+ T-cell responses in mild COVID-19 patients are significantly higher than in severe patients, whereas neutralizing antibody responses against SARS-CoV-2 virus significantly correlate with disease severity. Single-cell analyses of HLA-A2-restricted CD8+ T cells, which recognize highly conserved immunodominant SARS-CoV-2-specific epitopes, demonstrate divergent profiles in unvaccinated patients with mild versus severe disease. CD8+ T-cell types including cytotoxic KLRB1 + CD8αα cells with innate-like T-cell signatures, IFNG hi ID3 hi memory cells and IL7R + proliferative stem cell-like memory cells are preferentially observed in mild COVID-19, whereas distinct terminally-differentiated T-cell subsets are predominantly detected in severe COVID-19: highly activated FASL hi T-cell subsets and early-terminated or dysfunctional IL4R + GATA3 + stem cell-like memory T-cell subset. In conclusion, our findings suggest that unique and contrasting SARS-CoV-2-specific CD8+ T-cell profiles may dictate COVID-19 severity.
Collapse
Affiliation(s)
- Kazuya Masuda
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Sho Iketani
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Lihong Liu
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jing Huang
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Yujie Qiao
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jayesh Shah
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Meredith L. McNairy
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Christine Groso
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Christopher Ricupero
- Center for Dental & Craniofacial Regeneration, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Lucas F. Loffredo
- Department of Microbiology and Immunology, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Qian Wang
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Lawrence Purpura
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | | | - Zizhang Sheng
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Michael T Yin
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
- Lead contact
| |
Collapse
|
14
|
Xie J, Chen DG, Chour W, Ng RH, Zhang R, Yuan D, Choi J, McKasson M, Troisch P, Smith B, Jones L, Webster A, Rasheed Y, Li S, Edmark R, Hong S, Murray KM, Logue JK, Franko NM, Lausted CG, Piening B, Algren H, Wallick J, Magis AT, Watanabe K, Mease P, Greenberg PD, Chu H, Goldman JD, Su Y, Heath JR. APMAT analysis reveals the association between CD8 T cell receptors, cognate antigen, and T cell phenotype and persistence. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.08.631993. [PMID: 39829843 PMCID: PMC11741388 DOI: 10.1101/2025.01.08.631993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Elucidating the relationships between a class I peptide antigen, a CD8 T cell receptor (TCR) specific to that antigen, and the T cell phenotype that emerges following antigen stimulation, remains a mostly unsolved problem, largely due to the lack of large data sets that can be mined to resolve such relationships. Here, we describe Antigen-TCR Pairing and Multiomic Analysis of T-cells (APMAT), an integrated experimental-computational framework designed for the high-throughput capture and analysis of CD8 T cells, with paired antigen, TCR sequence, and single-cell transcriptome. Starting with 951 putative antigens representing a comprehensive survey of the SARS-CoV-2 viral proteome, we utilize APMAT for the capture and single cell analysis of CD8 T cells from 62 HLA A*02:01 COVID-19 participants. We leverage this unique, comprehensive dataset to integrate with peptide antigen properties, TCR CDR3 sequences, and T cell phenotypes to show that distinct physicochemical features of the antigen-TCR pairs strongly associate with both T cell phenotype and T cell persistence. This analysis suggests that CD8+ T cell phenotype following antigen stimulation is at least partially deterministic, rather than the result of stochastic biological properties.
Collapse
Affiliation(s)
- Jingyi Xie
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, 98195, USA
| | - Daniel G. Chen
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - William Chour
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Rachel H. Ng
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Rongyu Zhang
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Dan Yuan
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA
| | - Jongchan Choi
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | | | | | - Brett Smith
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Lesley Jones
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | | | - Yusuf Rasheed
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Sarah Li
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Rick Edmark
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Sunga Hong
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Kim M. Murray
- Institute of Systems Biology, Seattle, WA, 98109, USA
| | - Jennifer K. Logue
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Nicholas M. Franko
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | | | - Brian Piening
- Providence Health & Services, Seattle, WA, 99109, USA
| | - Heather Algren
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Julie Wallick
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | | | - Kino Watanabe
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Phil Mease
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Philip D. Greenberg
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
- Department of Immunology, University of Washington, Seattle, WA 98109, USA
| | - Helen Chu
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
| | - Jason D. Goldman
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Medicine, Allergy and Infectious Diseases, University of Washington, Seattle, WA 98109, USA
- Providence Health & Services, Seattle, WA, 99109, USA
- Providence Swedish Medical Center, Seattle, WA, 98122, USA
| | - Yapeng Su
- Institute of Systems Biology, Seattle, WA, 98109, USA
- Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- These authors jointly-supervised the work
| | - James R. Heath
- Institute of Systems Biology, Seattle, WA, 98109, USA
- These authors jointly-supervised the work
- Corresponding author, Leading contact
| |
Collapse
|
15
|
Zhang Y, Han S, Sun Q, Liu T, Wen Z, Yao M, Zhang S, Duan Q, Zhang X, Pang B, Kou Z, Jiang X. Single-cell transcriptome atlas of peripheral immune features to Omicron breakthrough infection under booster vaccination strategies. Front Immunol 2025; 15:1460442. [PMID: 39835127 PMCID: PMC11743671 DOI: 10.3389/fimmu.2024.1460442] [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: 07/06/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025] Open
Abstract
Introduction The high percentage of Omicron breakthrough infection in vaccinees is an emerging problem, of which we have a limited understanding of the phenomenon. Methods We performed single-cell transcriptome coupled with T-cell/B-cell receptor (TCR/BCR) sequencing in 15 peripheral blood mononuclear cell (PBMC) samples from Omicron infection and naïve with booster vaccination. Results We found that after breakthrough infection, multiple cell clusters showed activation of the type I IFN pathway and widespread expression of Interferon-stimulated genes (ISGs); T and B lymphocytes exhibited antiviral and proinflammatory-related differentiation features with pseudo-time trajectories; and large TCR clonal expansions were concentrated in effector CD8 T cells, and clonal expansions of BCRs showed a preference for IGHV3. In addition, myeloid cells in the BA.5.2 breakthrough infection with the fourth dose of aerosolized Ad5-nCoV were characterized by enhanced proliferation, chemotactic migration, and antigen presentation. Discussion Collectively, our study informs the comprehensive understandings of immune characterization for Omicron breakthrough infection, revealing the positive antiviral potential induced by booster doses of vaccine and the possible "trained immunity" phenomenon in the fourth dose of aerosolized Ad5-nCoV, providing a basis for the selection of vaccination strategies.
Collapse
MESH Headings
- Humans
- Immunization, Secondary
- COVID-19/immunology
- COVID-19/prevention & control
- COVID-19/genetics
- Single-Cell Analysis
- Transcriptome
- SARS-CoV-2/immunology
- COVID-19 Vaccines/immunology
- COVID-19 Vaccines/administration & dosage
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Leukocytes, Mononuclear/immunology
- Vaccination
- B-Lymphocytes/immunology
- Breakthrough Infections
Collapse
Affiliation(s)
- Yuwei Zhang
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Shanshan Han
- School of Public Health and Health Management, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Qingshuai Sun
- School of Public Health and Health Management, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Tao Liu
- Department of Infectious Disease Control, Yantai Center for Disease Control and Prevention, Yantai, Shandong, China
| | - Zixuan Wen
- School of Public Health, Weifang Medical University, Weifang, Shandong, China
| | - Mingxiao Yao
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Shu Zhang
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Qing Duan
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Xiaomei Zhang
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Bo Pang
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Zengqiang Kou
- Infectious Disease Prevention and Control Section, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| | - Xiaolin Jiang
- School of Public Health and Health Management, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- School of Public Health, Weifang Medical University, Weifang, Shandong, China
- Shandong Provincial Key Laboratory of Infectious Disease Control and Prevention, Shandong Center for Disease Control and Prevention, Jinan, Shandong, China
| |
Collapse
|
16
|
O'Donnell TJ, Kanduri C, Isacchini G, Limenitakis JP, Brachman RA, Alvarez RA, Haff IH, Sandve GK, Greiff V. Reading the repertoire: Progress in adaptive immune receptor analysis using machine learning. Cell Syst 2024; 15:1168-1189. [PMID: 39701034 DOI: 10.1016/j.cels.2024.11.006] [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: 06/23/2024] [Revised: 08/16/2024] [Accepted: 11/14/2024] [Indexed: 12/21/2024]
Abstract
The adaptive immune system holds invaluable information on past and present immune responses in the form of B and T cell receptor sequences, but we are limited in our ability to decode this information. Machine learning approaches are under active investigation for a range of tasks relevant to understanding and manipulating the adaptive immune receptor repertoire, including matching receptors to the antigens they bind, generating antibodies or T cell receptors for use as therapeutics, and diagnosing disease based on patient repertoires. Progress on these tasks has the potential to substantially improve the development of vaccines, therapeutics, and diagnostics, as well as advance our understanding of fundamental immunological principles. We outline key challenges for the field, highlighting the need for software benchmarking, targeted large-scale data generation, and coordinated research efforts.
Collapse
Affiliation(s)
| | - Chakravarthi Kanduri
- Department of Informatics, University of Oslo, Oslo, Norway; UiO:RealArt Convergence Environment, University of Oslo, Oslo, Norway
| | | | | | - Rebecca A Brachman
- Imprint Labs, LLC, New York, NY, USA; Cornell Tech, Cornell University, New York, NY, USA
| | | | - Ingrid H Haff
- Department of Mathematics, University of Oslo, 0371 Oslo, Norway
| | - Geir K Sandve
- Department of Informatics, University of Oslo, Oslo, Norway; UiO:RealArt Convergence Environment, University of Oslo, Oslo, Norway
| | - Victor Greiff
- Imprint Labs, LLC, New York, NY, USA; Department of Immunology, University of Oslo and Oslo University Hospital, Oslo, Norway.
| |
Collapse
|
17
|
Li Y, Lin Y, Yi Y, Zhu N, Cui X, Li X. COVID-19 Vaccination and Transient Increase in CD4/CD8 Cell Counts in People with HIV: Evidence from China. Vaccines (Basel) 2024; 12:1365. [PMID: 39772028 PMCID: PMC11680300 DOI: 10.3390/vaccines12121365] [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: 10/17/2024] [Revised: 11/26/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
Objectives: Accumulating evidence has confirmed the efficacy and safety of COVID-19 vaccines against SARS-CoV-2 infection. However, the effect of COVID-19 vaccination on immuno-virological parameters in people with HIV (PWH) is uncertain. Methods: A total of 372 PWH treated at Beijing Ditan Hospital were included. Unvaccinated PWH were matched 1:3 with vaccinated PWH using a propensity score matching algorithm. Differences in immuno-virological markers between the matched groups were analyzed. The Wilcoxon signed rank test was used to test for changes in CD4 and CD8 counts and HIV viral load over two months around vaccination. In addition, we investigated the long-term changes in HIV-related markers in different vaccination dose groups and in the entire vaccinated population. Results: Vaccinated PWH had a higher CD4/CD8 ratio (0.64 (0.49, 0.78) vs. 0.80 (0.56, 1.03), p = 0.037) than unvaccinated PWH within a two-month window after the third dose. There were 337 PWH who received COVID-19 vaccination, and 73.9% (n = 249) received three doses of vaccine. We observed a transient increase in CD4 count and CD4/CD8 ratio within a two-month window after vaccination, especially after the second dose (CD4 count: 583.5 (428.5, 706.8) vs. 618.0 (452.0, 744.0), p = 0.018; CD4/CD8 ratio: 0.70 (0.50, 0.91) vs. 0.71 (0.53, 0.96), p < 0.001)) and the third dose (CD4 count: 575.5 (435.5, 717.0) vs. 577.5 (440.8, 754.8), p = 0.001; CD4/CD8 ratio: 0.70 (0.52, 0.93) vs. 0.79 (0.53, 1.00), p < 0.001)). Recent CD4 counts and CD4/CD8 ratios were lower than after COVID-19 but remained higher than before COVID-19 in vaccinated PWH. In addition, COVID-19 vaccination had no negative effect on HIV viral load. Conclusions: A transient increase in CD4 count and CD4/CD8 ratio was observed after COVID-19 vaccination. However, the enhanced cellular immune response induced by vaccination may diminish over time and return to normal levels. There is no adverse effect of vaccination on HIV viral load.
Collapse
Affiliation(s)
- Yanyan Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Yingying Lin
- Center of Integrative Medicine, Peking University Ditan Teaching Hospital, Beijing 100015, China;
| | - Yunyun Yi
- Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Senior Department of Tuberculosis, The Eighth Medical Center of PLA General Hospital, PLA General Hospital, Beijing 100853, China;
| | - Na Zhu
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xinyu Cui
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xin Li
- Center of Integrative Medicine, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China; (Y.L.); (N.Z.); (X.C.)
- National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
- Center of Integrative Medicine, Peking University Ditan Teaching Hospital, Beijing 100015, China;
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
Wang L, Xu R, Huang D, Peng P, Sun K, Hu J, Liu BZ, Fang L, Zhang L, Sun X, Gu F, Tang N, Huang AL, Lin X, Lan X. Identification of virus epitopes and reactive T-cell receptors from memory T cells without peptide synthesis. Commun Biol 2024; 7:1432. [PMID: 39496850 PMCID: PMC11535475 DOI: 10.1038/s42003-024-07048-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 10/10/2024] [Indexed: 11/06/2024] Open
Abstract
Identifying epitopes and their corresponding T-cell receptor (TCR) sequences is crucial in the face of rapidly mutating viruses. Peptide synthesis is often required to confirm the exact epitope sequences, which is time-consuming and expensive. In this study, we introduce a scalable workflow to identify the exact sequences of virus epitopes and reactive TCRs targeting the epitopes from memory T cells. Following the narrowing down of epitopes to specific regions via the tandem minigene (TMG) system, our workflow incorporates the utilization of peptide-major histocompatibility complex-displaying yeasts (pMHC-displaying yeasts) to rapidly screen immunogenic epitopes' precise sequences, obviating the necessity for the chemical synthesis of peptides. Focusing on SARS-CoV-2, we identify the precise sequences of reactive TCRs, targeting conserved epitopes across the Coronaviridae family, from the blood of COVID-19-recovered individuals over 8 months. Notably, we reveal that at least 75% (6/8) of the tested donors harbor T cells targeting a shared epitope, KTFPPTEPK, derived from the N protein. Furthermore, several identified TCRs exhibit cross-reactivity to mutant epitopes, suggesting a potential mechanism for sustained T-cell responses against emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Lihui Wang
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Runda Xu
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Daosheng Huang
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Pai Peng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Keyong Sun
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Jie Hu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China
| | - Bei-Zhong Liu
- Yong-Chuan Hospital of Chongqing Medical University, Chongqing, China
| | - Liang Fang
- Yong-Chuan Hospital of Chongqing Medical University, Chongqing, China
| | - Liwen Zhang
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Xin Sun
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China
| | - Fei Gu
- Alibaba Group, 311121, Hangzhou, China.
| | - Ni Tang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
| | - Ai-Long Huang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, the Second Affiliated Hospital, Chongqing Medical University, 400016, Chongqing, China.
| | - Xin Lin
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China.
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.
| | - Xun Lan
- Department of Basic Medical Science, School of Medicine, Tsinghua University, 100084, Beijing, China.
- Tsinghua-Peking Joint Center for Life Sciences, Tsinghua University, 100084, Beijing, China.
- MOE Key Laboratory of Bioinformatics, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
21
|
Saiag E, Gamzu R, Padova H, Paran Y, Goldiner I, Cohen N, Bomze D. Antibody Response After a Fifth Dose (Third Booster) of BNT162b2 mRNA COVID-19 Vaccine in Healthcare Workers. J Clin Med 2024; 13:6538. [PMID: 39518677 PMCID: PMC11546691 DOI: 10.3390/jcm13216538] [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: 08/10/2024] [Revised: 10/19/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Although a fourth dose of SARS-CoV-2 vaccine was shown to be effective, the immunogenicity of a fifth dose in immunocompetent individuals had not been well described. This was a prospective observational cohort study of previously vaccinated healthcare workers at a single tertiary hospital in Israel. Individuals were administered up to three booster doses of the BNT162b2 mRNA vaccine (i.e., up to five overall doses), during the period between July 2021 and January 2023. Immunogenicity was assessed using the SARS-CoV-2 IgG (sCOVG) semi-quantitative assay, performed at several time points. The cohort consisted of 162 individuals (median age 69 years, 62% female). Of these, 104 (64%) received four doses and 58 (36%) received five doses. Anti-SARS-CoV-2 antibody levels increased in all cases, regardless of the baseline levels. The fold-change increase in the mean sCOVG index was 29.2 (SD 2.6) after the third vaccine, 3.8 (SD 2.4) after the fourth vaccine, and 3.6 (SD 3.0) after the fifth vaccine. A waning effect over time was seen in 78% and 43% of participants for the third and fourth doses, respectively. Adverse events following the fifth dose were limited and mild. Similar to previous booster vaccines, a fifth dose of BNT162b2 is immunogenic and safe in healthy individuals, although the clinical implications remain unclear.
Collapse
Affiliation(s)
- Esther Saiag
- Division of Information Systems and Operations, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel;
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel (N.C.)
| | - Ronni Gamzu
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel (N.C.)
- Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Hagit Padova
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel (N.C.)
- Department of Quality and Patient Safety, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Yael Paran
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel (N.C.)
- Department of Infectious Diseases and Infection Control, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Ilana Goldiner
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel (N.C.)
- Division of Clinical Laboratories, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - Neta Cohen
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel (N.C.)
- Emergency Department, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| | - David Bomze
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel (N.C.)
- Division of Dermatology, Tel Aviv Sourasky Medical Center, Tel Aviv 6423906, Israel
| |
Collapse
|
22
|
Henderson J, Nagano Y, Milighetti M, Tiffeau-Mayer A. Limits on inferring T cell specificity from partial information. Proc Natl Acad Sci U S A 2024; 121:e2408696121. [PMID: 39374400 PMCID: PMC11494314 DOI: 10.1073/pnas.2408696121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 09/03/2024] [Indexed: 10/09/2024] Open
Abstract
A key challenge in molecular biology is to decipher the mapping of protein sequence to function. To perform this mapping requires the identification of sequence features most informative about function. Here, we quantify the amount of information (in bits) that T cell receptor (TCR) sequence features provide about antigen specificity. We identify informative features by their degree of conservation among antigen-specific receptors relative to null expectations. We find that TCR specificity synergistically depends on the hypervariable regions of both receptor chains, with a degree of synergy that strongly depends on the ligand. Using a coincidence-based approach to measuring information enables us to directly bound the accuracy with which TCR specificity can be predicted from partial matches to reference sequences. We anticipate that our statistical framework will be of use for developing machine learning models for TCR specificity prediction and for optimizing TCRs for cell therapies. The proposed coincidence-based information measures might find further applications in bounding the performance of pairwise classifiers in other fields.
Collapse
Affiliation(s)
- James Henderson
- Division of Infection and Immunity, University College London, LondonWC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College London, LondonWC1E 6BT, United Kingdom
| | - Yuta Nagano
- Division of Infection and Immunity, University College London, LondonWC1E 6BT, United Kingdom
- Division of Medicine, University College London, LondonWC1E 6BT, United Kingdom
| | - Martina Milighetti
- Division of Infection and Immunity, University College London, LondonWC1E 6BT, United Kingdom
- Cancer Institute, University College London, LondonWC1E 6DD, United Kingdom
| | - Andreas Tiffeau-Mayer
- Division of Infection and Immunity, University College London, LondonWC1E 6BT, United Kingdom
- Institute for the Physics of Living Systems, University College London, LondonWC1E 6BT, United Kingdom
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Peña-Bates C, Lascurain R, Ortiz-Navarrete V, Chavez-Galan L. The BCG vaccine and SARS-CoV-2: Could there be a beneficial relationship? Heliyon 2024; 10:e38085. [PMID: 39347386 PMCID: PMC11437859 DOI: 10.1016/j.heliyon.2024.e38085] [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: 03/28/2024] [Revised: 09/17/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
The COVID-19 disease continues to cause complications and deaths worldwide. Identifying effective immune protection strategies remains crucial to address this ongoing challenge. The Bacillus Calmette-Guérin (BCG) vaccine, developed initially to prevent pulmonary tuberculosis, has gained relevance due to its ability to induce cross-protection against other pathogens of the airways. This review summarizes research on the immunological protection provided by BCG, along with its primary clinical and therapeutic uses. It also explores the immunological features of COVID-19, the mechanisms implicated in host cell death, and its association with chronic pulmonary illnesses such as tuberculosis, which has led to complications in diagnosis and management. While vaccines against COVID-19 have been administered globally, uncertainty still exists about its effectiveness. Additionally, it is uncertain whether the utilization of BCG can regulate the immune response to pathogens such as SARS-CoV-2.
Collapse
Affiliation(s)
- Carlos Peña-Bates
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Ricardo Lascurain
- Unidad de Enlace Científico, Faculty of Medicine, Universidad Nacional Autónoma de México en el Instituto Nacional de Medicina Genómica, Mexico City, 14610, Mexico
| | - Vianney Ortiz-Navarrete
- Department of Molecular Biomedicine, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional, Mexico City, Mexico
| | - Leslie Chavez-Galan
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| |
Collapse
|
26
|
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
|
27
|
Rowntree LC, Audsley J, Allen LF, McQuilten HA, Hagen RR, Chaurasia P, Petersen J, Littler DR, Tan HX, Murdiyarso L, Habel JR, Foo IJH, Zhang W, Ten Berge ERV, Ganesh H, Kaewpreedee P, Lee KWK, Cheng SMS, Kwok JSY, Jayasinghe D, Gras S, Juno JA, Wheatley AK, Kent SJ, Rossjohn J, Cheng AC, Kotsimbos TC, Trubiano JA, Holmes NE, Pang Chan KK, Hui DSC, Peiris M, Poon LLM, Lewin SR, Doherty PC, Thevarajan I, Valkenburg SA, Kedzierska K, Nguyen THO. SARS-CoV-2-specific CD8 + T cells from people with long COVID establish and maintain effector phenotype and key TCR signatures over 2 years. Proc Natl Acad Sci U S A 2024; 121:e2411428121. [PMID: 39284068 PMCID: PMC11441481 DOI: 10.1073/pnas.2411428121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 07/23/2024] [Indexed: 10/02/2024] Open
Abstract
Long COVID occurs in a small but important minority of patients following COVID-19, reducing quality of life and contributing to healthcare burden. Although research into underlying mechanisms is evolving, immunity is understudied. SARS-CoV-2-specific T cell responses are of key importance for viral clearance and COVID-19 recovery. However, in long COVID, the establishment and persistence of SARS-CoV-2-specific T cells are far from clear, especially beyond 12 mo postinfection and postvaccination. We defined ex vivo antigen-specific B cell and T cell responses and their T cell receptors (TCR) repertoires across 2 y postinfection in people with long COVID. Using 13 SARS-CoV-2 peptide-HLA tetramers, spanning 11 HLA allotypes, as well as spike and nucleocapsid probes, we tracked SARS-CoV-2-specific CD8+ and CD4+ T cells and B-cells in individuals from their first SARS-CoV-2 infection through primary vaccination over 24 mo. The frequencies of ORF1a- and nucleocapsid-specific T cells and B cells remained stable over 24 mo. Spike-specific CD8+ and CD4+ T cells and B cells were boosted by SARS-CoV-2 vaccination, indicating immunization, in fully recovered and people with long COVID, altered the immunodominance hierarchy of SARS-CoV-2 T cell epitopes. Meanwhile, influenza-specific CD8+ T cells were stable across 24 mo, suggesting no bystander-activation. Compared to total T cell populations, SARS-CoV-2-specific T cells were enriched for central memory phenotype, although the proportion of central memory T cells decreased following acute illness. Importantly, TCR repertoire composition was maintained throughout long COVID, including postvaccination, to 2 y postinfection. Overall, we defined ex vivo SARS-CoV-2-specific B cells and T cells to understand primary and recall responses, providing key insights into antigen-specific responses in people with long COVID.
Collapse
Affiliation(s)
- Louise C Rowntree
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jennifer Audsley
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lilith F Allen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hayley A McQuilten
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Ruth R Hagen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Priyanka Chaurasia
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Jan Petersen
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Dene R Littler
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Lydia Murdiyarso
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jennifer R Habel
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Isabelle J H Foo
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Wuji Zhang
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Elizabeth R V Ten Berge
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Hanujah Ganesh
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Prathanporn Kaewpreedee
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Kelly W K Lee
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Samuel M S Cheng
- Division of Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Janette S Y Kwok
- Division of Transplantation and Immunogenetics, Department of Pathology, Queen Mary Hospital, Hong Kong Special Administrative Region, China
| | - Dhilshan Jayasinghe
- Infection & Immunity Program, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3083, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3083, Australia
| | - Stephanie Gras
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Infection & Immunity Program, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3083, Australia
- Department of Biochemistry and Chemistry, School of Agriculture, Biomedicine and Environment, La Trobe University, Bundoora, VIC 3083, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
- Institute of Infection and Immunity, Cardiff University School of Medicine, Cardiff CF14 4XN, United Kingdom
| | - Allen C Cheng
- School of Public Health and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia
- Monash Infectious Diseases, Monash Health and School of Clinical Sciences, Monash University, Clayton, VIC 3168, Australia
| | - Tom C Kotsimbos
- Department of Respiratory Medicine, The Alfred Hospital, Melbourne, VIC 3004, Australia
- Department of Medicine, Central Clinical School, The Alfred Hospital, Monash University, Melbourne, VIC 3004, Australia
| | - Jason A Trubiano
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
- National Centre for Infections in Cancer, Peter McCallum Cancer Centre, Melbourne, VIC 3000, Australia
- Department of Medicine (Austin Health), University of Melbourne, Heidelberg, VIC 3084, Australia
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia
| | - Natasha E Holmes
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, VIC 3084, Australia
- Department of Critical Care, University of Melbourne, Parkville, VIC 3000, Australia
- Data Analytics Research and Evaluation Centre, Austin Health and University of Melbourne, Heidelberg, VIC 3084, Australia
| | - Ken Ka Pang Chan
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - David S C Hui
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Malik Peiris
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Division of Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| | - Leo L M Poon
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Division of Public Health Laboratory Sciences, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
- Centre for Immunology and Infection, Hong Kong Science and Technology Park, New Territories, Hong Kong Special Administrative Region, China
| | - Sharon R Lewin
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Infectious Disease, Alfred Hospital and Monash University, Melbourne, VIC 3000, Australia
| | - Peter C Doherty
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Irani Thevarajan
- Department of Infectious Diseases, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Sophie A Valkenburg
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- HKU-Pasteur Research Pole, School of Public Health, The University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Thi H O Nguyen
- Department of Microbiology and Immunology, University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| |
Collapse
|
28
|
Juhl AK, Dietz LL, Søgaard OS, Reekie J, Nielsen H, Johansen IS, Benfield T, Wiese L, Stærke NB, Jensen TØ, Olesen R, Iversen K, Fogh K, Bodilsen J, Madsen LW, Lindvig SO, Raben D, Andersen SD, Hvidt AK, Andreasen SR, Baerends EAM, Lundgren J, Østergaard L, Tolstrup M. Longitudinal Evaluation of Severe Acute Respiratory Syndrome Coronavirus 2 T-Cell Immunity Over 2 Years Following Vaccination and Infection. J Infect Dis 2024; 230:e605-e615. [PMID: 38687181 PMCID: PMC11420770 DOI: 10.1093/infdis/jiae215] [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: 01/05/2024] [Revised: 04/16/2024] [Accepted: 04/23/2024] [Indexed: 05/02/2024] Open
Abstract
BACKGROUND Within a year of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic, vaccines inducing a robust humoral and cellular immune response were implemented worldwide. However, emergence of novel variants and waning vaccine-induced immunity led to implementation of additional vaccine boosters. METHODS This prospective study evaluated the temporal profile of cellular and serological responses in a cohort of 639 SARS-CoV-2-vaccinated participants, of whom a large proportion experienced a SARS-CoV-2 infection. All participants were infection naïve at the time of their first vaccine dose. Proportions of SARS-CoV-2 spike-specific T cells were determined after each vaccine dose using the activation-induced marker assay, while levels of circulating SARS-CoV-2 antibodies were determined by the Meso Scale serology assay. RESULTS We found a significant increase in SARS-CoV-2 spike-specific CD4+ and CD8+ T-cell responses following the third dose of a SARS-CoV-2 messenger RNA vaccine as well as enhanced CD8+ T-cell responses after the fourth dose. Furthermore, increased age was associated with a poorer response. Finally, we observed that SARS-CoV-2 infection boosts both the cellular and humoral immune response, relative to vaccine-induced immunity alone. CONCLUSIONS Our findings highlight the boosting effect on T-cell immunity of repeated vaccine administration. The combination of multiple vaccine doses and SARS-CoV-2 infections maintains population T-cell immunity, although with reduced levels in the elderly.
Collapse
Affiliation(s)
- Anna Karina Juhl
- Department of Infectious Diseases, Aarhus University Hospital
- Department of Clinical Medicine, Aarhus University, Aarhus
| | - Lisa Loksø Dietz
- Department of Infectious Diseases, Aarhus University Hospital
- Department of Clinical Medicine, Aarhus University, Aarhus
| | - Ole Schmeltz Søgaard
- Department of Infectious Diseases, Aarhus University Hospital
- Department of Clinical Medicine, Aarhus University, Aarhus
| | - Joanne Reekie
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen
| | - Henrik Nielsen
- Department of Infectious Diseases, Aalborg University Hospital
- Department of Clinical Medicine, Aalborg University, Aalborg
| | - Isik Somuncu Johansen
- Department of Infectious Diseases, Odense University Hospital
- Department of Clinical Research, University of Southern Denmark, Odense
| | - Thomas Benfield
- Department of Infectious Diseases, Copenhagen University Hospital–Amager and Hvidovre, Hvidovre
- Department of Clinical Medicine, University of Copenhagen, Copenhagen
| | - Lothar Wiese
- Department of Medicine, Zealand University Hospital, Roskilde
| | - Nina Breinholt Stærke
- Department of Infectious Diseases, Aarhus University Hospital
- Department of Clinical Medicine, Aarhus University, Aarhus
| | - Tomas Østergaard Jensen
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen
| | - Rikke Olesen
- Department of Clinical Medicine, Aarhus University, Aarhus
| | - Kasper Iversen
- Departments of Cardiology and Emergency Medicine, Herlev Hospital, Herlev
| | - Kamille Fogh
- Departments of Cardiology and Emergency Medicine, Herlev Hospital, Herlev
| | - Jacob Bodilsen
- Department of Infectious Diseases, Aalborg University Hospital
- Department of Clinical Medicine, Aalborg University, Aalborg
| | - Lone Wulff Madsen
- Department of Infectious Diseases, Odense University Hospital
- Department of Regional Health Research, University of Southern Denmark, Odense
| | | | - Dorthe Raben
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen
| | | | | | | | | | - Jens Lundgren
- Center of Excellence for Health, Immunity and Infections, Rigshospitalet, University of Copenhagen, Copenhagen
- Department of Clinical Medicine, University of Copenhagen, Copenhagen
- Department of Infectious Diseases, Copenhagen University Hospital–Rigshospitalet, Copenhagen, Denmark
| | - Lars Østergaard
- Department of Infectious Diseases, Aarhus University Hospital
- Department of Clinical Medicine, Aarhus University, Aarhus
| | - Martin Tolstrup
- Department of Infectious Diseases, Aarhus University Hospital
- Department of Clinical Medicine, Aarhus University, Aarhus
| |
Collapse
|
29
|
Smith CL, Didion E, Aung H, Tamilselvan B, Bej T, Oyebanji OA, Shive CL, Wilson BM, Cameron M, Cameron C, Gravenstein S, Canaday DH. Longitudinal Analysis of Nursing Home Residents' T-Cell Responses After SARS-CoV-2 mRNA Vaccinations Shows Influence of Biological Sex and Infection History. J Infect Dis 2024; 230:635-644. [PMID: 38743816 PMCID: PMC11420774 DOI: 10.1093/infdis/jiae234] [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: 12/07/2023] [Revised: 04/02/2024] [Accepted: 04/30/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Vaccines and vaccine boosting have blunted excess morbidity and mortality from severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in older nursing home residents (NHR). However, the impact of repeated vaccination on the T-cell response based on biological sex and prior infection of NHR remain understudied. METHODS We examined T-cell responses to SARS-CoV-2 mRNA vaccines in a cohort of NHR and healthcare workers (HCW) over 2 years. We used interferon-γ ELIspot and flow cytometry to assess T-cell response before, 2 weeks, and 6 months after the initial series and each of 2 booster vaccines. We analyzed these data longitudinally with mixed-effect modeling and also examined subsets of our cohorts for additional changes in T-cell effector function. RESULTS Prior SARS-CoV-2 infection and female sex contributed to higher T-cell response in NHR but not HCW. When looking across time points, NHR but not HCW with prior infection had significantly higher T-cell responses than infection-naive subjects. These patterns of response were maintained across multiple booster vaccinations. CONCLUSIONS These results suggest that the age, multimorbidity, and/or frailty of the NHR cohort may accentuate sex and infection status differences in T-cell response to mRNA vaccination.
Collapse
Affiliation(s)
- Carson L Smith
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Elise Didion
- Division of Infectious Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Htin Aung
- Division of Infectious Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | | | - Taissa Bej
- Geriatric Research, Education, and Clinical Center, Louis Stokes Veterans Affairs Northeast Ohio Healthcare System, Cleveland, Ohio, USA
| | - Oladayo A Oyebanji
- Division of Infectious Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Carey L Shive
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Geriatric Research, Education, and Clinical Center, Louis Stokes Veterans Affairs Northeast Ohio Healthcare System, Cleveland, Ohio, USA
| | - Brigid M Wilson
- Geriatric Research, Education, and Clinical Center, Louis Stokes Veterans Affairs Northeast Ohio Healthcare System, Cleveland, Ohio, USA
- Division of Infectious Diseases and HIV Medicine, Case Western Reserve School of Medicine, Cleveland, Ohio, USA
| | - Mark Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Cheryl Cameron
- Department of Nutrition, Case Western Reserve University, Cleveland, Ohio, USA
| | - Stefan Gravenstein
- Division of Geriatrics and Palliative Medicine, Warren Alpert Medical School, Brown University, Providence, Rhode Island, USA
- Center on Innovation in Long-Term Services and Supports, Providence Veterans Administration Medical Center, Providence, Rhode Island, USA
| | - David H Canaday
- Division of Infectious Disease, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
- Geriatric Research, Education, and Clinical Center, Louis Stokes Veterans Affairs Northeast Ohio Healthcare System, Cleveland, Ohio, USA
| |
Collapse
|
30
|
Wagstaffe HR, Thwaites RS, Reynaldi A, Sidhu JK, McKendry R, Ascough S, Papargyris L, Collins AM, Xu J, Lemm NM, Siggins MK, Chain BM, Killingley B, Kalinova M, Mann A, Catchpole A, Davenport MP, Openshaw PJM, Chiu C. Mucosal and systemic immune correlates of viral control after SARS-CoV-2 infection challenge in seronegative adults. Sci Immunol 2024; 9:eadj9285. [PMID: 38335268 DOI: 10.1126/sciimmunol.adj9285] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 01/12/2024] [Indexed: 02/12/2024]
Abstract
Human infection challenge permits in-depth, early, and pre-symptomatic characterization of the immune response, enabling the identification of factors that are important for viral clearance. Here, we performed intranasal inoculation of 34 young adult, seronegative volunteers with a pre-Alpha severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) strain. Of these participants, 18 (53%) became infected and showed an interferon-dominated mediator response with divergent kinetics between nasal and systemic sites. Peripheral CD4+ and CD8+ T cell activation and proliferation were early and robust but showed distinct kinetic and phenotypic profiles; antigen-specific T cells were largely CD38+Ki67+ and displayed central and effector memory phenotypes. Both mucosal and systemic antibodies became detectable around day 10, but nasal antibodies plateaued after day 14 while circulating antibodies continued to rise. Intensively granular measurements in nasal mucosa and blood allowed modeling of immune responses to primary SARS-CoV-2 infection that revealed CD8+ T cell responses and early mucosal IgA responses strongly associated with viral control, indicating that these mechanisms should be targeted for transmission-reducing intervention.
Collapse
Affiliation(s)
- Helen R Wagstaffe
- Department of Infectious Disease, Imperial College London, London, UK
| | - Ryan S Thwaites
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | - Jasmin K Sidhu
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Richard McKendry
- Department of Infectious Disease, Imperial College London, London, UK
| | - Stephanie Ascough
- Department of Infectious Disease, Imperial College London, London, UK
| | - Loukas Papargyris
- Department of Infectious Disease, Imperial College London, London, UK
| | - Ashley M Collins
- Department of Infectious Disease, Imperial College London, London, UK
| | - Jiayun Xu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Nana-Marie Lemm
- Department of Infectious Disease, Imperial College London, London, UK
| | - Matthew K Siggins
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Benny M Chain
- UCL Division of Infection and Immunity, University College London, London, UK
| | - Ben Killingley
- Department of Infectious Diseases, University College London Hospital, London, UK
| | | | | | | | - Miles P Davenport
- Kirby Institute, University of New South Wales, Kensington, NSW, Australia
| | | | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| |
Collapse
|
31
|
Zheng K, Chong AY, Mentzer AJ. How could our genetics impact COVID-19 vaccine response? Expert Rev Clin Immunol 2024; 20:1027-1039. [PMID: 38676712 DOI: 10.1080/1744666x.2024.2346584] [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: 12/22/2023] [Accepted: 04/19/2024] [Indexed: 04/29/2024]
Abstract
INTRODUCTION The COVID-19 pandemic, caused by the SARS-CoV-2 virus, has posed unprecedented global health challenges since its emergence in December 2019. The rapid availability of vaccines has been estimated to save millions of lives, but there is variation in how individuals respond to vaccines, influencing their effectiveness at an individual, and population level. AREAS COVERED This review focuses on human genetic factors influencing the immune response and effectiveness of vaccines, highlighting the importance of associations across the HLA locus. Genome-Wide Association Studies (GWAS) and other genetic association analyses have identified statistically significant associations between specific HLA alleles including HLA-DRB1*13, DBQ1*06, and A*03 impacting antibody responses and the risk of breakthrough infections post-vaccination. Relationships between these associations and potential mechanisms and links with risks of natural infection or disease are explored, and this review concludes by emphasizing how understanding the mechanisms of these genetic determinants may inform the development of tailored vaccination strategies. EXPERT OPINION Although complex, we believe these findings from the SARS-CoV2 pandemic offer a unique opportunity to understand the relationships between HLA and infection and vaccine response, with a goal of optimizing individual protection against COVID-19 in the ongoing pandemic, and possibly influencing wider vaccine development in the future.
Collapse
Affiliation(s)
- Keyi Zheng
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Amanda Y Chong
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | | |
Collapse
|
32
|
Serdyuk YV, Zornikova KV, Dianov DV, Ivanova NO, Davydova VD, Fefelova EI, Nenasheva TA, Sheetikov SA, Bogolyubova AV. T-Cell Receptors Cross-Reactive to Coronaviral Epitopes Homologous to the SPR Peptide. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1631-1642. [PMID: 39418521 DOI: 10.1134/s0006297924090098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/26/2024] [Accepted: 08/09/2024] [Indexed: 10/19/2024]
Abstract
The COVID-19 pandemic caused by the rapid spread of the novel coronavirus SARS-CoV-2, has promoted an interest in studying the T-cell immune response. It was found that the polyclonal and cross-reactive T-cell response against seasonal coronaviruses and other SARS-CoV-2 strains reduced disease severity. We investigated the immunodominant T-cell epitope SPRWYFYYYL from the nucleocapsid protein of SARS-CoV-2. The immune response to this epitope is characterized by the formation of highly homologous (convergent) receptors that have been found in the T-cell receptor (TCR) repertoires of different individuals. This epitope belongs to a group of highly conserved peptides that are rarely mutated in novel SARS-CoV-2 strains and are homologous to the epitopes of seasonal coronaviruses. It has been suggested that the cross-reactive response to homologous peptides contributes to the reduction of COVID-19 severity. However, some investigators have questioned this hypothesis, suggesting that the low affinity of the cross-reactive receptors reduces the strength of the immune response. The aim of this study was to evaluate the effect of amino acid substitutions in the SPR epitope on its binding affinity to specific TCRs. For this, we performed antigen-dependent cellular expansions were performed using samples from four COVID-19-transfected donors and sequenced their TCR repertoires. The resulting SPR-specific repertoire of β-chains in TCRs had a greater sequence diversity than the repertoire of α-chains. However, the TCR repertoires of all four donors contained public receptors, three of which were cloned and used to generate the Jurkat E6-1 TPR cell line. Only one of these receptors was activated by the SPR peptide and recognized with the same affinity by its mutant homologue LPRWYFYYY from seasonal coronaviruses. This indicates that the presence of the mutation did not affect the strength of the immune response, which may explain why the cross-reactive response to the SPR epitope is so frequent and contributes positively to COVID-19 infection.
Collapse
Affiliation(s)
- Yana V Serdyuk
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Ksenia V Zornikova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Dmitry V Dianov
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Nataliia O Ivanova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Vassa D Davydova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Ekaterina I Fefelova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Tatiana A Nenasheva
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Saveliy A Sheetikov
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Apollinariya V Bogolyubova
- National Medical Research Center for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia.
| |
Collapse
|
33
|
Bodansky A, Mettelman RC, Sabatino JJ, Vazquez SE, Chou J, Novak T, Moffitt KL, Miller HS, Kung AF, Rackaityte E, Zamecnik CR, Rajan JV, Kortbawi H, Mandel-Brehm C, Mitchell A, Wang CY, Saxena A, Zorn K, Yu DJL, Pogorelyy MV, Awad W, Kirk AM, Asaki J, Pluvinage JV, Wilson MR, Zambrano LD, Campbell AP, Thomas PG, Randolph AG, Anderson MS, DeRisi JL. Molecular mimicry in multisystem inflammatory syndrome in children. Nature 2024; 632:622-629. [PMID: 39112696 PMCID: PMC11324515 DOI: 10.1038/s41586-024-07722-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 06/14/2024] [Indexed: 08/16/2024]
Abstract
Multisystem inflammatory syndrome in children (MIS-C) is a severe, post-infectious sequela of SARS-CoV-2 infection1,2, yet the pathophysiological mechanism connecting the infection to the broad inflammatory syndrome remains unknown. Here we leveraged a large set of samples from patients with MIS-C to identify a distinct set of host proteins targeted by patient autoantibodies including a particular autoreactive epitope within SNX8, a protein involved in regulating an antiviral pathway associated with MIS-C pathogenesis. In parallel, we also probed antibody responses from patients with MIS-C to the complete SARS-CoV-2 proteome and found enriched reactivity against a distinct domain of the SARS-CoV-2 nucleocapsid protein. The immunogenic regions of the viral nucleocapsid and host SNX8 proteins bear remarkable sequence similarity. Consequently, we found that many children with anti-SNX8 autoantibodies also have cross-reactive T cells engaging both the SNX8 and the SARS-CoV-2 nucleocapsid protein epitopes. Together, these findings suggest that patients with MIS-C develop a characteristic immune response to the SARS-CoV-2 nucleocapsid protein that is associated with cross-reactivity to the self-protein SNX8, demonstrating a mechanistic link between the infection and the inflammatory syndrome, with implications for better understanding a range of post-infectious autoinflammatory diseases.
Collapse
Affiliation(s)
- Aaron Bodansky
- Department of Pediatrics, Division of Critical Care, University of California San Francisco, San Francisco, CA, USA
| | - Robert C Mettelman
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Joseph J Sabatino
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Sara E Vazquez
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Janet Chou
- Division of Immunology, Department of Pediatrics, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Tanya Novak
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Anesthesia, Harvard Medical School, Boston, MA, USA
| | - Kristin L Moffitt
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Pediatric, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Haleigh S Miller
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Biological and Medical Informatics Program, University of California San Francisco, San Francisco, CA, USA
| | - Andrew F Kung
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Biological and Medical Informatics Program, University of California San Francisco, San Francisco, CA, USA
| | - Elze Rackaityte
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Colin R Zamecnik
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Jayant V Rajan
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - Hannah Kortbawi
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
- Medical Scientist Training Program, University of California San Francisco, San Francisco, CA, USA
| | - Caleigh Mandel-Brehm
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | | | | | - Aditi Saxena
- Chan Zuckerberg Biohub SF, San Francisco, CA, USA
| | - Kelsey Zorn
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA
| | - David J L Yu
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Mikhail V Pogorelyy
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Walid Awad
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Allison M Kirk
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - James Asaki
- Biomedical Sciences Program, University of California San Francisco, San Francisco, CA, USA
| | - John V Pluvinage
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Michael R Wilson
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Laura D Zambrano
- COVID-19 Response Team and Coronavirus and Other Respiratory Viruses Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Angela P Campbell
- COVID-19 Response Team and Coronavirus and Other Respiratory Viruses Division, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Paul G Thomas
- Department of Host-Microbe Interactions, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Adrienne G Randolph
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Anesthesiology, Critical Care and Pain Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Anesthesia, Harvard Medical School, Boston, MA, USA
| | - Mark S Anderson
- Diabetes Center, School of Medicine, University of California San Francisco, San Francisco, CA, USA.
- Department of Medicine, Division of Endocrinology and Metabolism, University of California San Francisco, San Francisco, CA, USA.
| | - Joseph L DeRisi
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, CA, USA.
- Chan Zuckerberg Biohub SF, San Francisco, CA, USA.
| |
Collapse
|
34
|
Zhang J. Immune responses in COVID-19 patients: Insights into cytokine storms and adaptive immunity kinetics. Heliyon 2024; 10:e34577. [PMID: 39149061 PMCID: PMC11325674 DOI: 10.1016/j.heliyon.2024.e34577] [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: 01/29/2024] [Revised: 07/11/2024] [Accepted: 07/11/2024] [Indexed: 08/17/2024] Open
Abstract
SARS-CoV-2 infection can trigger cytokine storm in some patients, which characterized by an excessive production of cytokines and chemical mediators. This hyperactive immune response may cause significant tissue damage and multiple organ failure (MOF). The severity of COVID-19 correlates with the intensity of cytokine storm, involving elements such as IFN, NF-κB, IL-6, HMGB1, etc. It is imperative to rapidly engage adaptive immunity to effectively control the disease progression. CD4+ T cells facilitate an immune response by improving B cells in the production of neutralizing antibodies and activating CD8+ T cells, which are instrumental in eradicating virus-infected cells. Meanwhile, antibodies from B cells can neutralize virus, obstructing further infection of host cells. In individuals who have recovered from the disease, virus-specific antibodies and memory T cells were observed, which could confer a level of protection, reducing the likelihood of re-infection or attenuating severity. This paper discussed the roles of macrophages, IFN, IL-6 and HMGB1 in cytokine release syndrome (CRS), the intricacies of adaptive immunity, and the persistence of immune memory, all of which are critical for the prevention and therapeutic strategies against COVID-19.
Collapse
Affiliation(s)
- Junguo Zhang
- Pulmonology Department, Fengdu General Hospital, Chongqing, 408200, China
| |
Collapse
|
35
|
Kanth SM, Huapaya JA, Gairhe S, Wang H, Tian X, Demirkale CY, Hou C, Ma J, Kuhns DB, Fink DL, Malayeri A, Turkbey E, Harmon SA, Chen MY, Regenold D, Lynch NF, Ramelli S, Li W, Krack J, Kuruppu J, Lionakis MS, Strich JR, Davey R, Childs R, Chertow DS, Kovacs JA, Parizi PT, Suffredini AF. Longitudinal analysis of the lung proteome reveals persistent repair months after mild to moderate COVID-19. Cell Rep Med 2024; 5:101642. [PMID: 38981485 PMCID: PMC11293333 DOI: 10.1016/j.xcrm.2024.101642] [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: 01/23/2024] [Revised: 04/23/2024] [Accepted: 06/13/2024] [Indexed: 07/11/2024]
Abstract
In order to assess homeostatic mechanisms in the lung after COVID-19, changes in the protein signature of bronchoalveolar lavage from 45 patients with mild to moderate disease at three phases (acute, recovery, and convalescent) are evaluated over a year. During the acute phase, inflamed and uninflamed phenotypes are characterized by the expression of tissue repair and host defense response molecules. With recovery, inflammatory and fibrogenic mediators decline and clinical symptoms abate. However, at 9 months, quantified radiographic abnormalities resolve in the majority of patients, and yet compared to healthy persons, all showed ongoing activation of cellular repair processes and depression of the renin-kallikrein-kinin, coagulation, and complement systems. This dissociation of prolonged reparative processes from symptom and radiographic resolution suggests that occult ongoing disruption of the lung proteome is underrecognized and may be relevant to recovery from other serious viral pneumonias.
Collapse
Affiliation(s)
- Shreya M Kanth
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Julio A Huapaya
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Salina Gairhe
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Honghui Wang
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xin Tian
- Office of Biostatistics Research, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Cumhur Y Demirkale
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Chunyan Hou
- Mass Spectrometry and Analytical Pharmacology Shared Resource, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, USA
| | - Junfeng Ma
- Mass Spectrometry and Analytical Pharmacology Shared Resource, Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Georgetown University, Washington, DC 20057, USA
| | - Douglas B Kuhns
- Neutrophil Monitoring Lab, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21701, USA
| | - Danielle L Fink
- Neutrophil Monitoring Lab, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Inc., Frederick, MD 21701, USA
| | - Ashkan Malayeri
- Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD 20892, USA
| | - Evrim Turkbey
- Radiology and Imaging Sciences, Clinical Center (CC), National Institutes of Health, Bethesda, MD 20892, USA
| | - Stephanie A Harmon
- Molecular Imaging Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Marcus Y Chen
- Cardiovascular Branch, National Institute of Heart, Lung, and Blood, National Institutes of Health, Bethesda, MD 20892, USA
| | - David Regenold
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nicolas F Lynch
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sabrina Ramelli
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Willy Li
- Pharmacy Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Janell Krack
- Pharmacy Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Janaki Kuruppu
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michail S Lionakis
- Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Jeffrey R Strich
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard Davey
- Laboratory of Immunoregulation, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Richard Childs
- Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Daniel S Chertow
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA; Laboratory of Virology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joseph A Kovacs
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Parizad Torabi- Parizi
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Anthony F Suffredini
- Critical Care Medicine Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA; Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
36
|
Drost F, An Y, Bonafonte-Pardàs I, Dratva LM, Lindeboom RGH, Haniffa M, Teichmann SA, Theis F, Lotfollahi M, Schubert B. Multi-modal generative modeling for joint analysis of single-cell T cell receptor and gene expression data. Nat Commun 2024; 15:5577. [PMID: 38956082 PMCID: PMC11220149 DOI: 10.1038/s41467-024-49806-9] [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: 10/10/2023] [Accepted: 05/23/2024] [Indexed: 07/04/2024] Open
Abstract
Recent advances in single-cell immune profiling have enabled the simultaneous measurement of transcriptome and T cell receptor (TCR) sequences, offering great potential for studying immune responses at the cellular level. However, integrating these diverse modalities across datasets is challenging due to their unique data characteristics and technical variations. Here, to address this, we develop the multimodal generative model mvTCR to fuse modality-specific information across transcriptome and TCR into a shared representation. Our analysis demonstrates the added value of multimodal over unimodal approaches to capture antigen specificity. Notably, we use mvTCR to distinguish T cell subpopulations binding to SARS-CoV-2 antigens from bystander cells. Furthermore, when combined with reference mapping approaches, mvTCR can map newly generated datasets to extensive T cell references, facilitating knowledge transfer. In summary, we envision mvTCR to enable a scalable analysis of multimodal immune profiling data and advance our understanding of immune responses.
Collapse
Affiliation(s)
- Felix Drost
- Computational Health Center, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354, Freising, Germany
| | - Yang An
- Computational Health Center, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- School of Computation, Information and Technology, Technical University of Munich, Boltzmannstraße 3, 85748, Garching bei München, Germany
| | - Irene Bonafonte-Pardàs
- Computational Health Center, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
| | - Lisa M Dratva
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
| | - Rik G H Lindeboom
- The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Biosciences Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK
- Department of Physics, Cavendish Laboratory, University of Cambridge, 19 JJ Thomson Avenue, Cambridge, UK
| | - Fabian Theis
- Computational Health Center, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany
- School of Life Sciences Weihenstephan, Technical University of Munich, Alte Akademie 8, 85354, Freising, Germany
- School of Computation, Information and Technology, Technical University of Munich, Boltzmannstraße 3, 85748, Garching bei München, Germany
| | - Mohammad Lotfollahi
- Computational Health Center, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, Cambridge, UK.
| | - Benjamin Schubert
- Computational Health Center, Helmholtz Munich, Ingolstädter Landstraße 1, 85764, Neuherberg, Germany.
- School of Computation, Information and Technology, Technical University of Munich, Boltzmannstraße 3, 85748, Garching bei München, Germany.
| |
Collapse
|
37
|
Qui M, Hariharaputran S, Hang SK, Zhang J, Tan CW, Chong CY, Low J, Wang L, Bertoletti A, Yung CF, Le Bert N. T cell hybrid immunity against SARS-CoV-2 in children: a longitudinal study. EBioMedicine 2024; 105:105203. [PMID: 38896919 PMCID: PMC11237860 DOI: 10.1016/j.ebiom.2024.105203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/27/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Hybrid immunity to SARS-CoV-2, resulting from both vaccination and natural infection, remains insufficiently understood in paediatric populations, despite increasing rates of breakthrough infections among vaccinated children. METHODS We conducted a prospective longitudinal study to investigate the magnitude, specificity, and cytokine profile of antigen-specific T cell responses elicited by breakthrough SARS-CoV-2 infection in a cohort of mRNA-vaccinated children (n = 29) aged 5-11. This longitudinal analysis involved six distinct time points spanning a 16-month period post-vaccination, during which we analysed a total of 159 blood samples. All children who were followed for at least 12 months (n = 26) experienced a breakthrough infection. We conducted cytokine release assays using minimal blood samples, and we verified the cellular origin of these responses through intracellular cytokine staining. FINDINGS After breakthrough infection, children who had received mRNA vaccines showed enhanced Th1 responses specific to Spike peptides. Additionally, their Spike-specific T cells exhibited a distinctive enrichment of CD4+ IFN-γ+IL10+ cells, a characteristic akin to adults with hybrid immunity. Importantly, vaccination did not impede the development of multi-specific T cell responses targeting Membrane, Nucleoprotein, and ORF3a/7/8 antigens. INTERPRETATION Children, previously primed with a Spike-based mRNA vaccine and experiencing either symptomatic or asymptomatic breakthrough infection, retained the ability to enhance and diversify Th1/IL-10 antigen-specific T cell responses against multiple SARS-CoV-2 proteins. These findings mirror characteristics associated with hybrid cellular immunity in adults, known to confer resistance against severe COVID-19. FUNDING This study was funded by the National Medical Research Council (NMRC) Singapore (COVID19RF-0019, MOH-000019, MOH-000535, OFLCG19May-0034 and MOH-OFYIRG19nov-0002).
Collapse
Affiliation(s)
- Martin Qui
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | | | - Shou Kit Hang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Jinyan Zhang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Chee Wah Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Infectious Diseases Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Chia Yin Chong
- KK Women's and Children's Hospital, Department of Paediatrics, Infectious Diseases Service, Singapore; Duke-NUS Medical School, Singapore; Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Jenny Low
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Singapore General Hospital, Department of Infectious Diseases, Singapore
| | - Linfa Wang
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Antonio Bertoletti
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore; Singapore Immunology Network, A∗STAR, Singapore
| | - Chee Fu Yung
- KK Women's and Children's Hospital, Department of Paediatrics, Infectious Diseases Service, Singapore; Duke-NUS Medical School, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Nina Le Bert
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore.
| |
Collapse
|
38
|
Tarke A, Ramezani-Rad P, Alves Pereira Neto T, Lee Y, Silva-Moraes V, Goodwin B, Bloom N, Siddiqui L, Avalos L, Frazier A, Zhang Z, da Silva Antunes R, Dan J, Crotty S, Grifoni A, Sette A. SARS-CoV-2 breakthrough infections enhance T cell response magnitude, breadth, and epitope repertoire. Cell Rep Med 2024; 5:101583. [PMID: 38781962 PMCID: PMC11228552 DOI: 10.1016/j.xcrm.2024.101583] [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: 12/12/2023] [Revised: 03/22/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024]
Abstract
Little is known about the effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2 or SARS2) vaccine breakthrough infections (BTIs) on the magnitude and breadth of the T cell repertoire after exposure to different variants. We studied samples from individuals who experienced symptomatic BTIs during Delta or Omicron waves. In the pre-BTI samples, 30% of the donors exhibited substantial immune memory against non-S (spike) SARS2 antigens, consistent with previous undiagnosed asymptomatic SARS2 infections. Following symptomatic BTI, we observed (1) enhanced S-specific CD4 and CD8 T cell responses in donors without previous asymptomatic infection, (2) expansion of CD4 and CD8 T cell responses to non-S targets (M, N, and nsps) independent of SARS2 variant, and (3) generation of novel epitopes recognizing variant-specific mutations. These variant-specific T cell responses accounted for 9%-15% of the total epitope repertoire. Overall, BTIs boost vaccine-induced immune responses by increasing the magnitude and by broadening the repertoire of T cell antigens and epitopes recognized.
Collapse
Affiliation(s)
- Alison Tarke
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Parham Ramezani-Rad
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | | | - Yeji Lee
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Vanessa Silva-Moraes
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL 34987, USA
| | - Benjamin Goodwin
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Nathaniel Bloom
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Leila Siddiqui
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Liliana Avalos
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - April Frazier
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Zeli Zhang
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | | | - Jennifer Dan
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| | - Alba Grifoni
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA.
| | - Alessandro Sette
- Center for Vaccine Innovation, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| |
Collapse
|
39
|
Tiffeau-Mayer A. Unbiased estimation of sampling variance for Simpson's diversity index. Phys Rev E 2024; 109:064411. [PMID: 39020976 DOI: 10.1103/physreve.109.064411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 06/03/2024] [Indexed: 07/20/2024]
Abstract
Quantification of measurement uncertainty is crucial for robust scientific inference, yet accurate estimates of this uncertainty remain elusive for ecological measures of diversity. Here, we address this longstanding challenge by deriving a closed-form unbiased estimator for the sampling variance of Simpson's diversity index. In numerical tests the estimator consistently outperforms existing approaches, particularly for applications in which species richness exceeds sample size. We apply the estimator to quantify biodiversity loss in marine ecosystems and to demonstrate ligand-dependent contributions of T-cell-receptor chains to specificity, illustrating its versatility across fields. The novel estimator provides researchers with a reliable method for comparing diversity between samples, essential for quantifying biodiversity trends and making informed conservation decisions.
Collapse
|
40
|
Aguilar-Bretones M, den Hartog Y, van Dijk LLA, Malahe SRK, Dieterich M, Mora HT, Mueller YM, Koopmans MPG, Reinders MEJ, Baan CC, van Nierop GP, de Vries RD. SARS-CoV-2-specific immune responses converge in kidney disease patients and controls with hybrid immunity. NPJ Vaccines 2024; 9:93. [PMID: 38806532 PMCID: PMC11133345 DOI: 10.1038/s41541-024-00886-0] [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: 10/31/2023] [Accepted: 05/07/2024] [Indexed: 05/30/2024] Open
Abstract
Healthy individuals with hybrid immunity, due to a SARS-CoV-2 infection prior to first vaccination, have stronger immune responses compared to those who were exclusively vaccinated. However, little is known about the characteristics of antibody, B- and T-cell responses in kidney disease patients with hybrid immunity. Here, we explored differences between kidney disease patients and controls with hybrid immunity after asymptomatic or mild coronavirus disease-2019 (COVID-19). We studied the kinetics, magnitude, breadth and phenotype of SARS-CoV-2-specific immune responses against primary mRNA-1273 vaccination in patients with chronic kidney disease or on dialysis, kidney transplant recipients, and controls with hybrid immunity. Although vaccination alone is less immunogenic in kidney disease patients, mRNA-1273 induced a robust immune response in patients with prior SARS-CoV-2 infection. In contrast, kidney disease patients with hybrid immunity develop SARS-CoV-2 antibody, B- and T-cell responses that are equally strong or stronger than controls. Phenotypic analysis showed that Spike (S)-specific B-cells varied between groups in lymph node-homing and memory phenotypes, yet S-specific T-cell responses were phenotypically consistent across groups. The heterogeneity amongst immune responses in hybrid immune kidney patients warrants further studies in larger cohorts to unravel markers of long-term protection that can be used for the design of targeted vaccine regimens.
Collapse
Affiliation(s)
| | - Yvette den Hartog
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Laura L A van Dijk
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - S Reshwan K Malahe
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Marjolein Dieterich
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Héctor Tejeda Mora
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Yvonne M Mueller
- Department of Immunology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marion P G Koopmans
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Marlies E J Reinders
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | - Carla C Baan
- Department of Internal Medicine, Nephrology and Transplantation, Erasmus Medical Center Transplant Institute, Rotterdam, The Netherlands
| | | | - Rory D de Vries
- Department of Viroscience, Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
41
|
Ingels J, De Cock L, Stevens D, Mayer RL, Théry F, Sanchez GS, Vermijlen D, Weening K, De Smet S, Lootens N, Brusseel M, Verstraete T, Buyle J, Van Houtte E, Devreker P, Heyns K, De Munter S, Van Lint S, Goetgeluk G, Bonte S, Billiet L, Pille M, Jansen H, Pascal E, Deseins L, Vantomme L, Verdonckt M, Roelandt R, Eekhout T, Vandamme N, Leclercq G, Taghon T, Kerre T, Vanommeslaeghe F, Dhondt A, Ferdinande L, Van Dorpe J, Desender L, De Ryck F, Vermassen F, Surmont V, Impens F, Menten B, Vermaelen K, Vandekerckhove B. Neoantigen-targeted dendritic cell vaccination in lung cancer patients induces long-lived T cells exhibiting the full differentiation spectrum. Cell Rep Med 2024; 5:101516. [PMID: 38626769 PMCID: PMC11148567 DOI: 10.1016/j.xcrm.2024.101516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 02/09/2024] [Accepted: 03/25/2024] [Indexed: 05/24/2024]
Abstract
Non-small cell lung cancer (NSCLC) is known for high relapse rates despite resection in early stages. Here, we present the results of a phase I clinical trial in which a dendritic cell (DC) vaccine targeting patient-individual neoantigens is evaluated in patients with resected NSCLC. Vaccine manufacturing is feasible in six of 10 enrolled patients. Toxicity is limited to grade 1-2 adverse events. Systemic T cell responses are observed in five out of six vaccinated patients, with T cell responses remaining detectable up to 19 months post vaccination. Single-cell analysis indicates that the responsive T cell population is polyclonal and exhibits the near-entire spectrum of T cell differentiation states, including a naive-like state, but excluding exhausted cell states. Three of six vaccinated patients experience disease recurrence during the follow-up period of 2 years. Collectively, these data support the feasibility, safety, and immunogenicity of this treatment in resected NSCLC.
Collapse
Affiliation(s)
- Joline Ingels
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Laurenz De Cock
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Dieter Stevens
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Rupert L Mayer
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Fabien Théry
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Guillem Sanchez Sanchez
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Institute for Medical Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; WELBIO Department, WEL Research Institute, 1300 Wavre, Walloon Brabant, Belgium
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Institute for Medical Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles, 1050 Brussels, Brussels, Belgium; WELBIO Department, WEL Research Institute, 1300 Wavre, Walloon Brabant, Belgium
| | - Karin Weening
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Saskia De Smet
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Nele Lootens
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Marieke Brusseel
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Tasja Verstraete
- Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Jolien Buyle
- Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Eva Van Houtte
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Pam Devreker
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Kelly Heyns
- GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Stijn De Munter
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Sandra Van Lint
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Glenn Goetgeluk
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Sarah Bonte
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Lore Billiet
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Melissa Pille
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Hanne Jansen
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Eva Pascal
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Lucas Deseins
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium
| | - Lies Vantomme
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Maarten Verdonckt
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Ria Roelandt
- VIB Single Cell Core, VIB, 9000/3000 Ghent/Leuven, East-Flanders/Flemish Brabant, Belgium
| | - Thomas Eekhout
- VIB Single Cell Core, VIB, 9000/3000 Ghent/Leuven, East-Flanders/Flemish Brabant, Belgium
| | - Niels Vandamme
- VIB Single Cell Core, VIB, 9000/3000 Ghent/Leuven, East-Flanders/Flemish Brabant, Belgium
| | - Georges Leclercq
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Tom Taghon
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Tessa Kerre
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium; Hematology, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Floris Vanommeslaeghe
- Nephrology, Ghent University Hospital, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Annemieke Dhondt
- Nephrology, Ghent University Hospital, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Liesbeth Ferdinande
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Pathology, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Jo Van Dorpe
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Pathology, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Liesbeth Desender
- Thoracic and Vascular Surgery, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Frederic De Ryck
- Thoracic and Vascular Surgery, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Frank Vermassen
- Thoracic and Vascular Surgery, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Veerle Surmont
- Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium
| | - Francis Impens
- Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium; VIB-UGent Center for Medical Biotechnology, VIB, 9000 Ghent, East-Flanders, Belgium
| | - Björn Menten
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Department of Biomolecular Medicine, Ghent University, 9000 Ghent, East-Flanders, Belgium
| | - Karim Vermaelen
- Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; Respiratory Medicine, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium.
| | - Bart Vandekerckhove
- Department of Diagnostic Sciences, Ghent University, 9000 Ghent, East-Flanders, Belgium; Cancer Research Institute Ghent (CRIG), 9000 Ghent, Easy-Flanders, Belgium; GMP Unit Cell Therapy, Ghent University Hospital, 9000 Ghent, East-Flanders, Belgium.
| |
Collapse
|
42
|
Leary AY, Scott D, Gupta NT, Waite JC, Skokos D, Atwal GS, Hawkins PG. Designing meaningful continuous representations of T cell receptor sequences with deep generative models. Nat Commun 2024; 15:4271. [PMID: 38769289 PMCID: PMC11106309 DOI: 10.1038/s41467-024-48198-0] [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: 06/18/2023] [Accepted: 04/24/2024] [Indexed: 05/22/2024] Open
Abstract
T Cell Receptor (TCR) antigen binding underlies a key mechanism of the adaptive immune response yet the vast diversity of TCRs and the complexity of protein interactions limits our ability to build useful low dimensional representations of TCRs. To address the current limitations in TCR analysis we develop a capacity-controlled disentangling variational autoencoder trained using a dataset of approximately 100 million TCR sequences, that we name TCR-VALID. We design TCR-VALID such that the model representations are low-dimensional, continuous, disentangled, and sufficiently informative to provide high-quality TCR sequence de novo generation. We thoroughly quantify these properties of the representations, providing a framework for future protein representation learning in low dimensions. The continuity of TCR-VALID representations allows fast and accurate TCR clustering and is benchmarked against other state-of-the-art TCR clustering tools and pre-trained language models.
Collapse
Affiliation(s)
- Allen Y Leary
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA.
| | - Darius Scott
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Namita T Gupta
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Janelle C Waite
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Dimitris Skokos
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Gurinder S Atwal
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA
| | - Peter G Hawkins
- Regeneron Pharmaceuticals Inc., 777 Old Saw Mill River Road, Tarrytown, NY, 10591, USA.
| |
Collapse
|
43
|
Margolis MG, Weizman S, Lazar L, Yakobovich-Gavan M, Tenenbaum A, Phillip M, Oron T. Clinical and immunological characteristics of children diagnosed with-Type 1 diabetes during the COVID-19 pandemic. Diabet Med 2024; 41:e15250. [PMID: 37897235 DOI: 10.1111/dme.15250] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 10/13/2023] [Accepted: 10/20/2023] [Indexed: 10/30/2023]
Abstract
AIMS To find clinical and immunological signatures of the SARS-CoV-2 and the COVID-19 pandemic on children newly diagnosed with type 1 diabetes (T1D). METHODS A single-centre, retrospective, observational study comparing the clinical and immunological characteristics of children diagnosed with T1D the year before and during the first 2 years of the COVID-19 pandemic. Data extracted from the medical records included clinical and demographic parameters, COVID-19 PCR results and the presence of anti-islet, thyroid and celiac-related antibodies. Also obtained from the medical records was a family history of T1D, celiac disease and autoimmune thyroid disease in a first-degree family member. RESULTS A total of 376 children were diagnosed with T1D during the study period. A total of 132 in the pre-COVID era and 246 in the first 2 years of the pandemic. At diagnosis, the pH in children with DKA was lower, and HbA1c tended to be higher in the COVID-19 group compared to the pre-COVID-19 group (7.30 [7.18, 7.35] vs 7.33 [7.19, 7.36], p = 0.046) and (110.9 [86.9, 129.5] vs 100 [80.3, 129.5], p = 0.067]) respectively. Multiple islet antibodies (IA) were significantly more common among patients in the pre-COVID-19 group compared to the COVID-19 group (72% vs 61%, p = 0.032). Tissue transglutaminase antibodies were more common among children diagnosed in the COVID-19 compared to the pre-COVID group (16.6% vs 7.9%, p = 0.022). CONCLUSIONS Our findings suggest that SARS-CoV-2 and the environmental alterations caused by the pandemic affected the clinical characteristics and the immunological profile of children diagnosed with T1D. It is, therefore, plausible that the virus plays a role in the autoimmune process causing T1D.
Collapse
Affiliation(s)
- Merav Gil Margolis
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Sarit Weizman
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Liora Lazar
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Yakobovich-Gavan
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ariel Tenenbaum
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Moshe Phillip
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tal Oron
- The Jesse Z and Sara Lea Shafer Institute for Endocrinology and Diabetes, National Center for Childhood Diabetes, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
44
|
Pavlova AV, Zvyagin IV, Shugay M. Detecting T-cell clonal expansions and quantifying clone survival using deep profiling of immune repertoires. Front Immunol 2024; 15:1321603. [PMID: 38633256 PMCID: PMC11021634 DOI: 10.3389/fimmu.2024.1321603] [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: 10/14/2023] [Accepted: 03/12/2024] [Indexed: 04/19/2024] Open
Abstract
An individual's T-cell repertoire constantly changes under the influence of external and internal factors. Cells that do not receive a stimulatory signal die, while those that encounter and recognize a pathogen or receive a co-stimulatory signal divide, resulting in clonal expansions. T-cell clones can be traced by monitoring the presence of their unique T-cell receptor (TCR) sequence, which is assembled de novo through a process known as V(D)J rearrangement. Tracking T cells can provide valuable insights into the survival of cells after hematopoietic stem cell transplantation (HSCT) or cancer treatment response and can indicate the induction of protective immunity by vaccination. In this study, we report a bioinformatic method for quantifying the T-cell repertoire dynamics from TCR sequencing data. We demonstrate its utility by measuring the T-cell repertoire stability in healthy donors, by quantifying the effect of donor lymphocyte infusion (DLI), and by tracking the fate of the different T-cell subsets in HSCT patients and the expansion of pathogen-specific clones in vaccinated individuals.
Collapse
Affiliation(s)
- Anastasia V. Pavlova
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Ivan V. Zvyagin
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Dmitriy Rogachev National Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Mikhail Shugay
- Institute of Translational Medicine, Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
45
|
Sheetikov SA, Khmelevskaya AA, Zornikova KV, Zvyagin IV, Shomuradova AS, Serdyuk YV, Shakirova NT, Peshkova IO, Titov A, Romaniuk DS, Shagina IA, Chudakov DM, Kiryukhin DO, Shcherbakova OV, Khamaganova EG, Dzutseva V, Afanasiev A, Bogolyubova AV, Efimov GA. Clonal structure and the specificity of vaccine-induced T cell response to SARS-CoV-2 Spike protein. Front Immunol 2024; 15:1369436. [PMID: 38629062 PMCID: PMC11018901 DOI: 10.3389/fimmu.2024.1369436] [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: 01/12/2024] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
Adenovirus vaccines, particularly the COVID-19 Ad5-nCoV adenovirus vaccine, have emerged as promising tools in the fight against infectious diseases. In this study, we investigated the structure of the T cell response to the Spike protein of the SARS-CoV-2 virus used in the COVID-19 Ad5-nCoV adenoviral vaccine in a phase 3 clinical trial (NCT04540419). In 69 participants, we collected peripheral blood samples at four time points after vaccination or placebo injection. Sequencing of T cell receptor repertoires from Spike-stimulated T cell cultures at day 14 from 17 vaccinated revealed a more diverse CD4+ T cell repertoire compared to CD8+. Nevertheless, CD8+ clonotypes accounted for more than half of the Spike-specific repertoire. Our longitudinal analysis showed a peak T cell response at day 14, followed by a decline until month 6. Remarkably, multiple T cell clonotypes persisted for at least 6 months after vaccination, as demonstrated by ex vivo stimulation. Examination of CDR3 regions revealed homologous sequences in both CD4+ and CD8+ clonotypes, with major CD8+ clonotypes sharing high similarity with annotated sequences specific for the NYNYLYRLF peptide, suggesting potential immunodominance. In conclusion, our study demonstrates the immunogenicity of the Ad5-nCoV adenoviral vaccine and highlights its ability to induce robust and durable T cell responses. These findings provide valuable insight into the efficacy of the vaccine against COVID-19 and provide critical information for ongoing efforts to control infectious diseases.
Collapse
Affiliation(s)
- Saveliy A. Sheetikov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Alexandra A. Khmelevskaya
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Ksenia V. Zornikova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Ivan V. Zvyagin
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Alina S. Shomuradova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
- Faculty of Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Yana V. Serdyuk
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Naina T. Shakirova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Iuliia O. Peshkova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Aleksei Titov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Dmitrii S. Romaniuk
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Irina A. Shagina
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
| | - Dmitry M. Chudakov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Genomics of Adaptive Immunity Department, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, Russia
- Central European Institute of Technology, Masaryk University, Brno, Czechia
| | - Dmitry O. Kiryukhin
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Olga V. Shcherbakova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Ekaterina G. Khamaganova
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| | - Vitalina Dzutseva
- Novosibirsk State University, Medical School, Novosibirsk, Russia
- NPO Petrovax Pharm LLC, Moscow, Russia
| | | | | | - Grigory A. Efimov
- Laboratory of Transplantation Immunology, National Medical Research Center for Hematology, Moscow, Russia
| |
Collapse
|
46
|
Galeota E, Bevilacqua V, Gobbini A, Gruarin P, Bombaci M, Pesce E, Favalli A, Lombardi A, Vincenti F, Ongaro J, Fabbris T, Curti S, Martinovic M, Toccafondi M, Lorenzo M, Critelli A, Clemente F, Crosti M, Sarnicola ML, Martinelli M, La Sala L, Espadas A, Donnici L, Borghi MO, De Feo T, De Francesco R, Prati D, Meroni PL, Notarbartolo S, Geginat J, Gori A, Bandera A, Abrignani S, Grifantini R. Tracking the immune response profiles elicited by the BNT162b2 vaccine in COVID-19 unexperienced and experienced individuals. Clin Immunol 2024; 261:110164. [PMID: 38417765 DOI: 10.1016/j.clim.2024.110164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 02/13/2024] [Accepted: 02/23/2024] [Indexed: 03/01/2024]
Abstract
Multiple vaccines have been approved to control COVID-19 pandemic, with Pfizer/BioNTech (BNT162b2) being widely used. We conducted a longitudinal analysis of the immune response elicited after three doses of the BNT162b2 vaccine in individuals who have previously experienced SARS-CoV-2 infection and in unexperienced ones. We conducted immunological analyses and single-cell transcriptomics of circulating T and B lymphocytes, combined to CITE-seq or LIBRA-seq, and VDJ-seq. We found that antibody levels against SARS-CoV-2 Spike, NTD and RBD from wild-type, delta and omicron VoCs show comparable dynamics in both vaccination groups, with a peak after the second dose, a decline after six months and a restoration after the booster dose. The antibody neutralization activity was maintained, with lower titers against the omicron variant. Spike-specific memory B cell response was sustained over the vaccination schedule. Clonal analysis revealed that Spike-specific B cells were polyclonal, with a partial clone conservation from natural infection to vaccination. Spike-specific T cell responses were oriented towards effector and effector memory phenotypes, with similar trends in unexperienced and experienced individuals. The CD8 T cell compartment showed a higher clonal expansion and persistence than CD4 T cells. The first two vaccinations doses tended to induce new clones rather than promoting expansion of pre-existing clones. However, we identified a fraction of Spike-specific CD8 T cell clones persisting from natural infection that were boosted by vaccination and clones specifically induced by vaccination. Collectively, our observations revealed a moderate effect of the second dose in enhancing the immune responses elicited after the first vaccination. Differently, we found that a third dose was necessary to restore comparable levels of neutralizing antibodies and Spike-specific T and B cell responses in individuals who experienced a natural SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Eugenia Galeota
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Valeria Bevilacqua
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Andrea Gobbini
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Paola Gruarin
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Mauro Bombaci
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Elisa Pesce
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Andrea Favalli
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; Ph.D. Program in Translational and Molecular Medicine, Dottorato in Medicina Molecolare e Traslazionale (DIMET), University of Milan-Bicocca, Monza, Italy
| | - Andrea Lombardi
- Infectious Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan 20122, Italy; Centre for Multidisciplinary Research in Health Science (MACH), University of Milano, Milan 20122, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan 20122, Italy
| | - Francesca Vincenti
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Jessica Ongaro
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Tanya Fabbris
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Serena Curti
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Martina Martinovic
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Mirco Toccafondi
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Mariangela Lorenzo
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Angelica Critelli
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Francesca Clemente
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Mariacristina Crosti
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Maria Lucia Sarnicola
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | | | | | - Alejandro Espadas
- Laboratory of Transplant Immunology - North Italy Transplant program (NITp) - Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico of Milan, Italy
| | - Lorena Donnici
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy
| | - Maria Orietta Borghi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy; IRCCS Istituto Auxologico Italiano, Immunorheumatology Research Laboratory, Milan, Italy
| | - Tullia De Feo
- Laboratory of Transplant Immunology - North Italy Transplant program (NITp) - Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico of Milan, Italy
| | - Raffaele De Francesco
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Daniele Prati
- Department of Transfusion Medicine and Hematology, Foundation IRCCS Cà Granda Ospedale Maggiore Policlinico of Milan, Italy
| | - Pier Luigi Meroni
- IRCCS Istituto Auxologico Italiano, Immunorheumatology Research Laboratory, Milan, Italy
| | - Samuele Notarbartolo
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; Infectious Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan 20122, Italy
| | - Jens Geginat
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Andrea Gori
- Centre for Multidisciplinary Research in Health Science (MACH), University of Milano, Milan 20122, Italy; Infectious Diseases Unit, Ospedale "Luigi Sacco", Milan, Italy
| | - Alessandra Bandera
- Infectious Diseases Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan 20122, Italy; Centre for Multidisciplinary Research in Health Science (MACH), University of Milano, Milan 20122, Italy; Department of Pathophysiology and Transplantation, University of Milan, Milan 20122, Italy
| | - Sergio Abrignani
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Renata Grifantini
- INGM, Istituto Nazionale Genetica Molecolare "Romeo ed Enrica Invernizzi", Milan, Italy; CheckmAb Srl, Milan, Italy.
| |
Collapse
|
47
|
van den Dijssel J, Duurland MC, Konijn VA, Kummer LY, Hagen RR, Kuijper LH, Wieske L, van Dam KP, Stalman EW, Steenhuis M, Geerdes DM, Mok JY, Kragten AH, Menage C, Koets L, Veldhuisen B, Verstegen NJ, van der Schoot CE, van Esch WJ, D'Haens GR, Löwenberg M, Volkers AG, Rispens T, Kuijpers TW, Eftimov F, van Gisbergen KP, van Ham SM, Ten Brinke A, van de Sandt CE. mRNA-1273 vaccinated inflammatory bowel disease patients receiving TNF inhibitors develop broad and robust SARS-CoV-2-specific CD8 + T cell responses. J Autoimmun 2024; 144:103175. [PMID: 38387105 DOI: 10.1016/j.jaut.2024.103175] [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: 11/10/2023] [Revised: 01/25/2024] [Accepted: 02/01/2024] [Indexed: 02/24/2024]
Abstract
SARS-CoV-2-specific CD8+ T cells recognize conserved viral peptides and in the absence of cross-reactive antibodies form an important line of protection against emerging viral variants as they ameliorate disease severity. SARS-CoV-2 mRNA vaccines induce robust spike-specific antibody and T cell responses in healthy individuals, but their effectiveness in patients with chronic immune-mediated inflammatory disorders (IMIDs) is less well defined. These patients are often treated with systemic immunosuppressants, which may negatively affect vaccine-induced immunity. Indeed, TNF inhibitor (TNFi)-treated inflammatory bowel disease (IBD) patients display reduced ability to maintain SARS-CoV-2 antibody responses post-vaccination, yet the effects on CD8+ T cells remain unclear. Here, we analyzed the impact of IBD and TNFi treatment on mRNA-1273 vaccine-induced CD8+ T cell responses compared to healthy controls in SARS-CoV-2 experienced and inexperienced patients. CD8+ T cells were analyzed for their ability to recognize 32 SARS-CoV-2-specific epitopes, restricted by 10 common HLA class I allotypes using heterotetramer combinatorial coding. This strategy allowed in-depth ex vivo profiling of the vaccine-induced CD8+ T cell responses using phenotypic and activation markers. mRNA vaccination of TNFi-treated and untreated IBD patients induced robust spike-specific CD8+ T cell responses with a predominant central memory and activated phenotype, comparable to those in healthy controls. Prominent non-spike-specific CD8+ T cell responses were observed in SARS-CoV-2 experienced donors prior to vaccination. Non-spike-specific CD8+ T cells persisted and spike-specific CD8+ T cells notably expanded after vaccination in these patient cohorts. Our data demonstrate that regardless of TNFi treatment or prior SARS-CoV-2 infection, IBD patients benefit from vaccination by inducing a robust spike-specific CD8+ T cell response.
Collapse
Affiliation(s)
- Jet van den Dijssel
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Mariël C Duurland
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Veronique Al Konijn
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Laura Yl Kummer
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Ruth R Hagen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - Lisan H Kuijper
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Luuk Wieske
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands; Department of Clinical Neurophysiology, St Antonius Hospital, Nieuwegein, Netherlands
| | - Koos Pj van Dam
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Eileen W Stalman
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Maurice Steenhuis
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | | | - Juk Yee Mok
- Sanquin Reagents B.V., Amsterdam, Netherlands
| | | | - Charlotte Menage
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Lianne Koets
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; National Screening Laboratory of Sanquin, Research and Laboratory Services, Amsterdam, Netherlands
| | - Barbera Veldhuisen
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Department of Immunohematology Diagnostics, Sanquin Diagnostic Services, Amsterdam, Netherlands
| | - Niels Jm Verstegen
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - C Ellen van der Schoot
- Department of Experimental Immunohematology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | | | - Geert Ram D'Haens
- Department of Gastroenterology and Hepatology, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Mark Löwenberg
- Department of Gastroenterology and Hepatology, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Adriaan G Volkers
- Department of Gastroenterology and Hepatology, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Theo Rispens
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Taco W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Disease, University of Amsterdam, Amsterdam, Netherlands
| | - Filip Eftimov
- Department of Neurology and Neurophysiology, Amsterdam Neuroscience, Amsterdam UMC Location AMC, University of Amsterdam, Amsterdam, Netherlands
| | - Klaas Pjm van Gisbergen
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands
| | - S Marieke van Ham
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Swammerdam Institute for Life Sciences, University of Amsterdam, Netherlands
| | - Anja Ten Brinke
- Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands; Department of Immunopathology, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands
| | - Carolien E van de Sandt
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam, Netherlands; Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia.
| |
Collapse
|
48
|
Xie Y, Chen H, Chellamuthu VR, Lajam ABM, Albani S, Low AHL, Petretto E, Behmoaras J. Comparative Analysis of Single-Cell RNA Sequencing Methods with and without Sample Multiplexing. Int J Mol Sci 2024; 25:3828. [PMID: 38612639 PMCID: PMC11011421 DOI: 10.3390/ijms25073828] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/20/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Single-cell RNA sequencing (scRNA-seq) has emerged as a powerful technique for investigating biological heterogeneity at the single-cell level in human systems and model organisms. Recent advances in scRNA-seq have enabled the pooling of cells from multiple samples into single libraries, thereby increasing sample throughput while reducing technical batch effects, library preparation time, and the overall cost. However, a comparative analysis of scRNA-seq methods with and without sample multiplexing is lacking. In this study, we benchmarked methods from two representative platforms: Parse Biosciences (Parse; with sample multiplexing) and 10x Genomics (10x; without sample multiplexing). By using peripheral blood mononuclear cells (PBMCs) obtained from two healthy individuals, we demonstrate that demultiplexed scRNA-seq data obtained from Parse showed similar cell type frequencies compared to 10x data where samples were not multiplexed. Despite relatively lower cell capture affecting library preparation, Parse can detect rare cell types (e.g., plasmablasts and dendritic cells) which is likely due to its relatively higher sensitivity in gene detection. Moreover, a comparative analysis of transcript quantification between the two platforms revealed platform-specific distributions of gene length and GC content. These results offer guidance for researchers in designing high-throughput scRNA-seq studies.
Collapse
Affiliation(s)
- Yi Xie
- Programme in Cardiovascular and Metabolic Disorders and Centre for Computational Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore; (Y.X.)
| | - Huimei Chen
- Programme in Cardiovascular and Metabolic Disorders and Centre for Computational Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore; (Y.X.)
| | - Vasuki Ranjani Chellamuthu
- Translational Immunology Institute, SingHealth/Duke-NUS Academic Medical Centre, Academia, Singapore 169856, Singapore; (V.R.C.)
| | - Ahmad bin Mohamed Lajam
- Translational Immunology Institute, SingHealth/Duke-NUS Academic Medical Centre, Academia, Singapore 169856, Singapore; (V.R.C.)
| | - Salvatore Albani
- Translational Immunology Institute, SingHealth/Duke-NUS Academic Medical Centre, Academia, Singapore 169856, Singapore; (V.R.C.)
| | - Andrea Hsiu Ling Low
- Department of Rheumatology and Immunology, Singapore General Hospital, Academia, Singapore 169856, Singapore;
- SingHealth Duke-NUS Medicine Academic Clinical Programme, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Enrico Petretto
- Programme in Cardiovascular and Metabolic Disorders and Centre for Computational Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore; (Y.X.)
- Institute for Big Data and Artificial Intelligence in Medicine, School of Science, China Pharmaceutical University, Nanjing 210009, China
| | - Jacques Behmoaras
- Programme in Cardiovascular and Metabolic Disorders and Centre for Computational Biology, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore; (Y.X.)
- Department of Immunology and Inflammation, Centre for Inflammatory Disease, Imperial College London, Hammersmith Hospital, London W12 0NN, UK
| |
Collapse
|
49
|
Aoki H, Kitabatake M, Abe H, Xu P, Tsunoda M, Shichino S, Hara A, Ouji-Sageshima N, Motozono C, Ito T, Matsushima K, Ueha S. CD8 + T cell memory induced by successive SARS-CoV-2 mRNA vaccinations is characterized by shifts in clonal dominance. Cell Rep 2024; 43:113887. [PMID: 38458195 DOI: 10.1016/j.celrep.2024.113887] [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: 07/20/2023] [Revised: 12/27/2023] [Accepted: 02/14/2024] [Indexed: 03/10/2024] Open
Abstract
mRNA vaccines against the spike glycoprotein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) elicit strong T cell responses. However, a clonal-resolution analysis of T cell responses to mRNA vaccination has not been performed. Here, we temporally track the CD8+ T cell repertoire in individuals who received three shots of the BNT162b2 mRNA vaccine through longitudinal T cell receptor sequencing with peptide-human leukocyte antigen (HLA) tetramer analysis. We demonstrate a shift in T cell responses between the clonotypes with different kinetics: from early responders that expand rapidly after the first shot to main responders that greatly expand after the second shot. Although the main responders re-expand after the third shot, their clonal diversity is skewed, and newly elicited third responders partially replace them. Furthermore, this shift in clonal dominance occurs not only between, but also within, clonotypes specific for spike epitopes. Our study will be a valuable resource for understanding vaccine-induced T cell responses in general.
Collapse
Affiliation(s)
- Hiroyasu Aoki
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda City, Chiba 2780022, Japan; Department of Preventive Medicine, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Masahiro Kitabatake
- Department of Immunology, Nara Medical University, Kashihara City, Nara 6348521, Japan
| | - Haruka Abe
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda City, Chiba 2780022, Japan
| | - Peng Xu
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda City, Chiba 2780022, Japan
| | - Mikiya Tsunoda
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda City, Chiba 2780022, Japan
| | - Shigeyuki Shichino
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda City, Chiba 2780022, Japan
| | - Atsushi Hara
- Department of Immunology, Nara Medical University, Kashihara City, Nara 6348521, Japan
| | - Noriko Ouji-Sageshima
- Department of Immunology, Nara Medical University, Kashihara City, Nara 6348521, Japan
| | - Chihiro Motozono
- Division of Infection and Immunity, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto City, Kumamoto 8600811, Japan
| | - Toshihiro Ito
- Department of Immunology, Nara Medical University, Kashihara City, Nara 6348521, Japan
| | - Kouji Matsushima
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda City, Chiba 2780022, Japan
| | - Satoshi Ueha
- Division of Molecular Regulation of Inflammatory and Immune Diseases, Research Institute for Biomedical Sciences, Tokyo University of Science, Noda City, Chiba 2780022, Japan.
| |
Collapse
|
50
|
Xing M, Hu G, Wang X, Wang Y, He F, Dai W, Wang X, Niu Y, Liu J, Liu H, Zhang X, Xu J, Cai Q, Zhou D. An intranasal combination vaccine induces systemic and mucosal immunity against COVID-19 and influenza. NPJ Vaccines 2024; 9:64. [PMID: 38509167 PMCID: PMC10954707 DOI: 10.1038/s41541-024-00857-5] [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: 10/19/2023] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Despite prolonged surveillance and interventions, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza viruses continue to pose a severe global health burden. Thus, we developed a chimpanzee adenovirus-based combination vaccine, AdC68-HATRBD, with dual specificity against SARS-CoV-2 and influenza virus. When used as a standalone vaccine, intranasal immunization with AdC68-HATRBD induced comprehensive and potent immune responses consisting of immunoglobin (Ig) G, mucosal IgA, neutralizing antibodies, and memory T cells, which protected the mice from BA.5.2 and pandemic H1N1 infections. When used as a heterologous booster, AdC68-HATRBD markedly improved the protective immune response of the licensed SARS-CoV-2 or influenza vaccine. Therefore, whether administered intranasally as a standalone or booster vaccine, this combination vaccine is a valuable strategy to enhance the overall vaccine efficacy by inducing robust systemic and mucosal immune responses, thereby conferring dual lines of immunological defenses for these two viruses.
Collapse
Affiliation(s)
- Man Xing
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Gaowei Hu
- MOE&NHC&CAMS Key Laboratory of Medical Molecular, Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Xiang Wang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Yihan Wang
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Furong He
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Weiqian Dai
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Xinyu Wang
- MOE&NHC&CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yixin Niu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Jiaojiao Liu
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China
| | - Hui Liu
- Chengdu Kanghua Biological Products Co., Ltd, Chengdu, China
| | - Xiaoyan Zhang
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China
| | - Jianqing Xu
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.
| | - Qiliang Cai
- MOE&NHC&CAMS Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infections Disease and Biosecurity, Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Dongming Zhou
- Department of Pathogen Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, 300070, China.
- Shanghai Public Health Clinical Center, Fudan University, Shanghai, 201508, China.
| |
Collapse
|