1
|
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
|
2
|
Lai W, Li Y, Luo OJ. MIST: An interpretable and flexible deep learning framework for single-T cell transcriptome and receptor analysis. SCIENCE ADVANCES 2025; 11:eadr7134. [PMID: 40184452 PMCID: PMC11970455 DOI: 10.1126/sciadv.adr7134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Accepted: 02/28/2025] [Indexed: 04/06/2025]
Abstract
Joint analysis of transcriptomic and T cell receptor (TCR) features at single-cell resolution provides a powerful approach for in-depth T cell immune function research. Here, we introduce a deep learning framework for single-T cell transcriptome and receptor analysis, MIST (Multi-insight for T cell). MIST features three latent spaces: gene expression, TCR, and a joint latent space. Through analyses of antigen-specific T cells, and T cell datasets related to lung cancer immunotherapy and COVID19, we demonstrate MIST's interpretability and flexibility. MIST easily and accurately resolves cell function and antigen specificity by vectorizing and integrating transcriptome and TCR data of T cells. In addition, using MIST, we identified the heterogeneity of CXCL13+ subsets in lung cancer infiltrating CD8+ T cells and their association with immunotherapy, providing additional insights into the functional transition of CXCL13+ T cells related to anti-PD-1 therapy that were not reported in the original study.
Collapse
Affiliation(s)
- Wenpu Lai
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Yangqiu Li
- The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou 510632, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou 510632, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou 510632, China
- Zhuhai Institute of Jinan University, Jinan University, Zhuhai 519070, China
| |
Collapse
|
3
|
Xie S, Wei J, Wang X. The intersection of influenza infection and autoimmunity. Front Immunol 2025; 16:1558386. [PMID: 40248710 PMCID: PMC12003283 DOI: 10.3389/fimmu.2025.1558386] [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: 01/10/2025] [Accepted: 03/17/2025] [Indexed: 04/19/2025] Open
Abstract
The relationship between viral infection and autoimmune manifestations has been emerging as a significant focus of study, underscoring the intricate interplay between viral infections and the immune system. Influenza infection can result in a spectrum of clinical outcomes, ranging from mild illness to severe disease, including mortality. Annual influenza vaccination remains the most effective strategy for preventing infection and its associated complications. The complications arising from acute influenza infection are attributable not only to the direct effects of the viral infection but also to the dysregulated immune response it elicits. Notably, associations between influenza and various autoimmune diseases, such as Guillain-Barré Syndrome (GBS), Type 1 Diabetes (T1D), and antiphospholipid syndrome, have been reported. While viral infections have long been recognized as potential triggers of autoimmunity, the underlying mechanisms remain to be elucidated. Here, we described the pathophysiology caused by influenza infection and the influenza-associated autoimmune manifestations. Current advances on the understanding of the underlying immune mechanisms that lead to the potential strategies were also summarized.
Collapse
Affiliation(s)
| | | | - Xiaohui Wang
- Guangzhou Institute of Paediatrics, Guangzhou Women and Children’s Medical Center, Guangdong Provincial Research Center for Child Health, State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
4
|
Dörnte C, Datsi A, Traska V, Kostyra J, Wagner M, Brauns O, Lamsfuß C, Winkels H, Balz V, Enczmann J, Adams O, Mueller L, Baurmann H, Eiz‐Vesper B, Bonifacius A, Sorg RV, Dose C, Schmitz J, Richter A, Fischer J, Schuster M. Distinct HLA Haplotypes Are Associated With an Altered Strength of SARS-CoV-2-Specific T-Cell Responses and Unfavorable Disease Courses. Eur J Immunol 2025; 55:e202451497. [PMID: 40259456 PMCID: PMC12012228 DOI: 10.1002/eji.202451497] [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: 09/03/2024] [Revised: 03/04/2025] [Accepted: 03/13/2025] [Indexed: 04/23/2025]
Abstract
Infection with SARS-CoV-2 results in mild to severe COVID-19 disease courses. Several studies showed the association of impaired T-cell responses and certain HLA haplotypes with disease severity. However, it remained unclear if T-cell activation was compromised due to a general reduction of presented epitopes or other intrinsic factors within APCs or T cells. Furthermore, a potential reduction of presented epitopes would suggest if an upcoming SARS-CoV-2 variant could escape T-cell immunity. Hence, knowledge about the T-cell epitope landscape of SARS-CoV-2 would allow to better understand mechanisms leading to severe disease and to estimate the potential stability of the T-cell response in light of virus evolution, which might provide insights for future vaccine designs. Hence, in the present study, the T-cell epitope landscape of SARS-CoV-2 was determined via in vitro T-cell stimulation plus in silico prediction. HLAs associated with mild and severe disease courses showed almost the same potential in epitope presentation, suggesting intrinsic factors of APCs or T cells as contributors to the more severe disease courses. As T-cell epitopes did also not originate from regions of SARS-CoV-2 having shown high mutation rates in the past, a relatively stable T-cell response can be expected regarding new SARS-CoV-2 strains in the future. Analysis of the T-cell epitope landscape of SARS-CoV-2 suggests T-cell intrinsic factors as likely modulators of disease severity and that the capacity of MHC-peptide presentation remains stable among circulating SARS-CoV-2 viral strains.
Collapse
Affiliation(s)
- C. Dörnte
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
- Clinic III for Internal MedicineUniversity of Cologne Faculty of Medicine and University Hospital CologneCologneGermany
| | - A. Datsi
- Institute for Transplantation Diagnostics and Cell TherapeuticsMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐University DüsseldorfDüsseldorfGermany
| | - V. Traska
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - J. Kostyra
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - M. Wagner
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - O. Brauns
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - C. Lamsfuß
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - H. Winkels
- Clinic III for Internal MedicineUniversity of Cologne Faculty of Medicine and University Hospital CologneCologneGermany
- Center for Molecular Medicine Cologne (CMMC)University of CologneCologneGermany
| | - V. Balz
- Institute for Transplantation Diagnostics and Cell TherapeuticsMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐University DüsseldorfDüsseldorfGermany
| | - J. Enczmann
- Institute for Transplantation Diagnostics and Cell TherapeuticsMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐University DüsseldorfDüsseldorfGermany
| | - O. Adams
- Institute for VirologyMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐UniversityDüsseldorfGermany
| | - L. Mueller
- Institute for VirologyMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐UniversityDüsseldorfGermany
| | - H. Baurmann
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - B. Eiz‐Vesper
- Institute of Transfusion Medicine and Transplant EngineeringMedizinische Hochschule HannoverHannoverGermany
| | - A. Bonifacius
- Institute of Transfusion Medicine and Transplant EngineeringMedizinische Hochschule HannoverHannoverGermany
| | - R. V. Sorg
- Institute for Transplantation Diagnostics and Cell TherapeuticsMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐University DüsseldorfDüsseldorfGermany
| | - C. Dose
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - J. Schmitz
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - A. Richter
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| | - J. Fischer
- Institute for Transplantation Diagnostics and Cell TherapeuticsMedical Faculty and University Hospital Düsseldorf Heinrich Heine‐University DüsseldorfDüsseldorfGermany
| | - M. Schuster
- Miltenyi Biotec B.V. & Co. KGBergisch GladbachGermany
| |
Collapse
|
5
|
Pujadas V, Chin C, Sankpal NV, Buhrmaster J, Arjuna A, Walia R, Smith MA, Eickelberg O, Bremner RM, Mohanakumar T, Sureshbabu A. Alveolar epithelial type 2 cell specific loss of IGFBP2 activates inflammation in COVID-19. Respir Res 2025; 26:111. [PMID: 40121473 PMCID: PMC11929192 DOI: 10.1186/s12931-025-03187-9] [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: 09/20/2024] [Accepted: 03/10/2025] [Indexed: 03/25/2025] Open
Abstract
The coronavirus disease 2019 (COVID-19) global pandemic is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). However, our understanding of SARS-CoV-2-induced inflammation in alveolar epithelial cells remains very limited. The contributions of intracellular insulin-like growth factor binding protein-2 (IGFBP2) to SARS-CoV-2 pathogenesis are also unclear. In this study, we have uncovered a critical role for IGFBP2, specifically in alveolar epithelial type 2 cells (AEC2), in the immunopathogenesis of COVID-19. Using bulk RNA sequencing, we show that IGFBP2 mRNA expression is significantly downregulated in primary AEC2 cells isolated from fibrotic lung regions from patients with COVID-19-acute respiratory distress syndrome (ARDS) compared to those with idiopathic pulmonary fibrosis (IPF) alone or IPF with a history of COVID-19. Using multicolor immunohistochemistry, we demonstrated that IGFBP2 and its selective ligands IGF1 and IGF2 were significantly reduced in AEC2 cells from patients with COVID-ARDS, IPF alone, or IPF with COVID history than in those from age-matched donor controls. Further, we demonstrated that lentiviral expression of Igfbp2 significantly reduced mRNA expression of proinflammatory cytokines-Tnf-α, Il1β, Il6, Stat3, Stat6 and chemokine receptors-Ccr2 and Ccr5-in mouse lung epithelial cells challenged with SARS-CoV-2 spike protein injury (S2; 500 ng/mL). Finally, we demonstrated higher levels of cytokines-TNF-α; IL-6 and chemokine receptor-CCR5 in AEC2 cells from COVID-ARDS patients compared to the IPF alone and the IPF with COVID history patients. Altogether, these data suggest that anti-inflammatory properties of IGFBP2 in AEC2 cells and its localized delivery may serve as potential therapeutic strategy for patients with COVID-19.
Collapse
Affiliation(s)
- Valentina Pujadas
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Chiahsuan Chin
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Narendra V Sankpal
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - James Buhrmaster
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Ashwini Arjuna
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Rajat Walia
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Michael A Smith
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Oliver Eickelberg
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Ross M Bremner
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Thalachallour Mohanakumar
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA
| | - Angara Sureshbabu
- Norton Thoracic Institute, St. Joseph's Hospital and Medical Center, 124 W. Thomas Road, Ste. 100, Phoenix, AZ, 85013, USA.
- Creighton University School of Medicine - Phoenix Regional Campus, Phoenix, AZ, USA.
| |
Collapse
|
6
|
Hao T, Ryan GE, Lydeamore MJ, Cromer D, Wood JG, McVernon J, McCaw JM, Shearer FM, Golding N. Predicting immune protection against outcomes of infectious disease from population-level effectiveness data with application to COVID-19. Vaccine 2025; 55:126987. [PMID: 40117726 DOI: 10.1016/j.vaccine.2025.126987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/20/2025] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
Quantifying the extent to which previous infections and vaccinations confer protection against future infection or disease outcomes is critical to managing the transmission and consequences of infectious diseases. We present a general statistical model for predicting the strength of protection conferred by different immunising exposures (numbers, types, and strains of both vaccines and infections), against multiple outcomes of interest, whilst accounting for immune waning. We predict immune protection against key clinical outcomes: developing symptoms, hospitalisation, and death. We also predict transmission-related outcomes: acquisition of infection and onward transmission in breakthrough infections. These enable quantification of the impact of immunity on population-level transmission dynamics. Our model calibrates the level of immune protection, drawing on both population-level data, such as vaccine effectiveness estimates, and neutralising antibody levels as a correlate of protection. This enables the model to learn realised immunity levels beyond those which can be predicted by antibody kinetics or other correlates alone. We demonstrate an application of the model for SARS-CoV-2, and predict the individual-level protective effectiveness conferred by natural infections with the Delta and the Omicron B.1.1.529 variants, and by the BioNTech-Pfizer (BNT162b2), Oxford-AstraZeneca (ChAdOx1), and 3rd-dose mRNA booster vaccines, against outcomes for both Delta and Omicron. We also demonstrate a use case of the model in late 2021 during the emergence of Omicron, showing how the model can be rapidly updated with emerging epidemiological data on multiple variants in the same population, to infer key immunogenicity and intrinsic transmissibility characteristics of the new variant, before the former can be more directly observed via vaccine effectiveness data. This model provided timely inference on rapidly evolving epidemic situations of significant concern during the early stages of the COVID-19 pandemic. The general nature of the model enables it to be used to support management of a range of infectious diseases.
Collapse
Affiliation(s)
- Tianxiao Hao
- The Kids Research Institute, Nedlands, Western Australia, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia.
| | - Gerard E Ryan
- The Kids Research Institute, Nedlands, Western Australia, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
| | - Michael J Lydeamore
- Department of Econometrics and Business Statistics, Monash University, Victoria, Australia
| | - Deborah Cromer
- Kirby Institute, University of New South Wales Sydney, New South Wales, Australia
| | - James G Wood
- School of Population Health, University of New South Wales Sydney, New South Wales, Australia
| | - Jodie McVernon
- The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Victoria, Australia; Victorian Infectious Disease Reference Laboratory Epidemiology Unit, The Royal Melbourne Hospital, Victoria, Australia
| | - James M McCaw
- Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia; School of Mathematics and Statistics, The University of Melbourne, Victoria, Australia
| | - Freya M Shearer
- The Kids Research Institute, Nedlands, Western Australia, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia
| | - Nick Golding
- The Kids Research Institute, Nedlands, Western Australia, Australia; Melbourne School of Population and Global Health, The University of Melbourne, Victoria, Australia; School of Population Health, Curtin University, Western Australia, Australia
| |
Collapse
|
7
|
Alrubayyi A, Huang H, Gaiha GD. Severe Acute Respiratory Syndrome Coronavirus 2 Immunology and Coronavirus Disease 2019 Clinical Outcomes. Infect Dis Clin North Am 2025:S0891-5520(25)00006-6. [PMID: 40089444 DOI: 10.1016/j.idc.2025.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
Abstract
The humoral and cellular immune response are the key players in preventing viral infection and limiting disease severity, particular in the context of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and coronavirus disease 2019. In this review, we discuss how immune responses generated by prior infection and vaccination influence the outcomes of SARS-CoV-2 infection. We aim to provide an overview of the role of humoral and cellular immunity, with a particular focus on CD8+ T cell responses, to delineate how different immune compartments contribute to the control of infection and modulation of disease outcomes.
Collapse
Affiliation(s)
| | - Hsinyen Huang
- Ragon Institute of MGB, MIT and Harvard, 600 Main Street, Cambridge, MA 02139
| | - Gaurav D Gaiha
- Ragon Institute of MGB, MIT and Harvard, 600 Main Street, Cambridge, MA 02139; Division of Gastroenterology, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02115.
| |
Collapse
|
8
|
Ying H, Wu X, Jia X, Yang Q, Liu H, Zhao H, Chen Z, Xu M, Wang T, Li M, Zhao Z, Zheng R, Wang S, Lin H, Xu Y, Lu J, Wang W, Ning G, Zheng J, Bi Y. Single-cell transcriptome-wide Mendelian randomization and colocalization reveals immune-mediated regulatory mechanisms and drug targets for COVID-19. EBioMedicine 2025; 113:105596. [PMID: 39933264 PMCID: PMC11867302 DOI: 10.1016/j.ebiom.2025.105596] [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: 08/20/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND COVID-19 continues to show long-term impacts on our health. Limited effective immune-mediated antiviral drugs have been launched. METHODS We conducted a Mendelian randomization (MR) and colocalization analysis using 26,597 single-cell expression quantitative trait loci (sc-eQTL) to proxy effects of expressions of 16,597 genes in 14 peripheral blood immune cells and tested them against four COVID-19 outcomes from COVID-19 Genetic Housing Initiative GWAS meta-analysis Round 7. We also carried out additional validations including colocalization, linkage disequilibrium check and host-pathogen interactome predictions. We integrated MR findings with clinical trial evidence from several drug gene related databases to identify drugs with repurposing potential. Finally, we developed a tier system and identified immune-cell-based prioritized drug targets for COVID-19. FINDINGS We identified 132 putative causal genes in 14 immune cells (343 MR associations) for COVID-19, with 58 genes that were not reported previously. 145 (73%) gene-COVID-19 pairs showed effects on COVID-19 in only one immune cell type, which implied widespread immune-cell specific effects. For pathway analyses, we found the putative causal genes were enriched in natural killer (NK) recruiting cells but de-enriched in NK cells. Using a deep learning model, we found 107 (81%) of the putative causal genes (41 novel genes) were predicted to interact with SARS-COV-2 proteins. Integrating the above evidence with drug trial information, we developed a tier system and prioritized 37 drug targets for COVID-19. INTERPRETATION Our study showcased the central role of immune-mediated regulatory mechanisms for COVID-19 and prioritized drug targets that might inform interventions for viral infectious diseases. FUNDING This work was supported by grants from the National Key Research and Development Program of China (2022YFC2505203).
Collapse
Affiliation(s)
- Hui Ying
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojing Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianqian Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyu Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiling Zhao
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Zhihe Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
9
|
Zhang Y, Li X, Yang Y, Yin Y, Zhong Y, Xu Q, Tu J, Deng J, Liang H, Shen T. Impact of SARS-CoV-2 inactivated vaccine on symptoms following omicron variant breakthrough infection. Vaccine 2025; 48:126722. [PMID: 39813973 DOI: 10.1016/j.vaccine.2025.126722] [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: 08/15/2024] [Revised: 12/02/2024] [Accepted: 01/07/2025] [Indexed: 01/18/2025]
Abstract
The SARS-CoV-2 Omicron variant and its sublineages continue to circulate widely. Clinical outcomes with this variant differ among individuals, primarily influenced by host immunity. Previous studies have explored the relationship between immune responses and severe diseases in infected or convalescent patients. However, the impact of vaccine-induced immune responses on disease severity, especially in cases of mild infection following breakthrough infection, remains unclear. This is primarily due to the lack of assessment of immune status in vaccinated individuals before infection. In this study, we aimed to elucidate the causality between virus-specific cellular and humoral immune responses and the severity of symptoms in breakthrough infected patients from a long-term follow-up post-vaccination cohort. A questionnaire survey was conducted to collect general symptoms upon breakthrough infection with the Omicron variants. Plasma levels of specific antibodies (neutralizing antibodies, anti-S IgG, and anti-N IgG) and T cell responses induced by inactivated SARS-CoV-2 vaccine were evaluated. The findings revealed that individuals with milder symptoms, particularly lower peak fever temperatures, exhibited higher antibody levels and enhanced T cell activation and responses prior to infection. This suggests that cellular and humoral immunity induced by inactivated vaccines may provide protection against severe clinical symptoms following breakthrough infection.
Collapse
Affiliation(s)
- Yuqi Zhang
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing 100191, China..
| | - Xinjie Li
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing 100191, China..
| | - Yingxiang Yang
- Senior Department of Hepato-Pancreato-Biliary Surgery, The First Medical Center of PLA General Hospital, Beijing 100853, China.
| | - Yue Yin
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing 100191, China..
| | - Yan Zhong
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| | - Qiang Xu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing 100191, China..
| | - Jing Tu
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing 100191, China..
| | - Juan Deng
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing 100191, China..
| | - Hua Liang
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Center for AIDS/STD Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China.
| | - Tao Shen
- Department of Microbiology and Infectious Disease Center, School of Basic Medical Sciences, Peking University, Beijing 100191, China..
| |
Collapse
|
10
|
Hurme A, Viinanen A, Teräsjärvi J, Jalkanen P, Feuth T, Löyttyniemi E, Vuorinen T, Kantele A, Oksi J, He Q, Julkunen I. Post-COVID-19 condition in prospective inpatient and outpatient cohorts. Sci Rep 2025; 15:6925. [PMID: 40011519 DOI: 10.1038/s41598-025-90819-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 02/17/2025] [Indexed: 02/28/2025] Open
Abstract
Viral persistence, immune dysregulation, hypocortisolism, and pulmonary tissue damage from acute infection are proposed as pathogenic mechanisms underlying post-COVID-19 condition (PCC). In this prospective observational study, we followed 62 COVID-19 inpatients and 53 COVID-19 outpatients for 24 months after the infection. During this period, we assessed prolonged symptoms, lung function, and a set of immunological markers and a proportion of the patient group was assessed with computed tomography three months post-infection. The prevalence of PCC, as assessed by four medical specialists, decreased from 51% at three months to 18% at 24 months. Risk factors included the severity of the acute infection and comorbidities of obstructive sleep apnea or obesity. Patients with PCC had higher serum levels of anti-SARS-CoV-2 S1 and N protein antibodies. In the whole group, spirometry results, orthostatic hypotension, or levels of soluble suppression of tumorigenicity 2, interleukin 6 (IL-6), high-sensitivity C-reactive protein (hs-CRP), or cortisol had no association with PCC. However, using symptom clusters, patients with cognitive problems had lower cortisol levels, while patients with ongoing respiratory or myalgic symptoms had higher levels of IL-6 and hs-CRP. However, more extensive studies with clustering are needed to validate these results.
Collapse
Affiliation(s)
- Antti Hurme
- Department of Infectious Diseases, Turku University Hospital and University of Turku, Turku, Finland.
- Institute of Biomedicine, University of Turku, Turku, Finland.
- Department of Internal Medicine, Lapland Central Hospital, Rovaniemi, Finland.
| | - Arja Viinanen
- Department of Pulmonary Diseases, Turku University Hospital and Department of Pulmonary Diseases and Clinical Allergology University of Turku, Turku, Finland
| | | | - Pinja Jalkanen
- Institute of Biomedicine, University of Turku, Turku, Finland
| | - Thijs Feuth
- Department of Pulmonary Diseases, Turku University Hospital and Department of Pulmonary Diseases and Clinical Allergology University of Turku, Turku, Finland
| | - Eliisa Löyttyniemi
- Department of Biostatistics, University of Turku and Turku University Hospital, Turku, Finland
| | - Tytti Vuorinen
- Institute of Biomedicine, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
| | - Anu Kantele
- Meilahti Vaccine Research Center, MeVac, Department of Infectious Diseases, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Jarmo Oksi
- Department of Infectious Diseases, Turku University Hospital and University of Turku, Turku, Finland
| | - Qiushui He
- Institute of Biomedicine, University of Turku, Turku, Finland
- InFlames Research Flagship Center, University of Turku, Turku, Finland
| | - Ilkka Julkunen
- Institute of Biomedicine, University of Turku, Turku, Finland
- InFlames Research Flagship Center, University of Turku, Turku, Finland
- Clinical Microbiology, Turku University Hospital, Turku, Finland
| |
Collapse
|
11
|
Whitaker-Hardin B, McGregor KM, Uswatte G, Lokken K. A Narrative Review of the Efficacy of Long COVID Interventions on Brain Fog, Processing Speed, and Other Related Cognitive Outcomes. Biomedicines 2025; 13:421. [PMID: 40002834 PMCID: PMC11853337 DOI: 10.3390/biomedicines13020421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
In the years following the global emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), or COVID-19, researchers have become acutely aware of long-term symptomology associated with this disease, often termed long COVID. Long COVID is associated with pervasive symptoms affecting multiple organ systems. Neurocognitive symptoms are reported by up to 40% of long COVID patients, with resultant effects of loss of daily functioning, employment issues, and enormous economic impact and high healthcare utilization. The literature on effective, safe, and non-invasive interventions for the remediation of the cognitive consequences of long COVID is scarce and poorly described. Of specific interest to this narrative review is the identification of potential interventions for long COVID-associated neurocognitive deficits. Articles were sourced from PubMed, EBSCO, Scopus, and Embase following Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Articles published between the dates of January 2020 and 30 June 2024 were included in the search. Twelve studies were included in the narrative review, including a feasibility study, a pilot study, a case series, a case study, and an observational study, in addition to three randomized clinical trials and four interventional studies. Overall, treatment interventions such as cognitive training, non-invasive brain stimulation therapy, exercise rehabilitation, targeted pharmacological intervention, and other related treatment paradigms show promise in reducing long COVID cognitive issues. This narrative review highlights the need for more rigorous experimental designs and future studies are needed to fully evaluate treatment interventions for persistent cognitive deficits associated with long COVID.
Collapse
Affiliation(s)
- Bryana Whitaker-Hardin
- Neuroscience Theme, Graduate Biomedical Sciences Doctoral Training Program, Joint Health Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Keith M. McGregor
- Birmingham Veterans Affairs Geriatric Research Education and Clinical Center, Birmingham Veterans Affairs Health Care System, Birmingham, AL 35294, USA;
- Department of Clinical and Diagnostic Sciences, School of Health Professions, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Gitendra Uswatte
- Departments of Psychology & Physical Therapy, College of Arts and Sciences, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Kristine Lokken
- Department of Psychiatry and Behavioral Neurobiology, Heersink School of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
12
|
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
|
13
|
Patel RS, Duque D, Bavananthasivam J, Hewitt M, Sandhu JK, Kumar R, Tran A, Agrawal B. Mixed lipopeptide-based mucosal vaccine candidate induces cross-variant immunity and protects against SARS-CoV-2 infection in hamsters. Immunohorizons 2025; 9:vlae011. [PMID: 39849995 PMCID: PMC11841972 DOI: 10.1093/immhor/vlae011] [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: 11/07/2024] [Accepted: 11/14/2024] [Indexed: 01/25/2025] Open
Abstract
The global dissemination of SARS-CoV-2 led to a worldwide pandemic in March 2020. Even after the official downgrading of the COVID-19 pandemic, infection with SARS-CoV-2 variants continues. The rapid development and deployment of SARS-CoV-2 vaccines helped to mitigate the pandemic to a great extent. However, the current vaccines are suboptimal; they elicit incomplete and short-lived protection and are ineffective against evolving virus variants. Updating the spike antigen according to the prevailing variant and repeated boosters is not the long-term solution. We have designed a lipopeptide-based, mucosal, pan-coronavirus vaccine candidate, derived from highly conserved and/or functional regions of the SARS-CoV-2 spike, nucleocapsid, and membrane proteins. Our studies demonstrate that the designed lipopeptides (LPMix) induced both cellular and humoral (mucosal and systemic) immune responses upon intranasal immunization in mice. Furthermore, the antibodies bound to the wild-type and mutated S proteins of SARS-CoV-2 variants of concern, including Alpha, Beta, Delta and Omicron, and also led to efficient neutralization in a surrogate viral neutralization assay. Our sequence alignment and 3-dimensional molecular modeling studies demonstrated that spike-derived epitopes, P1 and P2, are sequentially and/or structurally conserved among the SARS-CoV-2 variants. The addition of a novel mucosal adjuvant, heat-killed Caulobacter crescentus, to the lipopeptide vaccine significantly bolstered mucosal antibody responses. Finally, the lipopeptide-based intranasal vaccine demonstrated significant improvement in lung pathologies in a hamster model of SARS-CoV-2 infection. These studies are fundamentally important and open new avenues in the investigation of an innovative, broadly protective intranasal vaccine platform for SARS-CoV-2 and its variants.
Collapse
Affiliation(s)
- Raj S Patel
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| | - Diana Duque
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jegarubee Bavananthasivam
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Melissa Hewitt
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | - Jagdeep K Sandhu
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
| | | | - Anh Tran
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Babita Agrawal
- Department of Surgery, Faculty of Medicine and Dentistry, College of Health Sciences, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
14
|
Autaa G, Papagno L, Nogimori T, Boizard-Moracchini A, Korenkov D, Roy M, Suzuki K, Masuta Y, White E, Llewellyn-Lacey S, Yoshioka Y, Nicoli F, Price DA, Dechanet-Merville J, Yamamoto T, Pellegrin I, Appay V. Aging and inflammation limit the induction of SARS-CoV-2-specific CD8+ T cell responses in severe COVID-19. JCI Insight 2025; 10:e180867. [PMID: 39847442 PMCID: PMC11949069 DOI: 10.1172/jci.insight.180867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 01/13/2025] [Indexed: 01/24/2025] Open
Abstract
CD8+ T cells are critical for immune protection against severe COVID-19 during acute infection with SARS-CoV-2. However, the induction of antiviral CD8+ T cell responses varies substantially among infected people, and a better understanding of the mechanisms that underlie such immune heterogeneity is required for pandemic preparedness and risk stratification. In this study, we analyzed SARS-CoV-2-specific CD4+ and CD8+ T cell responses in relation to age, clinical status, and inflammation among patients infected primarily during the initial wave of the pandemic in France or Japan. We found that age-related contraction of the naive lymphocyte pool and systemic inflammation were associated with suboptimal SARS-CoV-2-specific CD4+ and, even more evidently, CD8+ T cell immunity in patients with acute COVID-19. No such differences were observed for humoral immune responses targeting the spike protein of SARS-CoV-2. We also found that the proinflammatory cytokine IL-18, concentrations of which were significantly elevated among patients with severe disease, suppressed the de novo induction and memory recall of antigen-specific CD8+ T cells, including those directed against SARS-CoV-2. These results potentially explain the vulnerability of older adults to infections that elicit a profound inflammatory response, exemplified by acute COVID-19.
Collapse
Affiliation(s)
- Gaëlle Autaa
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
| | - Laura Papagno
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
| | - Takuto Nogimori
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | | | - Daniil Korenkov
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
| | - Maeva Roy
- CHU Bordeaux, Laboratory of Immunology and Immunogenetics, 33000 Bordeaux, France
| | - Koichiro Suzuki
- The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Osaka, Japan
| | - Yuji Masuta
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Eoghann White
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
| | - Sian Llewellyn-Lacey
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Yasuo Yoshioka
- The Research Foundation for Microbial Diseases of Osaka University (BIKEN), Osaka, Japan
- Vaccine Creation Group, BIKEN Innovative Vaccine Research Alliance Laboratories, Research Institute for Microbial Diseases
- Laboratory of Nano-Design for Innovative Drug Development, Graduate School of Pharmaceutical Sciences, and
- Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Osaka, Japan
| | - Francesco Nicoli
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, Ferrara, Italy
| | - David A. Price
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Julie Dechanet-Merville
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
| | - Takuya Yamamoto
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| | - Isabelle Pellegrin
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
- CHU Bordeaux, Laboratory of Immunology and Immunogenetics, 33000 Bordeaux, France
| | - Victor Appay
- University of Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
- Laboratory of Precision Immunology, Center for Intractable Diseases and ImmunoGenomics, National Institutes of Biomedical Innovation, Health and Nutrition, Osaka, Japan
| |
Collapse
|
15
|
Iqbal NT, Khan H, Khalid A, Mahmood SF, Nasir N, Khanum I, de Siqueira I, Van Voorhis W. Chronic inflammation in post-acute sequelae of COVID-19 modulates gut microbiome: a review of literature on COVID-19 sequelae and gut dysbiosis. Mol Med 2025; 31:22. [PMID: 39849406 PMCID: PMC11756069 DOI: 10.1186/s10020-024-00986-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/01/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Long COVID or Post-acute sequelae of COVID-19 is an emerging syndrome, recognized in COVID-19 patients who suffer from mild to severe illness and do not recover completely. Most studies define Long COVID, through symptoms like fatigue, brain fog, joint pain, and headache prevailing four or more weeks post-initial infection. Global variations in Long COVID presentation and symptoms make it challenging to standardize features of Long COVID. Long COVID appears to be accompanied by an auto-immune multi-faceted syndrome where the virus or viral antigen persistence causes continuous stimulation of the immune response, resulting in multi-organ immune dysregulation. MAIN TEXT This review is focused on understanding the risk factors of Long COVID with a special emphasis on the dysregulation of the gut-brain axis. Two proposed mechanisms are discussed here. The first mechanism is related to the dysfunction of angiotensin-converting enzyme 2 receptor due to Severe Acute Respiratory Syndrome Corona Virus 2 infection, leading to impaired mTOR pathway activation, reduced AMP secretion, and causing dysbiotic changes in the gut. Secondly, gut-brain axis dysregulation accompanied by decreased production of short-chain fatty acids, impaired enteroendocrine cell function, and increased leakiness of the gut, which favors translocation of pathogens or lipopolysaccharide in circulation causing the release of pro-inflammatory cytokines. The altered Hypothalamic-Pituitary-Adrenal axis is accompanied by the reduced level of neurotransmitter, and decreased stimulation of the vagus nerve, which may cause neuroinflammation and dysregulation of serum cortisol levels. The dysbiotic microbiome in Long COVID patients is characterized by a decrease in beneficial short chain fatty acid-producing bacteria (Faecalibacterium, Ruminococcus, Dorea, and Bifidobacterium) and an increase in opportunistic bacteria (Corynebacterium, Streptococcus, Enterococcus). This dysbiosis is transient and may be impacted by interventions including probiotics, and dietary supplements. CONCLUSIONS Further studies are required to understand the geographic variation, racial and ethnic differences in phenotypes of Long COVID, the influence of viral strains on existing and emerging phenotypes, to explore long-term effects of gut dysbiosis, and gut-brain axis dysregulation, as well as the potential role of diet and probiotics in alleviating those symptoms.
Collapse
Affiliation(s)
- Najeeha Talat Iqbal
- Department of Biological and Biomedical Sciences, Department of Pediatrics and Child Health, Aga Khan University, Stadium Road, P. O Box 3500, Karachi, 74800, Pakistan.
- Department of Pediatrics & Child Health, Aga Khan University, Karachi, Pakistan.
| | - Hana Khan
- Undergraduate Medical Education (UGME), Year II, Aga Khan University, Karachi, Pakistan
| | - Aqsa Khalid
- Department of Pediatrics & Child Health, Aga Khan University, Karachi, Pakistan
| | | | - Nosheen Nasir
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | - Iffat Khanum
- Department of Medicine, Aga Khan University, Karachi, Pakistan
| | | | - Wes Van Voorhis
- Center for Emerging and Re-emerging Infectious Diseases (CERID), University of Washington, Seattle, USA
| |
Collapse
|
16
|
Guironnet-Paquet A, Hamzeh-Cognasse H, Berard F, Cognasse F, Richard JC, Yonis H, Mezidi M, Desebbe O, Delannoy B, Demeret S, Marois C, Saheb S, Le QV, Schoeffler M, Pugliesi PS, Debord S, Bastard P, Cobat A, Casanova JL, Pescarmona R, Viel S, Nicolas JF, Nosbaum A, Vocanson M, Hequet O. Therapeutic plasma exchange accelerates immune cell recovery in severe COVID-19. Front Immunol 2025; 15:1492672. [PMID: 39896810 PMCID: PMC11782122 DOI: 10.3389/fimmu.2024.1492672] [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: 09/07/2024] [Accepted: 12/04/2024] [Indexed: 02/04/2025] Open
Abstract
Background Immunological disturbances (anti-type I IFN auto-antibody production, cytokine storm, lymphopenia, T-cell hyperactivation and exhaustion) are responsible for disease exacerbation during severe COVID-19 infections. Methods In this study, we set up a prospective, randomised clinical trial (ClinicalTrials.gov ID: NCT04751643) and performed therapeutic plasma exchange (TPE) in severe COVID-19 patients in order to decrease excess cytokines and auto-antibodies and to assess whether adding TPE to the standard treatment (ST, including corticosteroids plus high-flow rate oxygen) could help restore immune parameters and limit the progression of acute respiratory distress syndrome (ARDS). Results As expected, performing TPE decreased the amount of anti-type I IFN auto-antibodies and improved the elimination or limited the production of certain inflammatory mediators (IL-18, IL-7, CCL2, CCL3, etc.) circulating in the blood of COVID-19 patients, compared to ST controls. Interestingly, while TPE did not influence changes in ARDS parameters throughout the protocol, it proved more effective than ST in reversing lymphopenia, preventing T-cell hyperactivation and reducing T-cell exhaustion, notably in a fraction of TPE patients who had an early favourable respiratory outcome. TPE also restored appropriate numbers of CD4+ and CD8+ T-cell memory populations and increased the number of circulating virus-specific T cells in these patients. Conclusion Our results therefore indicate that the addition of TPE sessions to the standard treatment accelerates immune cell recovery and contributes to the development of appropriate antiviral T-cell responses in some patients with severe COVID-19 disease.
Collapse
Affiliation(s)
- Aurelie Guironnet-Paquet
- Apheresis Unit, Etablissement Français du Sang Auvergne-Rhône-Alpes, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre Bénite, France
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| | - Hind Hamzeh-Cognasse
- University of Jean Monnet, Mines Saint-Étienne, Institut National de la Santé et de la Recherche Médicale (INSERM), U 1059 SAINBIOSE, Saint-Étienne, France
| | - Frederic Berard
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Fabrice Cognasse
- University of Jean Monnet, Mines Saint-Étienne, Institut National de la Santé et de la Recherche Médicale (INSERM), U 1059 SAINBIOSE, Saint-Étienne, France
- Scientific Department, Etablissement Français du Sang Auvergne-Rhône-Alpes, Saint-Etienne, France
| | - Jean Christophe Richard
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Hodane Yonis
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Mehdi Mezidi
- Intensive Care Unit, Centre Hospitalier Croix–Rousse, Hospices Civils de Lyon (HCL), Lyon, France
| | - Olivier Desebbe
- Department of Anesthesiology and Perioperative Medicine, Sauvegarde Clinic, Ramsay Santé, Lyon, France
| | - Bertrand Delannoy
- Department of Anesthesiology and Perioperative Medicine, Sauvegarde Clinic, Ramsay Santé, Lyon, France
| | - Sophie Demeret
- Neuro-Intensive Care Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Clemence Marois
- Neuro-Intensive Care Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
- Sorbonne Université, Institut du Cerveau, Paris Brain Institute, Institut du Cerveau et de la Moelle (ICM), Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Departement Médico-Universitaire (DMU) Neurosciences 6, Paris, France
- Groupe de Recherche Clinique en REanimation et Soins Intensifs du Patient en Insuffisance Respiratoire aiguE (GRC-RESPIRE), Sorbonne Université, Paris, France
| | - Samir Saheb
- Hemobiotherapy Unit, Assistance Publique des Hopitaux de Paris (AP-HP), Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Quoc Viet Le
- Intensive Care Unit, Medipôle Lyon Villeurbanne, Villeurbanne, France
| | - Mathieu Schoeffler
- Department of Anesthesiology and Intensive Care Unit, Centre Hospitalier de Montélimar, Montélimar, France
| | - Paul Simon Pugliesi
- Intensive Care Unit, Centre Hospitalier William Morey, Chalon sur Saône, France
| | - Sophie Debord
- Department of Anesthesiology and Intensive Care Medicine, Edouard Herriot Hospital, Hospices Civils de Lyon (HCL), Lyon, France
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique des Hopitaux de Paris (AP-HP), Paris, France
| | - Aurélie Cobat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
| | - Jean Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale (INSERM) U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States
- Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, Assistance Publique des Hopitaux de Paris (AP-HP), Paris, France
- Howards Hugues Medical Institute, New York, NY, United States
| | - Rémi Pescarmona
- Immun Monitorage Laboratory, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Sébastien Viel
- Plateforme de Biothérapies et de production de Médicaments de Thérapie Innovante (MTI), Hôpital Edouard Herriot, Hospices Civils de Lyon (HCL), Lyon, France
| | - Jean François Nicolas
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Audrey Nosbaum
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
- Clinical Immunology and Allergology, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre-Bénite, France
| | - Marc Vocanson
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| | - Olivier Hequet
- Apheresis Unit, Etablissement Français du Sang Auvergne-Rhône-Alpes, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon (HCL), Pierre Bénite, France
- International Center for Infectiology Research (CIRI), Université de Lyon, Institut National de la Santé et de la Recherche Médicale (INSERM), U1111, Lyon, France
| |
Collapse
|
17
|
Genova SN, Pencheva MM, Abadjieva TI, Atanasov NG. Cellular and immune response in fatal COVID-19 pneumonia. Pan Afr Med J 2024; 49:130. [PMID: 40190436 PMCID: PMC11971930 DOI: 10.11604/pamj.2024.49.130.45739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Accepted: 12/03/2024] [Indexed: 04/09/2025] Open
Abstract
Introduction the severity of COVID-19, causing fatal pneumonia, acute respiratory distress syndrome (ARDS), and thrombotic complications, is linked to intense inflammation. Elevated CD4+ and CD8+ cells in the lungs indicate harmful inflammation in severe cases. This study investigates immune responses in lung tissues of deceased patients across different stages of COVID-19 pneumonia. Methods lung tissues from 160 fatal COVID-19 cases, diagnosed via Real-Time RT-PCR, were histologically analyzed to identify pneumonia stages. Inflammatory cell counts were assessed immunohistochemically. Non-parametric tests analyzed categorical variables, while regression analysis evaluated relationships between continuous variables. Results the average patient age was 68.1 years (± 12.6). Microscopic analysis identified four pneumonia stages. CD4+, CD68 (macrophages), and IgG4 levels peaked by day 14, with notable elevation within seven days of symptom onset. CD4+ levels were significantly lower in DAD pneumonia (49.4% ± 15.7%) compared to ARDS (66.4% ± 19.3%) and thrombosis (70.2% ± 28.9%) (p < 0.05). Male patients had higher CD4+ values (68.5% ± 21.1%) than females (56.9% ± 22.4%) (p < 0.05). B cells (CD20) and NK cells were depleted across all stages. IgG4 expression reached 80-90% in acute phases but was nearly absent during organization and fibrosis stages. Conclusion a sharp decline in CD4+ and CD8+ during acute pneumonia and sepsis reflects immune exhaustion, while their elevation in ARDS and thrombosis likely triggers cytokine storms, causing severe lung damage. Elevated IgG4 levels in acute lung tissue correlate with fatal outcomes in severe COVID-19.
Collapse
Affiliation(s)
- Sylvia Nikolaeva Genova
- Department of General and Clinical Pathology, Medical Faculty, Medical University Plovdiv, Plovdiv, Bulgaria
- St George University Hospital, Plovdiv, Bulgaria
| | - Mina Miroslavova Pencheva
- Department of Medical Physics and Biophysics, Faculty of Pharmacy, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Tsvetana Ivanova Abadjieva
- St George University Hospital, Plovdiv, Bulgaria
- Department of Dermatology and Venereology, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Nikolay Georgiev Atanasov
- Department of Health Management and Health Economics, Faculty of Public Health, Medical University Plovdiv, Plovdiv, Bulgaria
| |
Collapse
|
18
|
Yang Y, Miller H, Byazrova MG, Cndotti F, Benlagha K, Camara NOS, Shi J, Forsman H, Lee P, Yang L, Filatov A, Zhai Z, Liu C. The characterization of CD8 + T-cell responses in COVID-19. Emerg Microbes Infect 2024; 13:2287118. [PMID: 37990907 PMCID: PMC10786432 DOI: 10.1080/22221751.2023.2287118] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/19/2023] [Indexed: 11/23/2023]
Abstract
This review gives an overview of the protective role of CD8+ T cells in SARS-CoV-2 infection. The cross-reactive responses intermediated by CD8+ T cells in unexposed cohorts are described. Additionally, the relevance of resident CD8+ T cells in the upper and lower airway during infection and CD8+ T-cell responses following vaccination are discussed, including recent worrisome breakthrough infections and variants of concerns (VOCs). Lastly, we explain the correlation between CD8+ T cells and COVID-19 severity. This review aids in a deeper comprehension of the association between CD8+ T cells and SARS-CoV-2 and broadens a vision for future exploration.
Collapse
Affiliation(s)
- Yuanting Yang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, USA
| | - Maria G. Byazrova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Fabio Cndotti
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kamel Benlagha
- Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Niels Olsen Saraiva Camara
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Junming Shi
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Huamei Forsman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Lu Yang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Alexander Filatov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Zhimin Zhai
- Department of Hematology, The Second Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| |
Collapse
|
19
|
Krishna B, Metaxaki M, Perera M, Wills M, Sithole N. Comparison of different T cell assays for the retrospective determination of SARS-CoV-2 infection. J Gen Virol 2024; 105. [PMID: 39704047 DOI: 10.1099/jgv.0.002055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2024] Open
Abstract
It is important to be able to retrospectively determine severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections with high accuracy, both for post-coronavirus disease 2019 (COVID-19) epidemiological studies, and to distinguish between Long COVID and other multi-syndromic diseases that have overlapping symptoms. Although serum antibody levels can be measured to retrospectively diagnose SARS-CoV-2 infections, peptide stimulation of memory T cell responses is a more sensitive approach. This is because robust memory T cells are generated after SARS-CoV-2 infection and persist even after antibodies wane below detectability thresholds. In this study, we compare T cell responses using FluoroSpot-based methods and overnight stimulation of whole blood with SARS-CoV-2 peptides followed by an ELISA. Both approaches have comparable sensitivity and specificity but require different equipment and samples to be used. Furthermore, the elimination of peptides that cross-react with other coronaviruses increases the assay specificity but trades off some sensitivity. Finally, this approach can be used on archival, cryopreserved PBMCs. This work shows comparative advantages for several methods to measure SARS-CoV-2 T cell responses that could be utilized by any laboratory studying the effects of the coronavirus disease 2019 pandemic.
Collapse
Affiliation(s)
- Benjamin Krishna
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Marina Metaxaki
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Marianne Perera
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
- Division of Virology, Department of Pathology, University of Cambridge, Cambridge, UK
| | - Mark Wills
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| | - Nyarie Sithole
- Cambridge Institute of Therapeutic Immunology and Infectious Disease (CITIID), Cambridge, UK
| |
Collapse
|
20
|
Li Q, Ma Y, He P, Chen D, Zhang T, Wang X, Xu Y, Li P, Wen W, Wang Z. Long COVID symptoms 6 months after acute infection among people living with HIV and people not living with HIV. Front Immunol 2024; 15:1430214. [PMID: 39669584 PMCID: PMC11634828 DOI: 10.3389/fimmu.2024.1430214] [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: 06/01/2024] [Accepted: 10/25/2024] [Indexed: 12/14/2024] Open
Abstract
Background Chronic viral infections, such as Human Immunodeficiency Virus (HIV), and their reactivation are considered potential contributing factors to Long-Corona Virus Disease (LC). However, research on the long-term sequelae of Long-COVID in individuals with HIV is limited. Methods We conducted a case-control study involving a total of 84 participants categorized into two groups: people living with HIV (PLWH) and people not living with HIV (PNLWH) within the six-month post-infection LC population. Differences in sequelae symptoms, cardiovascular biomarkers (VCAM-1, ICAM-1, and ACE2), Severe Acute Respiratory Syndrome Coronavirus 2 neutralization antibodies (SARS-CoV-2 nAb) and cytokines (IFN-γ, IL-6, and IL-17) were analyzed between the two groups. Results After 6 months of infection, PLWH exhibited significantly higher serum levels of ACE2, VCAM-1, and ICAM-1 (P < 0.01, respectively) compared to PNLWH with COVID-19. Additionally, sequelae symptoms were more pronounced in PNLWH, and there were no differences in serum levels of IFN-γ, TNF-α, IL-6, and IL-17 between the two groups (P > 0.05, respectively). Conclusion PLWH had lower symptoms of LC and reduced frequency of symptoms, increased cardiovascular risk factors, and no differences in levels of inflammation or SARS-CoV-2 nAb levels when compared to PNLWH.
Collapse
Affiliation(s)
- Qin Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming, China
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, China
| | - Yijie Ma
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Peng He
- Traditional Chinese Medicine Department, Tengchong People's Hospital, Tengchong, China
| | - Dongqiong Chen
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Tingrui Zhang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiaoying Wang
- Research Management Section, Kunming Municipal Hospital of Traditional Chinese Medicine, Kunming, China
| | - Ying Xu
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Peiming Li
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Weibo Wen
- Yunnan Key Laboratory of Integrated Traditional Chinese and Western Medicine for Chronic Disease in Prevention and Treatment, Yunnan University of Chinese Medicine, Kunming, China
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Zefeng Wang
- College of Traditional Chinese Medicine, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
21
|
Yosri M, Dokhan M, Aboagye E, Al Moussawy M, Abdelsamed HA. Mechanisms governing bystander activation of T cells. Front Immunol 2024; 15:1465889. [PMID: 39669576 PMCID: PMC11635090 DOI: 10.3389/fimmu.2024.1465889] [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: 07/17/2024] [Accepted: 10/31/2024] [Indexed: 12/14/2024] Open
Abstract
The immune system is endowed with the capacity to distinguish between self and non-self, so-called immune tolerance or "consciousness of the immune system." This type of awareness is designed to achieve host protection by eliminating cells expressing a wide range of non-self antigens including microbial-derived peptides. Such a successful immune response is associated with the secretion of a whole spectrum of soluble mediators, e.g., cytokines and chemokines, which not only contribute to the clearance of infected host cells but also activate T cells that are not specific to the original cognate antigen. This kind of non-specific T-cell activation is called "bystander activation." Although it is well-established that this phenomenon is cytokine-dependent, there is evidence in the literature showing the involvement of peptide/MHC recognition depending on the type of T-cell subset (naive vs. memory). Here, we will summarize our current understanding of the mechanism(s) of bystander T-cell activation as well as its biological significance in a wide range of diseases including microbial infections, cancer, auto- and alloimmunity, and chronic inflammatory diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Mohammed Yosri
- The Regional Center for Mycology and Biotechnology, Al-Azhar University, Cairo, Egypt
| | - Mohamed Dokhan
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Elizabeth Aboagye
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
| | - Mouhamad Al Moussawy
- Starzl Transplantation Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, United States
| | - Hossam A. Abdelsamed
- Immunology Center of Georgia (IMMCG), Medical College of Georgia (MCG), Augusta University, Augusta, GA, United States
- Department of Physiology, Augusta University, Augusta, GA, United States
| |
Collapse
|
22
|
Ćorović A, Zhao X, Huang Y, Newland SR, Gopalan D, Harrison J, Giakomidi D, Chen S, Yarkoni NS, Wall C, Peverelli M, Sriranjan R, Gallo A, Graves MJ, Sage A, Lyons PA, Sithole N, Bennett MR, Rudd JHF, Mallat Z, Zhao TX, Nus M, Tarkin JM. Coronavirus disease 2019-related myocardial injury is associated with immune dysregulation in symptomatic patients with cardiac magnetic resonance imaging abnormalities. Cardiovasc Res 2024; 120:1752-1767. [PMID: 39073768 PMCID: PMC11587552 DOI: 10.1093/cvr/cvae159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 05/01/2024] [Accepted: 05/23/2024] [Indexed: 07/30/2024] Open
Abstract
AIMS While acute cardiovascular complications of coronavirus disease 2019 (COVID-19) are well described, less is known about longer-term cardiac sequelae. For many individuals with cardiac signs or symptoms arising after COVID-19 infection, the aetiology remains unclear. We examined immune profiles associated with magnetic resonance imaging (MRI) abnormalities in patients with unexplained cardiac injury after COVID-19. METHODS AND RESULTS Twenty-one participants {mean age 47 [standard deviation (SD) 13] years, 71% female} with long COVID-19 (n = 17), raised troponin (n = 2), or unexplained new-onset heart failure (n = 2), who did not have pre-existing heart conditions or recent steroid/immunosuppression treatment, were enrolled a mean 346 (SD 191) days after COVID-19 infection in a prospective observational study. Cardiac MRI and blood sampling for deep immunophenotyping using mass cytometry by time of flight and measurement of proteomic inflammatory markers were performed. Nine of the 21 (43%) participants had MRI abnormalities (MRI(+)), including non-ischaemic patterns of late gadolinium enhancement and/or visually overt myocardial oedema in 8 people. One patient had mildly impaired biventricular function without fibrosis or oedema, and two had severe left ventricular (LV) impairment. MRI(+) individuals had higher blood CCL3, CCL7, FGF-23, and CD4 Th2 cells, and lower CD8 T effector memory (TEM) cells, than MRI(-). Cluster analysis revealed lower expression of inhibitory receptors PD1 and TIM3 in CD8 TEM cells from MRI(+) patients than MRI(-) patients, and functional studies of CD8 T αβ cells showed higher proportions of cytotoxic granzyme B+(GZB+)-secreting cells upon stimulation. CD8 TEM cells and CCL7 were the strongest predictors of MRI abnormalities in a least absolute shrinkage and selection operator regression model (composite area under the curve 0.96, 95% confidence interval 0.88-1.0). CCL7 was correlated with diffuse myocardial fibrosis/oedema detected by quantitative T1 mapping (r = 0.47, P = 0.04). CONCLUSION COVID-19-related cardiac injury in symptomatic patients with non-ischaemic myocarditis-like MRI abnormalities is associated with immune dysregulation, including decreased peripheral CD8 TEM cells and increased CCL7, persisting long after the initial infection.
Collapse
Affiliation(s)
- Andrej Ćorović
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Xiaohui Zhao
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Yuan Huang
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Stephen R Newland
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Deepa Gopalan
- Department of Radiology, Cambridge University Hospitals NHS Trust, Cambridge, UK
| | - James Harrison
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Despina Giakomidi
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Shanna Chen
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Natalia S Yarkoni
- Cell Phenotyping Hub, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Christopher Wall
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Marta Peverelli
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Rouchelle Sriranjan
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Arianna Gallo
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Martin J Graves
- Department of Radiology, University of Cambridge, Cambridge, UK
| | - Andrew Sage
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Nyarie Sithole
- Infectious Diseases, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Martin R Bennett
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - James H F Rudd
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ziad Mallat
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Tian X Zhao
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Meritxell Nus
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Jason M Tarkin
- Section of Cardiorespiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, UK
| |
Collapse
|
23
|
H-Vázquez J, Cal-Sabater P, Arribas-Rodríguez E, Fiz-López A, Perez-Segurado C, Martín-Muñoz Á, De Prado Á, Perez Mazzali M, de Castro CG, del Hierro AG, de la Fuente Graciani I, Pérez González S, Gutiérrez S, Tellería P, Novoa C, Rojo Rello S, Garcia-Blesa A, Sedano R, Martínez García AM, Garcinuño Pérez S, Domínguez-Gil M, Hernán García C, Guerra MM, Muñoz-Sánchez E, Barragan-Pérez C, Diez Morales S, Casazza Donnarumma O, Ramos Pollo D, Santamarta Solla N, Álvarez Manzanares PM, Bravo S, García Alonso C, Avendaño Fernández LA, Gay Alonso J, Garrote JA, Arranz E, Eiros JM, Rescalvo Santiago F, Quevedo Villegas C, Tamayo E, Orduña A, Dueñas C, Peñarrubia MJ, Cuesta-Sancho S, Montoya M, Bernardo D. Unbiased immunome characterisation correlates with COVID-19 mRNA vaccine failure in immunocompromised adults. Front Immunol 2024; 15:1405217. [PMID: 39611158 PMCID: PMC11602488 DOI: 10.3389/fimmu.2024.1405217] [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: 03/22/2024] [Accepted: 10/23/2024] [Indexed: 11/30/2024] Open
Abstract
Introduction Coronavirus disease 2019 (COVID-19) affects the population unequally, with a greater impact on older and immunosuppressed people. Methods Hence, we performed a prospective experimental cohort study to characterise the effect of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccination in immune-compromised patients (older adults and oncohaematologic patients), compared with healthy counterparts, based on deep characterisation of the circulating immune cell subsets. Results and discussion While acquired humoral and cellular memory did not predict subsequent infection 18 months after full vaccination, spectral and computational cytometry revealed several subsets within the CD8+ T-cells, B-cells, natural killer (NK) cells, monocytes and TEMRA Tγδ cells that were differentially expressed in individuals who were subsequently infected and not infected not just following immunisation, but also prior to vaccination. Of note, we found up to seven clusters within the TEMRA Tγδ cell population, with some of them being expanded and others decreased in subsequently infected individuals. Moreover, some of these cellular clusters were also related to COVID-19-induced hospitalisation in oncohaematologic patients. Therefore, we have identified a cellular signature that even before vaccination is related to COVID-19 vulnerability as opposed to the acquisition of cellular and/or humoral memory following vaccination with SARS-CoV-2 messenger RNA (mRNA) vaccines.
Collapse
Affiliation(s)
- Juan H-Vázquez
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - Paloma Cal-Sabater
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - Elisa Arribas-Rodríguez
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - Aida Fiz-López
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - Candido Perez-Segurado
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - Álvaro Martín-Muñoz
- Flow Cytometry Facility, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - Ángel De Prado
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - Marina Perez Mazzali
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - Carolina G. de Castro
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - Alejandro G. del Hierro
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | | | - Sonia Pérez González
- Department of Hematology, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Sara Gutiérrez
- Internal Medicine Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Pablo Tellería
- Internal Medicine Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Cristina Novoa
- Internal Medicine Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Silvia Rojo Rello
- Microbiology and Inmunology Unit, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Antonio Garcia-Blesa
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - Rosa Sedano
- Internal Medicine Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | | | - Sonsoles Garcinuño Pérez
- Microbiology and Inmunology Unit, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Marta Domínguez-Gil
- Microbiology Department, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Cristina Hernán García
- Servicio de Medicina Preventiva, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Ma Mercedes Guerra
- Servicio de Medicina Preventiva, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Eduardo Muñoz-Sánchez
- Unidad de Cuidados Paliativos del Hospital Universitario Río Hortega, Valladolid, Spain
| | | | - Soraya Diez Morales
- Unidad de Cuidados Paliativos del Hospital Universitario Río Hortega, Valladolid, Spain
| | | | - Daniel Ramos Pollo
- Unidad de Cuidados Paliativos del Hospital Universitario Río Hortega, Valladolid, Spain
| | | | | | - Sara Bravo
- Department of Hematology, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Cristina García Alonso
- Department of Hematology, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | | | | | - José A. Garrote
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - Eduardo Arranz
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - José María Eiros
- Microbiology Department, Hospital Universitario Río Hortega, Valladolid, Spain
| | - Fernando Rescalvo Santiago
- Servicio de Prevención de Riesgos Laborales, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Carolina Quevedo Villegas
- Servicio de Prevención de Riesgos Laborales, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Eduardo Tamayo
- Unidad Investigación, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Antonio Orduña
- Microbiology and Inmunology Unit, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Carlos Dueñas
- Internal Medicine Department, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - María Jesús Peñarrubia
- Department of Hematology, Hospital Clínico Universitario de Valladolid, Valladolid, Spain
| | - Sara Cuesta-Sancho
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
| | - María Montoya
- Viral Immunology, Therapies and Vaccines Lab, Centro de Investigaciones Biológicas Margarita Salas (CIB), Madrid, Spain
| | - David Bernardo
- Mucosal Immunology Lab, Unit of Excellence Institute of Biomedicine and Molecular Genetics of Valladolid (IBGM, University of Valladolid-CSIC), Valladolid, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| |
Collapse
|
24
|
Babcock BR, Kosters A, Eddins DJ, Donaire MSB, Sarvadhavabhatla S, Pae V, Beltran F, Murray VW, Gill G, Xie G, Dobosh BS, Giacalone VD, Tirouvanziam RM, Ramonell RP, Jenks SA, Sanz I, Lee FEH, Roan NR, Lee SA, Ghosn EEB. Transient anti-interferon autoantibodies in the airways are associated with recovery from COVID-19. Sci Transl Med 2024; 16:eadq1789. [PMID: 39504354 PMCID: PMC11924959 DOI: 10.1126/scitranslmed.adq1789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024]
Abstract
Preexisting anti-interferon-α (anti-IFN-α) autoantibodies in blood are associated with susceptibility to life-threatening COVID-19. However, it is unclear whether anti-IFN-α autoantibodies in the airways, the initial site of infection, can also determine disease outcomes. In this study, we developed a multiparameter technology, FlowBEAT, to quantify and profile the isotypes of anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and anti-IFN-α antibodies in longitudinal samples collected over 20 months from the airways and blood of 129 donors spanning mild to severe COVID-19. We found that nasal IgA1 anti-IFN-α autoantibodies were induced after infection onset in more than 70% of mild and moderate COVID-19 cases and were associated with robust anti-SARS-CoV-2 immunity, fewer symptoms, and efficient recovery. Nasal anti-IFN-α autoantibodies followed the peak of host IFN-α production and waned with disease recovery, revealing a regulated balance between IFN-α and anti-IFN-α response. In contrast, systemic IgG1 anti-IFN-α autoantibodies appeared later and were detected only in a subset of patients with elevated systemic inflammation and worsening symptoms. These data reveal a protective role for nasal anti-IFN-α in the immunopathology of COVID-19 and suggest that anti-IFN-α autoantibodies may serve a homeostatic function to regulate host IFN-α after viral infection in the respiratory mucosa.
Collapse
Affiliation(s)
- Benjamin R Babcock
- Division of Immunology and Rheumatology, Department of Medicine, Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322, USA
| | - Astrid Kosters
- Division of Immunology and Rheumatology, Department of Medicine, Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322, USA
| | - Devon J Eddins
- Division of Immunology and Rheumatology, Department of Medicine, Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322, USA
| | - Maria Sophia Baluyot Donaire
- Division of HIV, Infectious Diseases & Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Sannidhi Sarvadhavabhatla
- Division of HIV, Infectious Diseases & Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Vivian Pae
- Division of HIV, Infectious Diseases & Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Fiona Beltran
- Division of HIV, Infectious Diseases & Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Victoria W Murray
- Division of HIV, Infectious Diseases & Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Gurjot Gill
- Division of HIV, Infectious Diseases & Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Guorui Xie
- Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Brian S Dobosh
- Department of Pediatrics, Emory University, Atlanta, GA 30322, USA
| | | | | | - Richard P Ramonell
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322, USA
| | - Scott A Jenks
- Division of Immunology and Rheumatology, Department of Medicine, Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322, USA
| | - Ignacio Sanz
- Division of Immunology and Rheumatology, Department of Medicine, Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322, USA
| | - F Eun-Hyung Lee
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Department of Medicine, Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322, USA
| | - Nadia R Roan
- Gladstone Institutes, San Francisco, CA 94158, USA
- Department of Urology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Sulggi A Lee
- Division of HIV, Infectious Diseases & Global Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Eliver E B Ghosn
- Division of Immunology and Rheumatology, Department of Medicine, Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322, USA
- Emory Vaccine Center, Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| |
Collapse
|
25
|
Perdiguero B, Álvarez E, Marcos-Villar L, Sin L, López-Bravo M, Valverde JR, Sorzano CÓS, Falqui M, Coloma R, Esteban M, Guerra S, Gómez CE. B and T Cell Bi-Cistronic Multiepitopic Vaccine Induces Broad Immunogenicity and Provides Protection Against SARS-CoV-2. Vaccines (Basel) 2024; 12:1213. [PMID: 39591118 PMCID: PMC11598604 DOI: 10.3390/vaccines12111213] [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: 09/18/2024] [Revised: 10/18/2024] [Accepted: 10/22/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND The COVID-19 pandemic, caused by SARS-CoV-2, has highlighted the need for vaccines targeting both neutralizing antibodies (NAbs) and long-lasting cross-reactive T cells covering multiple viral proteins to provide broad and durable protection against emerging variants. METHODS To address this, here we developed two vaccine candidates, namely (i) DNA-CoV2-TMEP, expressing the multiepitopic CoV2-TMEP protein containing immunodominant and conserved T cell regions from SARS-CoV-2 structural proteins, and (ii) MVA-CoV2-B2AT, encoding a bi-cistronic multiepitopic construct that combines conserved B and T cell overlapping regions from SARS-CoV-2 structural proteins. RESULTS Both candidates were assessed in vitro and in vivo demonstrating their ability to induce robust immune responses. In C57BL/6 mice, DNA-CoV2-TMEP enhanced the recruitment of innate immune cells and stimulated SARS-CoV-2-specific polyfunctional T cells targeting multiple viral proteins. MVA-CoV2-B2AT elicited NAbs against various SARS-CoV-2 variants of concern (VoCs) and reduced viral replication and viral yields against the Beta variant in susceptible K18-hACE2 mice. The combination of MVA-CoV2-B2AT with a mutated ISG15 form as an adjuvant further increased the magnitude, breadth and polyfunctional profile of the response. CONCLUSION These findings underscore the potential of these multiepitopic proteins when expressed from DNA or MVA vectors to provide protection against SARS-CoV-2 and its variants, supporting their further development as next-generation COVID-19 vaccines.
Collapse
Affiliation(s)
- Beatriz Perdiguero
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Enrique Álvarez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
| | - Laura Marcos-Villar
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - Laura Sin
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| | - María López-Bravo
- Department of Microbial Biotechnology, CNB-CSIC, 28049 Madrid, Spain;
| | | | | | - Michela Falqui
- Department of Preventive Medicine, Public Health and Microbiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.F.); (R.C.); (S.G.)
| | - Rocío Coloma
- Department of Preventive Medicine, Public Health and Microbiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.F.); (R.C.); (S.G.)
| | - Mariano Esteban
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
| | - Susana Guerra
- Department of Preventive Medicine, Public Health and Microbiology, Faculty of Medicine, Universidad Autónoma de Madrid, 28029 Madrid, Spain; (M.F.); (R.C.); (S.G.)
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Carmen Elena Gómez
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología (CNB), Consejo Superior de Investigaciones Científicas (CSIC), 28049 Madrid, Spain; (E.Á.); (L.M.-V.); (M.E.)
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain;
| |
Collapse
|
26
|
Bozkus CC, Brown M, Velazquez L, Thomas M, Wilson EA, O’Donnell T, Ruchnewitz D, Geertz D, Bykov Y, Kodysh J, Oguntuyo KY, Roudko V, Hoyos D, Srivastava KD, Kleiner G, Alshammary H, Karekar N, McClain C, Gopal R, Nie K, Del Valle D, Delbeau-Zagelbaum D, Rodriguez D, Setal J, Carroll E, Wiesendanger M, Gulko PS, Charney A, Merad M, Kim-Schulze S, Lee B, Wajnberg A, Simon V, Greenbaum BD, Chowell D, Vabret N, Luksza M, Bhardwaj N. T cell epitope mapping reveals immunodominance of evolutionarily conserved regions within SARS-CoV-2 proteome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619918. [PMID: 39484455 PMCID: PMC11527131 DOI: 10.1101/2024.10.23.619918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
As SARS-CoV-2 variants continue to emerge capable of evading neutralizing antibodies, it has become increasingly important to fully understand the breadth and functional profile of T cell responses to determine their impact on the immune surveillance of variant strains. Here, sampling healthy individuals, we profiled the kinetics and polyfunctionality of T cell immunity elicited by mRNA vaccination. Modeling of anti-spike T cell responses against ancestral and variant strains of SARS-CoV-2 suggested that epitope immunodominance and cross-reactivity are major predictive determinants of T cell immunity. To identify immunodominant epitopes across the viral proteome, we generated a comprehensive map of CD4+ and CD8+ T cell epitopes within non-spike proteins that induced polyfunctional T cell responses in convalescent patients. We found that immunodominant epitopes mainly resided within regions that were minimally disrupted by mutations in emerging variants. Conservation analysis across historical human coronaviruses combined with in silico alanine scanning mutagenesis of non-spike proteins underscored the functional importance of mutationally-constrained immunodominant regions. Collectively, these findings identify immunodominant T cell epitopes across the mutationally-constrained SARS-CoV-2 proteome, potentially providing immune surveillance against emerging variants, and inform the design of next-generation vaccines targeting antigens throughout SARS-CoV-2 proteome for broader and more durable protection.
Collapse
Affiliation(s)
- Cansu Cimen Bozkus
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Parker Institute of Cancer Immunotherapy, San Francisco, CA, USA
| | - Matthew Brown
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Parker Institute of Cancer Immunotherapy, San Francisco, CA, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Leandra Velazquez
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marcus Thomas
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Eric A. Wilson
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Timothy O’Donnell
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Denis Ruchnewitz
- Institute for Biological Physics, University of Cologne, 50937 Cologne, Germany
| | - Douglas Geertz
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yonina Bykov
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Julia Kodysh
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kasopefoluwa Y. Oguntuyo
- The Department of Medicine, The Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vladimir Roudko
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Hoyos
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Komal D. Srivastava
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giulio Kleiner
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Hala Alshammary
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Neha Karekar
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher McClain
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ramya Gopal
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kai Nie
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Diane Del Valle
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Denise Rodriguez
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica Setal
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | | | - Emily Carroll
- The Department of Medicine, The Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Margrit Wiesendanger
- The Department of Medicine, The Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Percio S. Gulko
- The Department of Medicine, The Division of Rheumatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Charney
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Miriam Merad
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Seunghee Kim-Schulze
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Ania Wajnberg
- The Department of Medicine, The Division of General Internal Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Viviana Simon
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Benjamin D Greenbaum
- Computational Oncology, Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Physiology, Biophysics & Systems Biology, Weill Cornell Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Diego Chowell
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nicolas Vabret
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marta Luksza
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nina Bhardwaj
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- The Department of Medicine, The Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Parker Institute of Cancer Immunotherapy, San Francisco, CA, USA
| |
Collapse
|
27
|
Bean DJ, Liang YM, Sagar M. Recent Endemic Coronavirus Infection Associates With Higher SARS-CoV-2 Cross-Reactive Fc Receptor Binding Antibodies. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619886. [PMID: 39484477 PMCID: PMC11527020 DOI: 10.1101/2024.10.23.619886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Recent documented infection with an endemic coronavirus (eCoV) associates with less severe coronavirus disease 2019 (COVID-19), yet the immune mechanism behind this protection has not been fully explored. We measured both antibody and T cell responses against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in SARS-CoV-2 naïve individuals classified into two groups, either with or without presumed recent eCoV infections. There was no difference in neutralizing antibodies and T cell responses against SARS-CoV-2 antigens between the two groups. SARS-CoV-2 naïve individuals with recent presumed eCoV infection, however, had higher levels of Fc receptor (FcR) binding antibodies against eCoV spikes (S) and SARS-CoV-2 S2. There was also a significant correlation between eCoV and SARS-CoV-2 FcR binding antibodies. Recent eCoV infection boosts cross-reactive antibodies that can mediate Fc effector functions, and this may play a role in the observed heterotypic immune protection against severe COVID-19.
Collapse
Affiliation(s)
- David J. Bean
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine; Boston, MA
| | - Yan Mei Liang
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine; Boston, MA
| | - Manish Sagar
- Department of Virology, Immunology and Microbiology, Boston University Chobanian & Avedisian School of Medicine; Boston, MA
- Department of Medicine, Boston University Chobanian & Avedisian School of Medicine; Boston, MA
| |
Collapse
|
28
|
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
|
29
|
Im KI, Kim N, Lee J, Oh UH, Lee HW, Lee DG, Min GJ, Lee R, Lee J, Kim S, Cho SG. SARS-CoV-2-Specific T-Cell as a Potent Therapeutic Strategy against Immune Evasion of Emerging COVID-19 Variants. Int J Mol Sci 2024; 25:10512. [PMID: 39408840 PMCID: PMC11477143 DOI: 10.3390/ijms251910512] [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: 09/05/2024] [Revised: 09/24/2024] [Accepted: 09/26/2024] [Indexed: 10/20/2024] Open
Abstract
Despite advances in vaccination and therapies for coronavirus disease, challenges remain due to reduced antibody longevity and the emergence of virulent variants like Omicron (BA.1) and its subvariants (BA.1.1, BA.2, BA.3, and BA.5). This study explored the potential of adoptive immunotherapy and harnessing the protective abilities using virus-specific T cells (VSTs). Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) VSTs were generated by stimulating donor-derived peripheral blood mononuclear cells with spike, nucleocapsid, and membrane protein peptide mixtures. Phenotypic characterization, including T-cell receptor (TCR) vβ and pentamer analyses, was performed on the ex vivo-expanded cells. We infected human leukocyte antigen (HLA)-partially matched human Calu-3 cells with various authentic SARS-CoV-2 strains in a Biosafety Level 3 facility and co-cultured them with VSTs. VSTs exhibited a diverse TCR vβ repertoire, confirming their ability to target a broad range of SARS-CoV-2 antigens from both the ancestral and mutant strains, including Omicron BA.1 and BA.5. These ex vivo-expanded cells exhibited robust cytotoxicity and low alloreactivity against HLA-partially matched SARS-CoV-2-infected cells. Their cytotoxic effects were consistent across variants, targeting conserved spike and nucleocapsid epitopes. Our findings suggest that third-party partial HLA-matching VSTs could counter immune-escape mechanisms posed by emerging variants of concern.
Collapse
Affiliation(s)
- Keon-Il Im
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Nayoun Kim
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Junseok Lee
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
| | - Ui-Hyeon Oh
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Hye-Won Lee
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
| | - Dong-Gun Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.-G.L.); (R.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Gi-June Min
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Department of Hematology, Seoul St. Mary’s Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Raeseok Lee
- Division of Infectious Diseases, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.-G.L.); (R.L.)
- Vaccine Bio Research Institute, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jinah Lee
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea; (J.L.); (S.K.)
| | - Seungtaek Kim
- Zoonotic Virus Laboratory, Institut Pasteur Korea, Seongnam 13488, Republic of Korea; (J.L.); (S.K.)
| | - Seok-Goo Cho
- Institute for Translational Research and Molecular Imaging, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (K.-I.I.); (N.K.); (J.L.); (G.-J.M.)
- Research and Development Division, LucasBio Co., Ltd., Seoul 06591, Republic of Korea; (U.-H.O.); (H.-W.L.)
- Department of Hematology, Seoul St. Mary’s Hematology Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
30
|
Rappuoli R, Alter G, Pulendran B. Transforming vaccinology. Cell 2024; 187:5171-5194. [PMID: 39303685 PMCID: PMC11736809 DOI: 10.1016/j.cell.2024.07.021] [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: 01/24/2024] [Revised: 06/24/2024] [Accepted: 07/12/2024] [Indexed: 09/22/2024]
Abstract
The COVID-19 pandemic placed the field of vaccinology squarely at the center of global consciousness, emphasizing the vital role of vaccines as transformative public health tools. The impact of vaccines was recently acknowledged by the award of the 2023 Nobel Prize in Physiology or Medicine to Katalin Kariko and Drew Weissman for their seminal contributions to the development of mRNA vaccines. Here, we provide a historic perspective on the key innovations that led to the development of some 27 licensed vaccines over the past two centuries and recent advances that promise to transform vaccines in the future. Technological revolutions such as reverse vaccinology, synthetic biology, and structure-based design transformed decades of vaccine failures into successful vaccines against meningococcus B and respiratory syncytial virus (RSV). Likewise, the speed and flexibility of mRNA vaccines profoundly altered vaccine development, and the advancement of novel adjuvants promises to revolutionize our ability to tune immunity. Here, we highlight exciting new advances in the field of systems immunology that are transforming our mechanistic understanding of the human immune response to vaccines and how to predict and manipulate them. Additionally, we discuss major immunological challenges such as learning how to stimulate durable protective immune response in humans.
Collapse
Affiliation(s)
| | - Galit Alter
- Moderna Therapeutics, Cambridge, MA 02139, USA.
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
31
|
Li Q, Zhou X, Wang W, Xu Q, Wang Q, Li J. Structural basis of rosmarinic acid inhibitory mechanism on SARS-CoV-2 main protease. Biochem Biophys Res Commun 2024; 724:150230. [PMID: 38865813 DOI: 10.1016/j.bbrc.2024.150230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
The SARS-CoV-2 coronavirus is characterized by high mutation rates and significant infectivity, posing ongoing challenges for therapeutic intervention. To address potential challenges in the future, the continued development of effective drugs targeting SARS-CoV-2 remains an important task for the scientific as well as the pharmaceutical community. The main protease (Mpro) of SARS-CoV-2 is an ideal therapeutic target for COVID-19 drug development, leading to the introduction of various inhibitors, both covalent and non-covalent, each characterized by unique mechanisms of action and possessing inherent strengths and limitations. Natural products, being compounds naturally present in the environment, offer advantages such as low toxicity and diverse activities, presenting a viable source for antiviral drug development. Here, we identified a natural compound, rosmarinic acid, which exhibits significant inhibitory effects on the Mpro of the SARS-CoV-2. Through detailed structural biology analysis, we elucidated the precise crystal structure of the complex formed between rosmarinic acid and SARS-CoV-2 Mpro, revealing the molecular basis of its inhibitory mechanism. These findings not only enhance our understanding of the antiviral action of rosmarinic acid, but also provide valuable structural information and mechanistic insights for the further development of therapeutic strategies against SARS-CoV-2.
Collapse
Affiliation(s)
- Qianhui Li
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China; Chinese Academy of Sciences, Beijing, 100049, China
| | - Xuelan Zhou
- College of Pharmaceutical Sciences, Gannan Medical University, Ganzhou, 341000, China
| | - Weiwei Wang
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, China
| | - Qin Xu
- Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, China
| | - Qisheng Wang
- Shanghai Institute of Applied Physics, Chinese Academy of Sciences, Shanghai, 201800, China; Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201204, China.
| | - Jian Li
- College of Pharmaceutical Sciences, Gannan Medical University, Ganzhou, 341000, China.
| |
Collapse
|
32
|
Ma S, Wei D, Hu W, Xi M, Zhang Y, Chen X, Chen J. A case report of prolonged viral shedding of SARS-CoV-2 in a patient who receive ibrutinib for CLL therapy. BMC Infect Dis 2024; 24:895. [PMID: 39218877 PMCID: PMC11367913 DOI: 10.1186/s12879-024-09794-z] [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: 12/10/2023] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
Patients on B cell immunosuppressive treatments have been shown to have persistent infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this report, a woman treated with ibrutinib for chronic lymphocytic leukemia experienced more than 40 days of coronavirus disease 2019 (COVID-19) infection. Unexpectedly, her peripheral blood experiments showed a normal SARS-CoV-2-specific antibody level and a relatively elevated percentage of CD19 + B cells, while an obvious decrease in the percentages of NK cells, CD4 + T cells and CD8 + T cells. Further SARS-CoV-2-specific T cell analysis in this patient indicated a significant decrease in the percentage of SARS-CoV-2-specific IFN-γ, TNF-α or IL-2 producing CD4 + T or CD8 + T cells. Most notably, ten days after the cease of ibrutinib, the PCR for SARS-CoV-2 turned negative and the reduced proportions of peripheral CD4 + T cells and CD8 + T cells recovered. Our research predicted that the depleted B-cell function therapies may play considerable role in the development of long COVID-19 and the abnormal T-cell subset distribution might be the underlying mechanism.
Collapse
Affiliation(s)
- Siyuan Ma
- Department of Infectious Diseases, Shanghai Sixth People's HospitalAffiliated to, Shanghai Jiao Tong University School of Medicine , Shanghai, 200233, China
| | - Dong Wei
- Department of Infectious Diseases, Research Laboratory of Clinical Virology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
- Shanghai Key Laboratory of Emergency Prevention, Diagnosis and Treatment of Respiratory Infectious Diseases, Shanghai, People's Republic of China
| | - Weiwei Hu
- Department of Infectious Diseases, Shanghai Sixth People's HospitalAffiliated to, Shanghai Jiao Tong University School of Medicine , Shanghai, 200233, China
| | - Min Xi
- Department of Infectious Diseases, Shanghai Sixth People's HospitalAffiliated to, Shanghai Jiao Tong University School of Medicine , Shanghai, 200233, China
| | - Yi Zhang
- Department of Infectious Diseases, Shanghai Sixth People's HospitalAffiliated to, Shanghai Jiao Tong University School of Medicine , Shanghai, 200233, China
| | - Xiaohua Chen
- Department of Infectious Diseases, Shanghai Sixth People's HospitalAffiliated to, Shanghai Jiao Tong University School of Medicine , Shanghai, 200233, China.
| | - Jie Chen
- Department of Infectious Diseases, Shanghai Sixth People's HospitalAffiliated to, Shanghai Jiao Tong University School of Medicine , Shanghai, 200233, China.
| |
Collapse
|
33
|
Markov NS, Ren Z, Senkow KJ, Grant RA, Gao CA, Malsin ES, Sichizya L, Kihshen H, Helmin KA, Jovisic M, Arnold JM, Pérez-Leonor XG, Abdala-Valencia H, Swaminathan S, Nwaezeapu J, Kang M, Rasmussen L, Ozer EA, Lorenzo-Redondo R, Hultquist JF, Simons LM, Rios-Guzman E, Misharin AV, Wunderink RG, Budinger GRS, Singer BD, Morales-Nebreda L. Distinctive evolution of alveolar T cell responses is associated with clinical outcomes in unvaccinated patients with SARS-CoV-2 pneumonia. Nat Immunol 2024; 25:1607-1622. [PMID: 39138384 PMCID: PMC11490290 DOI: 10.1038/s41590-024-01914-w] [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: 01/15/2024] [Accepted: 07/03/2024] [Indexed: 08/15/2024]
Abstract
The evolution of T cell molecular signatures in the distal lung of patients with severe pneumonia is understudied. Here, we analyzed T cell subsets in longitudinal bronchoalveolar lavage fluid samples from 273 patients with severe pneumonia, including unvaccinated patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or with respiratory failure not linked to pneumonia. In patients with SARS-CoV-2 pneumonia, activation of interferon signaling pathways, low activation of the NF-κB pathway and preferential targeting of spike and nucleocapsid proteins early after intubation were associated with favorable outcomes, whereas loss of interferon signaling, activation of NF-κB-driven programs and specificity for the ORF1ab complex late in disease were associated with mortality. These results suggest that in patients with severe SARS-CoV-2 pneumonia, alveolar T cell interferon responses targeting structural SARS-CoV-2 proteins characterize individuals who recover, whereas responses against nonstructural proteins and activation of NF-κB are associated with poor outcomes.
Collapse
Affiliation(s)
- Nikolay S Markov
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ziyou Ren
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Dermatology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Karolina J Senkow
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Rogan A Grant
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Catherine A Gao
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elizabeth S Malsin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lango Sichizya
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hermon Kihshen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Kathryn A Helmin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Milica Jovisic
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jason M Arnold
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Xóchitl G Pérez-Leonor
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Hiam Abdala-Valencia
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Suchitra Swaminathan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Julu Nwaezeapu
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mengjia Kang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Luke Rasmussen
- Division of Health and Biomedical Informatics, Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Egon A Ozer
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ramon Lorenzo-Redondo
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Judd F Hultquist
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lacy M Simons
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Estefany Rios-Guzman
- Division of Infectious Diseases, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Center for Pathogen Genomics and Microbial Evolution, Institute for Global Health, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Alexander V Misharin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Richard G Wunderink
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G R Scott Budinger
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Benjamin D Singer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
- Simpson Querrey Institute for Epigenetics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Luisa Morales-Nebreda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
- Simpson Querrey Lung Institute for Translational Science, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
34
|
Benhamouda N, Besbes A, Bauer R, Mabrouk N, Gadouas G, Desaint C, Chevrier L, Lefebvre M, Radenne A, Roelens M, Parfait B, Weiskopf D, Sette A, Gruel N, Courbebaisse M, Appay V, Paul S, Gorochov G, Ropers J, Lebbah S, Lelievre JD, Johannes L, Ulmer J, Lebeaux D, Friedlander G, De Lamballerie X, Ravel P, Kieny MP, Batteux F, Durier C, Launay O, Tartour E. Cytokine profile of anti-spike CD4 +T cells predicts humoral and CD8 +T cell responses after anti-SARS-CoV-2 mRNA vaccination. iScience 2024; 27:110441. [PMID: 39104410 PMCID: PMC11298648 DOI: 10.1016/j.isci.2024.110441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/31/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
Coordinating immune responses - humoral and cellular - is vital for protection against severe Covid-19. Our study evaluates a multicytokine CD4+T cell signature's predictive for post-vaccinal serological and CD8+T cell responses. A cytokine signature composed of four cytokines (IL-2, TNF-α, IP10, IL-9) excluding IFN-γ, and generated through machine learning, effectively predicted the CD8+T cell response following mRNA-1273 or BNT162b2 vaccine administration. Its applicability extends to murine vaccination models, encompassing diverse immunization routes (such as intranasal) and vaccine platforms (including adjuvanted proteins). Notably, we found correlation between CD4+T lymphocyte-produced IL-21 and the humoral response. Consequently, we propose a test that offers a rapid overview of integrated immune responses. This approach holds particular relevance for scenarios involving immunocompromised patients because they often have low cell counts (lymphopenia) or pandemics. This study also underscores the pivotal role of CD4+T cells during a vaccine response and highlights their value in vaccine immunomonitoring.
Collapse
Affiliation(s)
- Nadine Benhamouda
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Anissa Besbes
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | | | - Nesrine Mabrouk
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Gauthier Gadouas
- Bioinformatics and Cancer System Biology Team, IRCM-INSERM U1194, Institut de Recherche en Cancerologie de Montpellier, Montpellier, France
| | - Corinne Desaint
- INSERM SC10-US019, Villejuif, France
- Université Paris Cité, INSERM, CIC 1417, F-CRIN, Innovative Clinical Research Network in Vaccinology (I-REIVAC), APHP, CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Lucie Chevrier
- Université Paris Cité, INSERM U1016 Insitut Cochin, Hôpital Cochin, APHP, Centre Service d’immunologie Biologique, Paris, France
| | - Maeva Lefebvre
- Service de maladies infectieuses et tropicales, Centre de prévention des maladies infectieuses et transmissibles CHU de Nantes, Nantes, France
| | - Anne Radenne
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière-Hôpitaux Universitaires Pitié Salpêtrière - Charles Foix, APHP, Paris, France
| | - Marie Roelens
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| | - Béatrice Parfait
- Centre de ressources Biologiques, Hôpital Cochin, APHP, Paris, France
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, School of Medicine in Health Sciences, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA, USA
| | - Nadège Gruel
- INSERM U830, Équipe Labellisée Ligue Nationale Contre le Cancer, Diversity and Plasticity of Childhood Tumors Lab, Centre de Recherche, Institut Curie, Université PSL, Paris, France
- Department of Translational Research, Centre de Recherche, Institut Curie, Université PSL, Paris, France
| | - Marie Courbebaisse
- Faculté de Médecine, Université Paris Cité, Paris, France
- Explorations fonctionnelles rénales, Physiologie, Hôpital Européen Georges-Pompidou, APHP, Paris, France
| | - Victor Appay
- Université de Bordeaux, CNRS UMR 5164, INSERM ERL 1303, ImmunoConcEpT, 33000 Bordeaux, France
- International Research Center of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Stephane Paul
- Centre International de Recherche en Infectiologie, Team GIMAP, Université Jean Monnet, Université Claude Bernard Lyon, INSERM, CIC 1408 INSERM Vaccinology, Immunology Department, iBiothera Reference Center, University Hospital of Saint-Etienne, Saint-Etienne, France
| | - Guy Gorochov
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses, APHP, Hôpital Pitié-Salpêtrière, Paris, France
| | - Jacques Ropers
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière –Hôpitaux Universitaires Pitié Salpêtrière- Charles Foix, APHP, Paris, France
| | - Said Lebbah
- Unité de Recherche Clinique des Hôpitaux Universitaires Pitié Salpêtrière –Hôpitaux Universitaires Pitié Salpêtrière- Charles Foix, APHP, Paris, France
| | - Jean-Daniel Lelievre
- Vaccine Research Institute, Créteil, France
- INSERM U955, Université Paris-Est Créteil, Créteil, France
- Groupe Henri-Mondor Albert-Chenevier, APHP, Créteil, France
| | - Ludger Johannes
- Cellular and Chemical Biology Unit, U1143 INSERM, UMR3666 CNRS, Institut Curie, Centre de Recherche, Université PSL, Paris, France
| | - Jonathan Ulmer
- Cellular and Chemical Biology Unit, U1143 INSERM, UMR3666 CNRS, Institut Curie, Centre de Recherche, Université PSL, Paris, France
| | - David Lebeaux
- Université Paris Cité, Service de maladies infectieuses Hôpital Saint Louis/Lariboisère APHP, INSERM, Paris, France
| | - Gerard Friedlander
- Department of « Croissance et Signalisation », Institut Necker Enfants Malades, INSERM U1151, CNRS UMR 8253, Université de Paris Cité, Paris, France
| | - Xavier De Lamballerie
- Unité des Virus Émergents, UVE: Aix-Marseille Université, IRD 190, INSERM 1207 Marseille, France
| | - Patrice Ravel
- Bioinformatics and Cancer System Biology Team, IRCM-INSERM U1194, Institut de Recherche en Cancerologie de Montpellier, Montpellier, France
| | - Marie Paule Kieny
- Institut National de la Santé et de la Recherche Médicale, INSERM, Paris, France
| | - Fréderic Batteux
- Université Paris Cité, INSERM U1016 Insitut Cochin, Hôpital Cochin, APHP, Centre Service d’immunologie Biologique, Paris, France
| | | | - Odile Launay
- Université Paris Cité, INSERM, CIC 1417, F-CRIN, Innovative Clinical Research Network in Vaccinology (I-REIVAC), APHP, CIC Cochin Pasteur, Hôpital Cochin, Paris, France
| | - Eric Tartour
- Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
- Université Paris Cité, INSERM U970, PARCC, Department of Immunology, Hôpital Européen Georges-Pompidou, Hôpital Necker Department of Immunology, Paris, France
| |
Collapse
|
35
|
Kenny G, Saini G, Gaillard CM, Negi R, Alalwan D, Garcia Leon A, McCann K, Tinago W, Kelly C, Cotter AG, de Barra E, Horgan M, Yousif O, Gautier V, Landay A, McAuley D, Feeney ER, O'Kane C, Mallon PWG. Early inflammatory profiles predict maximal disease severity in COVID-19: An unsupervised cluster analysis. Heliyon 2024; 10:e34694. [PMID: 39144942 PMCID: PMC11320140 DOI: 10.1016/j.heliyon.2024.e34694] [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: 03/14/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024] Open
Abstract
Background The inflammatory changes that underlie the heterogeneous presentations of COVID-19 remain incompletely understood. In this study we aimed to identify inflammatory profiles that precede the development of severe COVID-19, that could serve as targets for optimised delivery of immunomodulatory therapies and provide insights for the development of new therapies. Methods We included individuals sampled <10 days from COVID-19 symptom onset, recruited from both inpatient and outpatient settings. We measured 61 biomarkers in plasma, including markers of innate immune and T cell activation, coagulation, tissue repair and lung injury. We used principal component analysis and hierarchical clustering to derive biomarker clusters, and ordinal logistic regression to explore associations between cluster membership and maximal disease severity, adjusting for known risk factors for severe COVID-19. Results In 312 individuals, median (IQR) 7 (4-9) days from symptom onset, we found four clusters. Cluster 1 was characterised by low overall inflammation, cluster 2 was characterised by higher levels of growth factors and markers of endothelial activation (EGF, VEGF, PDGF, TGFα, PAI-1 and p-selectin). Cluster 3 and 4 both had higher overall inflammation. Cluster 4 had the highest levels of most markers including markers of innate immune activation (IL6, procalcitonin, CRP, TNFα), and coagulation (D-dimer, TPO), in contrast cluster 3 had the highest levels of alveolar epithelial injury markers (RAGE, ST2), but relative downregulation of growth factors and endothelial activation markers, suggesting a dysfunctional inflammatory pattern. In unadjusted and adjusted analysis, compared to cluster 1, cluster 3 had the highest odds of progressing to more severe disease (unadjusted OR (95%CI) 9.02 (4.53-17.96), adjusted OR (95%CI) 6.02 (2.70-13.39)). Conclusion Early inflammatory profiles predicted subsequent maximal disease severity independent of risk factors for severe COVID-19. A cluster with downregulation of growth factors and endothelial activation markers, and early evidence of alveolar epithelial injury, had the highest risk of severe COVID-19.
Collapse
Affiliation(s)
- Grace Kenny
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Gurvin Saini
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Colette Marie Gaillard
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Riya Negi
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Dana Alalwan
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Alejandro Garcia Leon
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Kathleen McCann
- Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland
| | - Willard Tinago
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Christine Kelly
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Aoife G. Cotter
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Eoghan de Barra
- Department of International Health and Tropical Medicine, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mary Horgan
- Department of Infectious Diseases, Cork University Hospital, Wilton, Cork, Ireland
| | - Obada Yousif
- Department of Endocrinology, Wexford General Hospital, Wexford, Ireland
| | - Virginie Gautier
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
| | - Alan Landay
- Department of Internal Medicine, Rush University, Chicago, IL, USA
| | | | - Eoin R. Feeney
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland
| | | | - Patrick WG. Mallon
- Centre for Experimental Pathogen Host Research, University College Dublin, Dublin, Ireland
- Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland
| |
Collapse
|
36
|
Fischer K, Lulla A, So TY, Pereyra-Gerber P, Raybould MIJ, Kohler TN, Yam-Puc JC, Kaminski TS, Hughes R, Pyeatt GL, Leiss-Maier F, Brear P, Matheson NJ, Deane CM, Hyvönen M, Thaventhiran JED, Hollfelder F. Rapid discovery of monoclonal antibodies by microfluidics-enabled FACS of single pathogen-specific antibody-secreting cells. Nat Biotechnol 2024:10.1038/s41587-024-02346-5. [PMID: 39143416 DOI: 10.1038/s41587-024-02346-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 06/27/2024] [Indexed: 08/16/2024]
Abstract
Monoclonal antibodies are increasingly used to prevent and treat viral infections and are pivotal in pandemic response efforts. Antibody-secreting cells (ASCs; plasma cells and plasmablasts) are an excellent source of high-affinity antibodies with therapeutic potential. Current methods to study antigen-specific ASCs either have low throughput, require expensive and labor-intensive screening or are technically demanding and therefore not widely accessible. Here we present a straightforward technology for the rapid discovery of monoclonal antibodies from ASCs. Our approach combines microfluidic encapsulation of single cells into an antibody capture hydrogel with antigen bait sorting by conventional flow cytometry. With our technology, we screened millions of mouse and human ASCs and obtained monoclonal antibodies against severe acute respiratory syndrome coronavirus 2 with high affinity (<1 pM) and neutralizing capacity (<100 ng ml-1) in 2 weeks with a high hit rate (>85% of characterized antibodies bound the target). By facilitating access to the underexplored ASC compartment, the approach enables efficient antibody discovery and immunological studies into the generation of protective antibodies.
Collapse
Affiliation(s)
- Katrin Fischer
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Aleksei Lulla
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Tsz Y So
- MRC Toxicology Unit, Gleeson Building, Cambridge, UK
| | - Pehuén Pereyra-Gerber
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
| | - Matthew I J Raybould
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford, UK
| | - Timo N Kohler
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | - Tomasz S Kaminski
- Department of Biochemistry, University of Cambridge, Cambridge, UK
- Department of Molecular Biology, Institute of Biochemistry, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Robert Hughes
- MRC Toxicology Unit, Gleeson Building, Cambridge, UK
| | | | | | - Paul Brear
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Nicholas J Matheson
- Cambridge Institute for Therapeutic Immunology and Infectious Disease (CITIID), Department of Medicine, University of Cambridge, Cambridge, UK
- NHS Blood and Transplant, Cambridge, UK
| | - Charlotte M Deane
- Oxford Protein Informatics Group, Department of Statistics, University of Oxford, Oxford, UK
| | - Marko Hyvönen
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | | | |
Collapse
|
37
|
Grosso D, Wagner JL, O’Connor A, Keck K, Huang Y, Wang ZX, Mehler H, Leiby B, Flomenberg P, Gergis U, Nikbakht N, Morris M, Karp J, Peedin A, Flomenberg N. Safety and feasibility of third-party cytotoxic T lymphocytes for high-risk patients with COVID-19. Blood Adv 2024; 8:4113-4124. [PMID: 38885482 PMCID: PMC11345373 DOI: 10.1182/bloodadvances.2024013344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 05/24/2024] [Accepted: 06/12/2024] [Indexed: 06/20/2024] Open
Abstract
ABSTRACT Cytotoxic T lymphocytes (CTLs) destroy virally infected cells and are critical for the elimination of viral infections such as those caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Delayed and dysfunctional adaptive immune responses to SARS-CoV-2 are associated with poor outcomes. Treatment with allogeneic SARS-CoV-2-specific CTLs may enhance cellular immunity in high-risk patients providing a safe, direct mechanism of treatment. Thirty high-risk ambulatory patients with COVID-19 were enrolled in a phase 1 trial assessing the safety of third party, SARS-CoV-2-specific CTLs. Twelve interventional patients, 6 of whom were immunocompromised, matched the HLA-A∗02:01 restriction of the CTLs and received a single infusion of 1 of 4 escalating doses of a product containing 68.5% SARS-CoV-2-specific CD8+ CTLs/total cells. Symptom improvement and resolution in these patients was compared with an observational group of 18 patients lacking HLA-A∗02:01 who could receive standard of care. No dose-limiting toxicities were observed at any dosing level. Nasal swab polymerase chain reaction testing showed ≥88% and >99% viral elimination from baseline in all patients at 4 and 14 days after infusion, respectively. The CTLs did not interfere with the development of endogenous anti-SARS-CoV-2 humoral or cellular responses. T-cell receptor β analysis showed persistence of donor-derived SARS-CoV-2-specific CTLs through the end of the 6-month follow-up period. Interventional patients consistently reported symptomatic improvement 2 to 3 days after infusion, whereas improvement was more variable in observational patients. SARS-CoV-2-specific CTLs are a potentially feasible cellular therapy for COVID-19 illness. This trial was registered at www.clinicaltrials.gov as #NCT04765449.
Collapse
Affiliation(s)
- Dolores Grosso
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - John L. Wagner
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Allyson O’Connor
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Kaitlyn Keck
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Yanping Huang
- Department of Pathology and Genomic Medicine, Histocompatibility and Immunogenetics Laboratory, Thomas Jefferson University, Philadelphia, PA
| | - Zi-Xuan Wang
- Departments of Surgery and Pathology, Molecular and Genomic Pathology Laboratory, Thomas Jefferson University, Philadelphia, PA
| | - Hilary Mehler
- Department of Pathology and Genomic Medicine, Histocompatibility and Immunogenetics Laboratory, Thomas Jefferson University, Philadelphia, PA
| | - Benjamin Leiby
- Division of Biostatistics, Department of Pharmacology, Physiology, and Cancer Biology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Phyllis Flomenberg
- Division of Infectious Diseases, Department of Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Usama Gergis
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| | - Neda Nikbakht
- Department of Dermatology and Cutaneous Biology, Cutaneous Lymphoma Clinic, Thomas Jefferson University, Philadelphia, PA
| | - Michael Morris
- Department of Emergency Medicine, Thomas Jefferson University Washington Township Hospital, Sewell, NJ
| | - Julie Karp
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Alexis Peedin
- Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA
| | - Neal Flomenberg
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
38
|
Gregory TA, Knight SR, Aaroe AE, Highsmith KN, Janatpour ZC, O’Brien BJ, Majd NK, Loghin ME, Patel CB, Weathers SP, Puduvalli VK, Kamiya-Matsuoka C. Accelerated tumor progression after COVID-19 infection in patients with glioblastoma: A retrospective case-control study. Neurooncol Pract 2024; 11:475-483. [PMID: 39006516 PMCID: PMC11241387 DOI: 10.1093/nop/npae029] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024] Open
Abstract
Background We observed rapid tumor progression following COVID-19 infection among patients with glioblastoma and sought to systematically characterize their disease course in a retrospective case-control study. Methods Using an institutional database, we retrospectively identified a series of COVID-19-positive glioblastoma cases and matched them by age and sex 1:2 to glioblastoma controls who had a negative COVID-19 test during their disease course. Demographic and clinical data were analyzed. Hyperprogression was defined using modified response evaluation criteria in solid tumors criteria. Time to progression and overall survival were estimated using the Kaplan-Meier method. Results Thirty-two glioblastoma cases with positive COVID-19 testing were matched to 64 glioblastoma controls with negative testing; age, sex, and molecular profiles did not differ between groups. Progression events occurred in 27 cases (84%) and 46 controls (72%). Of these, 14 cases (52%) presented with multifocal disease or leptomeningeal disease at progression compared with 10 controls (22%; P = .0082). Hyperprogression was identified in 13 cases (48%) but only 4 controls (9%; P = .0001). Cases had disease progression at a median of 35 days following COVID-19 testing, compared with 164 days for controls (P = .0001). Median survival from COVID-19 testing until death was 8.3 months for cases but 17 months for controls (P = .0016). Median overall survival from glioblastoma diagnosis was 20.7 months for cases and 24.6 months for controls (P = .672). Conclusions Patients with glioblastoma may have accelerated disease progression in the first 2 months after COVID-19 infection. Infected patients should be monitored vigilantly. Future investigations should explore tumor-immune microenvironment changes linking tumor progression and COVID-19.
Collapse
Affiliation(s)
- Timothy A Gregory
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
- Department of Neurology, Madigan Army Medical Center, Tacoma, Washington, USA
| | - Stephanie R Knight
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Ashley E Aaroe
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Kaitlin N Highsmith
- Department of Neuro-Oncology, Pharmacy Clinical Programs, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Zachary C Janatpour
- Department of Neurology, Madigan Army Medical Center, Tacoma, Washington, USA
| | - Barbara J O’Brien
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Nazanin K Majd
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Monica E Loghin
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Chirag B Patel
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Shiao-Pei Weathers
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Vinay K Puduvalli
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Carlos Kamiya-Matsuoka
- Department of Neuro-Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
39
|
Hurler L, Mescia F, Bergamaschi L, Kajdácsi E, Sinkovits G, Cervenak L, Prohászka Z, Lyons PA, Toonen EJ. sMR and PTX3 levels associate with COVID-19 outcome and survival but not with Long COVID. iScience 2024; 27:110162. [PMID: 39027374 PMCID: PMC11255846 DOI: 10.1016/j.isci.2024.110162] [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: 12/06/2023] [Revised: 02/28/2024] [Accepted: 05/29/2024] [Indexed: 07/20/2024] Open
Abstract
Biomarkers for monitoring COVID-19 disease course are lacking. Study aim was to identify biomarkers associated with disease severity, survival, long-term outcome, and Long COVID. As excessive macrophages activation is a hallmark of COVID-19 and complement activation is key in this, we selected the following proteins involved in these processes: PTX3, C1q, C1-INH, C1s/C1-INH, and sMR. EDTA-plasma concentrations were measured in 215 patients and 47 controls using ELISA. PTX3, sMR, C1-INH, and C1s/C1-INH levels were associated with disease severity. PTX3 and sMR were also associated with survival and long-term immune recovery. Lastly, sMR levels associate with ICU admittance. sMR (AUC 0.85) and PTX3 (AUC 0.78) are good markers for disease severity, especially when used in combination (AUC 0.88). No association between biomarker levels and Long COVID was observed. sMR has not previously been associated with COVID-19 disease severity, ICU admittance or survival and may serve as marker for disease course.
Collapse
Affiliation(s)
- Lisa Hurler
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - Federica Mescia
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Cambridge Institute of Therapeutic Immunology and Infectious Disease-National Institute of Health Research (CITIID-NIHR) COVID BioResource Collaboration
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
- Research Group for Immunology and Haematology, Semmelweis University - Eötvös Loránd Research Network (Office for Supported Research Groups), Budapest, Hungary
- Research and Development Department, Hycult Biotech, Uden, the Netherlands
| | - Erika Kajdácsi
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - György Sinkovits
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - László Cervenak
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
| | - Zoltán Prohászka
- Department of Internal Medicine and Haematology, Semmelweis University, Budapest, Hungary
- Research Group for Immunology and Haematology, Semmelweis University - Eötvös Loránd Research Network (Office for Supported Research Groups), Budapest, Hungary
| | - Paul A. Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge CB2 0AW, UK
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge CB2 0QQ, UK
| | - Erik J.M. Toonen
- Research and Development Department, Hycult Biotech, Uden, the Netherlands
| |
Collapse
|
40
|
Li C, Qian W, Wei X, Narasimhan H, Wu Y, Arish M, Cheon IS, Tang J, de Almeida Santos G, Li Y, Sharifi K, Kern R, Vassallo R, Sun J. Comparative single-cell analysis reveals IFN-γ as a driver of respiratory sequelae after acute COVID-19. Sci Transl Med 2024; 16:eadn0136. [PMID: 39018367 DOI: 10.1126/scitranslmed.adn0136] [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: 11/17/2023] [Revised: 03/12/2024] [Accepted: 06/10/2024] [Indexed: 07/19/2024]
Abstract
Postacute sequelae of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection (PASC) represent an urgent public health challenge and are estimated to affect more than 60 million individuals globally. Although a growing body of evidence suggests that dysregulated immune reactions may be linked with PASC symptoms, most investigations have primarily centered around blood-based studies, with few focusing on samples derived from affected tissues. Furthermore, clinical studies alone often provide correlative insights rather than causal mechanisms. Thus, it is essential to compare clinical samples with relevant animal models and conduct functional experiments to understand the etiology of PASC. In this study, we comprehensively compared bronchoalveolar lavage fluid single-cell RNA sequencing data derived from clinical PASC samples and a mouse model of PASC. This revealed a pro-fibrotic monocyte-derived macrophage response in respiratory PASC, as well as abnormal interactions between pulmonary macrophages and respiratory resident T cells, in both humans and mice. Interferon-γ (IFN-γ) emerged as a key node mediating the immune anomalies in respiratory PASC. Neutralizing IFN-γ after the resolution of acute SARS-CoV-2 infection reduced lung inflammation and tissue fibrosis in mice. Together, our study underscores the importance of performing comparative analysis to understand the cause of PASC and suggests that the IFN-γ signaling axis might represent a therapeutic target.
Collapse
Affiliation(s)
- Chaofan Li
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Wei Qian
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Xiaoqin Wei
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Harish Narasimhan
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| | - Yue Wu
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Mohd Arish
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - In Su Cheon
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Jinyi Tang
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Gislane de Almeida Santos
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Ying Li
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Kamyar Sharifi
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
| | - Ryan Kern
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Robert Vassallo
- Division of Pulmonary and Critical Medicine, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jie Sun
- Beirne B. Carter Center for Immunology Research, University of Virginia, Charlottesville, VA 22908, USA
- Division of Infectious Disease and International Health, Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
41
|
Li J, Zhou J, Tan Y, Hu C, Meng Q, Gao J, Xing L. Clinical characteristics and risk factors for mortality in pneumonia-associated acute respiratory distress syndrome patients: a single center retrospective cohort study. Front Cell Infect Microbiol 2024; 14:1396088. [PMID: 39045130 PMCID: PMC11263095 DOI: 10.3389/fcimb.2024.1396088] [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: 03/05/2024] [Accepted: 06/20/2024] [Indexed: 07/25/2024] Open
Abstract
Background Pathogenic diversity may have contributed to the high mortality of pneumonia-associated acute respiratory distress syndrome (p-ARDS). Metagenomics next-generation sequencing (mNGS) serves as a valuable diagnostic tool for early pathogen identification. However, its clinical utility in p-ARDS remains understudied. There are still limited researches on the etiology, clinical characteristics and risk factors for 28-day mortality in p-ARDS patients. Methods A single center retrospective cohort study of 75 p-ARDS patients was conducted. Patients were categorized into survival and deceased groups based on their 28-day outcomes. A comprehensive clinical evaluation was conducted, including baseline characteristics, laboratory indicators, outcomes and pathogen identification by mNGS and traditional microbiological testing. We then evaluated the diagnostic value of mNGS and identified clinical characteristics and risk factors for 28-day mortality in p-ARDS. Result The overall ICU mortality was 26.67%, and the 28-day mortality was 57.33%, with 32 cases (42.67%) in the survival group, and 43 cases (57.33%) in the deceased group. Patients in the deceased group were older than those in the survival group (68(59,73) years vs. 59(44,67) years, P=0.04). The average lengths of ICU and hospital stay were 9(5,13) days and 14(7,21) days, respectively. The survival group had longer lengths of ICU and hospital stay (ICU: 11(7,17) days and hospital: 17(9,27) days) compared to the deceased group (ICU: 8(4,11) days and hospital: 12(6,19) days) (P<0.05). Survival patients exhibited lower Acute Physiology and Chronic Health Evaluation (APACHE) II score on the 3rd and 7th days, higher lymphocyte counts, higher CD3+ and CD8+ T cell counts compared to deceased patients (P<0.05). Multivariate logistic regression analysis identified age, APACHE II scores on 3rd and 7th days, CD8+ T cell count and length of ICU as independent risk factors for 28-day mortality in p-ARDS patients. mNGS demonstrated a significantly higher overall pathogen detection rate (70/75, 93.33%) compared to the traditional method (50/75, 66.67%, P=0.022). The average turnaround time (TAT) for mNGS was significantly shorter at 1(1,1) day compared to 4(3,5) days for the traditional method (P<0.001). Conclusion Metagenome next-generation sequencing can be used as a valuable tool for identifying pathogens in p-ARDS, reducing diagnostic time and improving accuracy. Early application of mNGS alongside traditional methods is recommended for p-ARDS. Furthermore, older age, higher APACHE II scores, lower lymphocyte counts and lymphocyte subset counts were associated with increased mortality in p-ARDS patients, highlighting the importance of timely assessment of immune status and disease severity, especially in elderly.
Collapse
Affiliation(s)
| | | | | | | | | | - Jing Gao
- Department of Respiratory Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lihua Xing
- Department of Respiratory Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
42
|
Ichise H, Speranza E, La Russa F, Veres TZ, Chu CJ, Gola A, Germain RN. Rebalancing Viral and Immune Damage versus Tissue Repair Prevents Death from Lethal Influenza Infection. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.04.601620. [PMID: 39372755 PMCID: PMC11451654 DOI: 10.1101/2024.07.04.601620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Maintaining tissue function while eliminating infected cells is fundamental to host defense. Innate inflammatory damage contributes to lethal influenza and COVID-19, yet other than steroids, immunomodulatory drugs have modest effects. Among more than 50 immunomodulatory regimes tested in mouse lethal influenza infection, only the previously reported early depletion of neutrophils showed efficacy, suggesting that the infected host passes an early tipping point in which limiting innate immune damage alone cannot rescue physiological function. To re-balance the system late in infection, we investigated whether partial limitation of viral spread using oseltamivir (Tamiflu) together with enhancement of epithelial repair by blockade of interferon signaling or the limitation of further epithelial cell loss mediated by cytotoxic CD8 + T cells would prevent death. These treatments salvaged a large fraction of infected animals, providing new insight into the importance of repair processes and the timing of adaptive immune responses in survival of pulmonary infections.
Collapse
|
43
|
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
|
44
|
Hernandez-Galicia G, Gomez-Morales L, Lopez-Bailon LU, Valdovinos-Torres H, Contreras-Ochoa CO, Díaz Benítez CE, Martinez-Barnetche J, Alpuche-Aranda C, Ortiz-Navarrete V. Presence of SARS-CoV-2-specific T cells before vaccination in the Mexican population. J Leukoc Biol 2024; 116:95-102. [PMID: 38717738 DOI: 10.1093/jleuko/qiae111] [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: 08/01/2023] [Revised: 04/04/2024] [Accepted: 04/17/2024] [Indexed: 06/30/2024] Open
Abstract
The immune response to SARS-CoV-2 has been extensively studied following the pandemic outbreak in 2020; however, the presence of specific T cells against SARS-CoV-2 before vaccination has not been evaluated in Mexico. In this study, we estimated the frequency of T CD4+ and T CD8+ cells that exhibit a specific response to S (spike) and N (nucleocapsid) proteins in a Mexican population. We collected 78 peripheral blood samples from unvaccinated subjects, and the presence of antibodies against spike (RBD) and N protein was determined. Peripheral blood mononuclear cells were isolated and stimulated with a pool of S or N protein peptides (Wuhan-Hu-1 strain). IL-1β, IL-4, IL-6, IL-10, IL-2, IL-8, TNF-α, IFN-γ, and GM-CSF levels were quantified in the supernatant of the activated cells, and the cells were stained to assess the activation and memory phenotypes. Differential activation frequency dependent on serological status was observed in CD4+ cells but not in CD8+ cells. The predominantly activated population was the central memory T CD4+ cells. Only 10% of the population exhibited the same phenotype with respect to the response to nucleocapsid peptides. The cytokine profile differed between the S and N responses. S peptides induced a more proinflammatory response compared with the N peptides. In conclusion, in a Mexican cohort before vaccination, there was a significant response to the S and N SARS-CoV-2 proteins resulting from previous infections with seasonal coronaviruses or previous undetected exposure to SARS-CoV-2.
Collapse
Affiliation(s)
- Gabriela Hernandez-Galicia
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
| | - Luis Gomez-Morales
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
- Department of Immunology, National School of Biological Sciences, National Polytechnic Institute, Manuel Carpio and Plan de Ayala St, 11340, Mexico City, Mexico
| | - Luis Uriel Lopez-Bailon
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
- Department of Immunology, National School of Biological Sciences, National Polytechnic Institute, Manuel Carpio and Plan de Ayala St, 11340, Mexico City, Mexico
| | - Humberto Valdovinos-Torres
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Carla O Contreras-Ochoa
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Cinthya Estefhany Díaz Benítez
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Jesus Martinez-Barnetche
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Celia Alpuche-Aranda
- Center for Infectious Diseases Research, National Institute of Public Health, 655 Universidad Avenue, 62100, Cuernavaca, Mexico
| | - Vianney Ortiz-Navarrete
- Department of Molecular Biomedicine, Center for Research and Advanced Studies of the National Polytechnic Institute, 2508 Instituto Politécnico Nacional Avenue, 07360, Mexico City, Mexico
| |
Collapse
|
45
|
Theel ES, Kirby JE, Pollock NR. Testing for SARS-CoV-2: lessons learned and current use cases. Clin Microbiol Rev 2024; 37:e0007223. [PMID: 38488364 PMCID: PMC11237512 DOI: 10.1128/cmr.00072-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024] Open
Abstract
SUMMARYThe emergence and worldwide dissemination of SARS-CoV-2 required both urgent development of new diagnostic tests and expansion of diagnostic testing capacity on an unprecedented scale. The rapid evolution of technologies that allowed testing to move out of traditional laboratories and into point-of-care testing centers and the home transformed the diagnostic landscape. Four years later, with the end of the formal public health emergency but continued global circulation of the virus, it is important to take a fresh look at available SARS-CoV-2 testing technologies and consider how they should be used going forward. This review considers current use case scenarios for SARS-CoV-2 antigen, nucleic acid amplification, and immunologic tests, incorporating the latest evidence for analytical/clinical performance characteristics and advantages/limitations for each test type to inform current debates about how tests should or should not be used.
Collapse
Affiliation(s)
- Elitza S. Theel
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - James E. Kirby
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Nira R. Pollock
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Laboratory Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
46
|
Rakebrandt N, Yassini N, Kolz A, Schorer M, Lambert K, Goljat E, Estrada Brull A, Rauld C, Balazs Z, Krauthammer M, Carballido JM, Peters A, Joller N. Innate acting memory Th1 cells modulate heterologous diseases. Proc Natl Acad Sci U S A 2024; 121:e2312837121. [PMID: 38838013 PMCID: PMC11181110 DOI: 10.1073/pnas.2312837121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 05/08/2024] [Indexed: 06/07/2024] Open
Abstract
Through immune memory, infections have a lasting effect on the host. While memory cells enable accelerated and enhanced responses upon rechallenge with the same pathogen, their impact on susceptibility to unrelated diseases is unclear. We identify a subset of memory T helper 1 (Th1) cells termed innate acting memory T (TIA) cells that originate from a viral infection and produce IFN-γ with innate kinetics upon heterologous challenge in vivo. Activation of memory TIA cells is induced in response to IL-12 in combination with IL-18 or IL-33 but is TCR independent. Rapid IFN-γ production by memory TIA cells is protective in subsequent heterologous challenge with the bacterial pathogen Legionella pneumophila. In contrast, antigen-independent reactivation of CD4+ memory TIA cells accelerates disease onset in an autoimmune model of multiple sclerosis. Our findings demonstrate that memory Th1 cells can acquire additional TCR-independent functionality to mount rapid, innate-like responses that modulate susceptibility to heterologous challenges.
Collapse
Affiliation(s)
- Nikolas Rakebrandt
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Nima Yassini
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Anna Kolz
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
| | - Michelle Schorer
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Katharina Lambert
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
| | - Eva Goljat
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Anna Estrada Brull
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Celine Rauld
- Novartis Biomedical Research, 4002Basel, Switzerland
| | - Zsolt Balazs
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | - Michael Krauthammer
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| | | | - Anneli Peters
- Institute of Clinical Neuroimmunology, University Hospital, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität München, 82152Planegg, Germany
| | - Nicole Joller
- Institute of Experimental Immunology, University of Zurich, 8057Zurich, Switzerland
- Department of Quantitative Biomedicine, University of Zurich, 8057Zurich, Switzerland
| |
Collapse
|
47
|
Milentijević M, Katanić N, Joksimović B, Pavlović A, Filimonović J, Anđelković M, Bojović K, Elek Z, Ristić S, Vasiljević M, Stevanović J, Radomirović D, Elez-Burnjaković N, Lalović N, Kulić M, Kulić J, Milić M. The Impact of Cytokines on Coagulation Profile in COVID-19 Patients: Controlled for Socio-Demographic, Clinical, and Laboratory Parameters. Biomedicines 2024; 12:1281. [PMID: 38927488 PMCID: PMC11201770 DOI: 10.3390/biomedicines12061281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024] Open
Abstract
Background: Severe coagulation abnormalities are common in patients with COVID-19 infection. We aimed to investigate the relationship between pro-inflammatory cytokines and coagulation parameters concerning socio-demographic, clinical, and laboratory characteristics. Methods: Our study included patients hospitalized during the second wave of COVID-19 in the Republic of Serbia. We collected socio-demographic, clinical, and blood-sample data for all patients. Cytokine levels were measured using flow cytometry. Results: We analyzed data from 113 COVID-19 patients with an average age of 58.15 years, of whom 79 (69.9%) were male. Longer duration of COVID-19 symptoms before hospitalization (B = 69.672; p = 0.002) and use of meropenem (B = 1237.220; p = 0.014) were predictive of higher D-dimer values. Among cytokines, higher IL-5 values significantly predicted higher INR values (B = 0.152; p = 0.040) and longer prothrombin times (B = 0.412; p = 0.043), and higher IL-6 (B = 0.137; p = 0.003) predicted longer prothrombin times. Lower IL-17F concentrations at admission (B = 0.024; p = 0.050) were predictive of higher INR values, and lower IFN-γ values (B = -0.306; p = 0.017) were predictive of higher aPTT values. Conclusions: Our findings indicate a significant correlation between pro-inflammatory cytokines and coagulation-related parameters. Factors such as the patient's level of education, gender, oxygen-therapy use, symptom duration before hospitalization, meropenem use, and serum concentrations of IL-5, IL-6, IL-17F, and IFN-γ were associated with worse coagulation-related parameters.
Collapse
Affiliation(s)
- Milica Milentijević
- Department of Infective Diseases, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.M.); (N.K.)
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
| | - Nataša Katanić
- Department of Infective Diseases, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.M.); (N.K.)
| | - Bojan Joksimović
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Aleksandar Pavlović
- Department of Surgery, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia;
| | - Jelena Filimonović
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (J.F.); (J.S.)
| | - Milena Anđelković
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
| | - Ksenija Bojović
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Zlatan Elek
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
- Department of Surgery, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia;
| | - Siniša Ristić
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Miloš Vasiljević
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Jasmina Stevanović
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (J.F.); (J.S.)
| | - Danica Radomirović
- Clinical Hospital Center Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (M.A.); (Z.E.); (D.R.)
| | - Nikolina Elez-Burnjaković
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Nenad Lalović
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Milan Kulić
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Jovan Kulić
- Faculty of Medicine Foča, University of East Sarajevo, 73300 Foča, Republic of Srpska, Bosnia and Herzegovina; (K.B.); (S.R.); (M.V.); (N.E.-B.); (N.L.); (M.K.); (J.K.)
| | - Marija Milić
- Department of Epidemiology, Faculty of Medicine, University of Pristina Temporarily Settled in Kosovska Mitrovica, 38220 Kosovska Mitrovica, Serbia; (J.F.); (J.S.)
| |
Collapse
|
48
|
Sbierski-Kind J, Schlickeiser S, Feldmann S, Ober V, Grüner E, Pleimelding C, Gilberg L, Brand I, Weigl N, Ahmed MIM, Ibarra G, Ruzicka M, Benesch C, Pernpruner A, Valdinoci E, Hoelscher M, Adorjan K, Stubbe HC, Pritsch M, Seybold U, Roider J. Persistent immune abnormalities discriminate post-COVID syndrome from convalescence. Infection 2024; 52:1087-1097. [PMID: 38326527 PMCID: PMC11142964 DOI: 10.1007/s15010-023-02164-y] [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: 11/13/2023] [Accepted: 12/19/2023] [Indexed: 02/09/2024]
Abstract
BACKGROUND Innate lymphoid cells (ILCs) are key organizers of tissue immune responses and regulate tissue development, repair, and pathology. Persistent clinical sequelae beyond 12 weeks following acute COVID-19 disease, named post-COVID syndrome (PCS), are increasingly recognized in convalescent individuals. ILCs have been associated with the severity of COVID-19 symptoms but their role in the development of PCS remains poorly defined. METHODS AND RESULTS Here, we used multiparametric immune phenotyping, finding expanded circulating ILC precursors (ILCPs) and concurrent decreased group 2 innate lymphoid cells (ILC2s) in PCS patients compared to well-matched convalescent control groups at > 3 months after infection or healthy controls. Patients with PCS showed elevated expression of chemokines and cytokines associated with trafficking of immune cells (CCL19/MIP-3b, FLT3-ligand), endothelial inflammation and repair (CXCL1, EGF, RANTES, IL-1RA, PDGF-AA). CONCLUSION These results define immunological parameters associated with PCS and might help find biomarkers and disease-relevant therapeutic strategies.
Collapse
Affiliation(s)
- Julia Sbierski-Kind
- Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Internal Medicine IV, Division of Diabetology, Endocrinology and Nephrology, University Hospital, Eberhard-Karls-Universität Tübingen, Tübingen, Germany
- The M3 Research Center, University Clinic Tübingen (UKT), Medical Faculty, Otfried-Müllerstr. 37, Tübingen, Germany
| | - Stephan Schlickeiser
- Charité, Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt- Universität Zu Berlin, Institute of Medical Immunology, Augustenburger Platz 1, 13353, Berlin, Germany
- Berlin Institute of Health (BIH) at Charité, Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Charitéplatz 1, 10117, Berlin, Germany
| | - Svenja Feldmann
- Department of Infectious Diseases, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Veronica Ober
- Department of Infectious Diseases, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Eva Grüner
- Department of Infectious Diseases, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Claire Pleimelding
- Department of Infectious Diseases, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Leonard Gilberg
- Department of Infectious Diseases, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Isabel Brand
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Nikolas Weigl
- Department of Medicine IV, Division of Clinical Pharmacology, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Mohamed I M Ahmed
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Gerardo Ibarra
- The M3 Research Center, University Clinic Tübingen (UKT), Medical Faculty, Otfried-Müllerstr. 37, Tübingen, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Ruzicka
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Christopher Benesch
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Medicine II, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Anna Pernpruner
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Medicine II, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Elisabeth Valdinoci
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Medicine II, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Hoelscher
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Kristina Adorjan
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Psychiatry and Psychotherapy, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Hans Christian Stubbe
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
- Department of Medicine II, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Michael Pritsch
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- Division of Infectious Diseases and Tropical Medicine, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ulrich Seybold
- Department of Infectious Diseases, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Julia Roider
- Department of Infectious Diseases, Department of Medicine IV, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany.
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany.
| |
Collapse
|
49
|
Niehaus C, Klein S, Strunz B, Freyer E, Maasoumy B, Wedemeyer H, Björkström NK, Kraft ARM, Cornberg M. CXCR6 +CD69 + CD8 + T cells in ascites are associated with disease severity in patients with cirrhosis. JHEP Rep 2024; 6:101074. [PMID: 38882602 PMCID: PMC11179582 DOI: 10.1016/j.jhepr.2024.101074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 03/16/2024] [Accepted: 03/20/2024] [Indexed: 06/18/2024] Open
Abstract
Background & Aims Patients with advanced cirrhosis often develop hepatic decompensation, which is accompanied by systemic inflammation and may eventually lead to acute-on-chronic liver failure. One important cause of systemic hyperinflammation is a dysregulated overshooting immune response in ascites in the abdominal cavity. In this study, we analyzed the role of CD8+ T cells in the ascites immune compartment. Methods Peripheral blood and ascites fluid were collected from 50 patients with decompensated cirrhosis. Phenotype and functional responses of CD8+ T cells were analyzed, and obtained data were compared with each other as well as with healthy controls and patients with compensated cirrhosis. Results High-dimensional flow cytometry revealed that CD8+ T cells are abundant in the ascites of patients with cirrhosis and exhibit a chronically activated bystander phenotype with innate-like functions. Indeed, we identified distinct CXCR6+CD69+ clusters of late effector memory CD8+ T cells that were rarely found in blood and correlated with clinical parameters of disease severity. Moreover, this CD8+ T-cell population was hyperresponsive to innate cytokines and exhibited cytokine-mediated bystander activation. Interestingly, the Janus kinase (JAK) inhibitor tofacitinib was able to effectively block bystander-activated CXCR6+CD69+ CD8+ T cells and significantly suppress effector molecule production. Conclusions The results indicate that CXCR6+CD69+ CD8+ T cells in ascites are associated with disease severity and may contribute to inflammation in patients with decompensated cirrhosis, suggesting that targeted inhibition of this immune cell subset may be a viable therapeutic option. Impact and Implications Patients with advanced cirrhosis often develop hepatic decompensation, which is accompanied by systemic inflammation and eventually leads to acute-on-chronic liver failure. One important cause of systemic hyperinflammation is a dysregulated overshooting immune response in ascites in the abdominal cavity. In this study, we demonstrate that CXCR6+CD69+ CD8+ T cells are abundant in the ascites of patients with cirrhosis, exhibit a chronically activated bystander phenotype, and correlate with clinical parameters of disease severity. Moreover, we show that the Janus kinase (JAK) inhibitor tofacitinib can effectively block these bystander-activated CXCR6+CD69+ CD8+ T cells, suggesting that targeted inhibition of this immune cell subset may be a potential therapeutic strategy. Clinical trial number Prospective registry: INFEKTA (DRKS00010664).
Collapse
Affiliation(s)
- Christian Niehaus
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Sebastian Klein
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany
- CAIMed - Center for AI in Medicine, Joint Venture of Leibniz University Hannover and Hannover Medical School, Hannover, Germany
| | - Benedikt Strunz
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Erich Freyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany
| | - Benjamin Maasoumy
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany
| | - Heiner Wedemeyer
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Niklas K Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Anke R M Kraft
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Markus Cornberg
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School, Hannover, Germany
- Centre for Individualised Infection Medicine (CiiM), a joint venture between the Helmholtz Centre for Infection Research (HZI) and Hannover Medical School (MHH), Hannover, Germany
- Twincore, Centre for Experimental and Clinical Infection Research, a joint venture between the Helmholtz Centre for Infection Research (HZI) and the Hannover Medical School, Hannover, Germany
- German Center for Infection Research (DZIF), Partner-site Hannover-Braunschweig, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- German Center for Infection Research, HepNet Study-House German Liver Foundation, Hannover, Germany
| |
Collapse
|
50
|
Wilk P, Stranges S, Cuschieri S. Does sex modify the effect of pre-pandemic body mass index on the risk of Long COVID? Evidence from the longitudinal analysis of the Survey of Health, Ageing and Retirement in Europe. Int J Obes (Lond) 2024; 48:821-829. [PMID: 38287094 DOI: 10.1038/s41366-024-01477-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 01/31/2024]
Abstract
BACKGROUND Research on Long COVID risk factors is ongoing. High body mass index (BMI) may increase Long COVID risk, yet no evidence has been established regarding sex differences in the relationship between BMI and the risk of Long COVID. Investigating the nature of this relationship was the main objective of this study. METHODS A population-based prospective study involving a sample of respondents aged 50 years and older (n = 4004) from 27 European countries that participated in the 2020 and 2021 Survey of Health, Ageing and Retirement in Europe's (SHARE) Corona Surveys and in Waves 7 and 8 of the main SHARE survey. Logistic regression models were estimated to produce unadjusted and adjusted estimates of the sex differences in the relationship between BMI and Long COVID. RESULTS Linear relationship for females, with probability of Long COVID increasing with BMI (68% at BMI = 18, 93% at BMI = 45). Non-linear relationship for males, with probability of Long COVID of 27% at BMI = 18, 68% at BMI = 33, and 40% at BMI = 45. Relationships remained significant after adjusting for known Long COVID risk factors (age and COVID-19 hospitalization), presence of chronic diseases, and respondents' place of residence. CONCLUSION Sex differences appear to play an important role in the relationship between BMI and risk of Long COVID. Overall, females were more likely to have Long COVID, regardless of their BMI. Males at the higher end of the BMI spectrum had a lower risk of Long COVID as opposed to their female counterparts. Sex-specific research is recommended for better understanding of Long COVID risk factors.
Collapse
Affiliation(s)
- Piotr Wilk
- Department of Epidemiology and Biostatistics, Western University, London, ON, Canada
- Department of Epidemiology, Maastricht University, Maastricht, the Netherlands
| | - Saverio Stranges
- Department of Epidemiology and Biostatistics, Western University, London, ON, Canada
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
- Department of Behavioural and Cognitive Sciences, University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Sarah Cuschieri
- Department of Epidemiology and Biostatistics, Western University, London, ON, Canada.
- Faculty of Medicine and Surgery, University of Malta, Msida, Malta.
| |
Collapse
|