201
|
Fischer DS, Ansari M, Wagner KI, Jarosch S, Huang Y, Mayr CH, Strunz M, Lang NJ, D'Ippolito E, Hammel M, Mateyka L, Weber S, Wolff LS, Witter K, Fernandez IE, Leuschner G, Milger K, Frankenberger M, Nowak L, Heinig-Menhard K, Koch I, Stoleriu MG, Hilgendorff A, Behr J, Pichlmair A, Schubert B, Theis FJ, Busch DH, Schiller HB, Schober K. Single-cell RNA sequencing reveals ex vivo signatures of SARS-CoV-2-reactive T cells through 'reverse phenotyping'. Nat Commun 2021; 12:4515. [PMID: 34312385 PMCID: PMC8313584 DOI: 10.1038/s41467-021-24730-4] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 06/16/2021] [Indexed: 02/07/2023] Open
Abstract
The in vivo phenotypic profile of T cells reactive to severe acute respiratory syndrome (SARS)-CoV-2 antigens remains poorly understood. Conventional methods to detect antigen-reactive T cells require in vitro antigenic re-stimulation or highly individualized peptide-human leukocyte antigen (pHLA) multimers. Here, we use single-cell RNA sequencing to identify and profile SARS-CoV-2-reactive T cells from Coronavirus Disease 2019 (COVID-19) patients. To do so, we induce transcriptional shifts by antigenic stimulation in vitro and take advantage of natural T cell receptor (TCR) sequences of clonally expanded T cells as barcodes for 'reverse phenotyping'. This allows identification of SARS-CoV-2-reactive TCRs and reveals phenotypic effects introduced by antigen-specific stimulation. We characterize transcriptional signatures of currently and previously activated SARS-CoV-2-reactive T cells, and show correspondence with phenotypes of T cells from the respiratory tract of patients with severe disease in the presence or absence of virus in independent cohorts. Reverse phenotyping is a powerful tool to provide an integrated insight into cellular states of SARS-CoV-2-reactive T cells across tissues and activation states.
Collapse
Affiliation(s)
- David S Fischer
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, München, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Meshal Ansari
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, München, Germany
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Karolin I Wagner
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Sebastian Jarosch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Yiqi Huang
- Institute of Virology, Technische Universität München (TUM), Munich, Germany
| | - Christoph H Mayr
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Maximilian Strunz
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Niklas J Lang
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Elvira D'Ippolito
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Monika Hammel
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Laura Mateyka
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Simone Weber
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
| | - Lisa S Wolff
- Institute of Virology, Technische Universität München (TUM), Munich, Germany
| | - Klaus Witter
- Laboratory of Immunogenetics and Molecular Diagnostics, Department of Transfusion Medicine, Cell Therapeutic Agents and Hemostaseology, LMU Munich, Munich, Germany
- Department of Medicine V, University Hospital, LMU Munich, Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for lung research (DZL), Munich, Germany
| | - Isis E Fernandez
- Department of Medicine V, University Hospital, LMU Munich, Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for lung research (DZL), Munich, Germany
| | - Gabriela Leuschner
- Department of Medicine V, University Hospital, LMU Munich, Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for lung research (DZL), Munich, Germany
| | - Katrin Milger
- Department of Medicine V, University Hospital, LMU Munich, Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for lung research (DZL), Munich, Germany
| | - Marion Frankenberger
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich (LMU) and Asklepios Lung Clinic Munich-Gauting, Munich and Gauting, Munich, Germany
| | - Lorenz Nowak
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich (LMU) and Asklepios Lung Clinic Munich-Gauting, Munich and Gauting, Munich, Germany
| | - Katharina Heinig-Menhard
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich (LMU) and Asklepios Lung Clinic Munich-Gauting, Munich and Gauting, Munich, Germany
| | - Ina Koch
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
- Asklepios Biobank for pulmonary diseases, Gauting, Germany
- Member of the German Center for Lung Research (DZL), Center for Comprehensive Developmental Care (CDeCLMU), Department of Neonatology, Perinatal Center, Munich, Germany
| | - Mircea G Stoleriu
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
- Asklepios Biobank for pulmonary diseases, Gauting, Germany
- Member of the German Center for Lung Research (DZL), Center for Comprehensive Developmental Care (CDeCLMU), Department of Neonatology, Perinatal Center, Munich, Germany
| | - Anne Hilgendorff
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany
- German Center for Infection Research (DZIF), partner site Munich, Munich, Germany
| | - Jürgen Behr
- Department of Medicine V, University Hospital, LMU Munich, Comprehensive Pneumology Center Munich (CPC-M), Member of the German Center for lung research (DZL), Munich, Germany
- Center for Thoracic Surgery Munich, Ludwig-Maximilians-University of Munich (LMU) and Asklepios Lung Clinic Munich-Gauting, Munich and Gauting, Munich, Germany
| | - Andreas Pichlmair
- Institute of Virology, Technische Universität München (TUM), Munich, Germany
- Department of Mathematics, Technical University of Munich, Garching, Germany
| | - Benjamin Schubert
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, München, Germany
- Focus Group 'Clinical Cell Processing and Purification", Institute for Advanced Study, TUM, Munich, Germany
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, Neuherberg, München, Germany
- Focus Group 'Clinical Cell Processing and Purification", Institute for Advanced Study, TUM, Munich, Germany
| | - Dirk H Busch
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany
- Department of Mathematics, Technical University of Munich, Garching, Germany
- Grosshadern, Hospital of the Ludwig-Maximilians University (LMU), Munich, Germany
| | - Herbert B Schiller
- Institute of Lung Biology and Disease and Comprehensive Pneumology Center with the CPC-M bioArchive, Helmholtz Zentrum Muenchen, Member of the German Center for Lung Research (DZL), Munich, Germany.
- Institute of Lung Biology and Disease, Comprehensive Pneumology Center, Helmholtz Zentrum München, Neuherberg, München, Germany.
| | - Kilian Schober
- Institute for Medical Microbiology, Immunology and Hygiene, Technische Universität München (TUM), Munich, Germany.
- Microbiological Institute-Institute of Clinical Microbiology, Immunology and Hygiene, University Hospital of Erlangen, Erlangen, Germany.
| |
Collapse
|
202
|
Budinger GS, Misharin AV, Ridge KM, Singer BD, Wunderink RG. Distinctive features of severe SARS-CoV-2 pneumonia. J Clin Invest 2021; 131:149412. [PMID: 34263736 PMCID: PMC8279580 DOI: 10.1172/jci149412] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic is among the most important public health crises of our generation. Despite the promise of prevention offered by effective vaccines, patients with severe COVID-19 will continue to populate hospitals and intensive care units for the foreseeable future. The most common clinical presentation of severe COVID-19 is hypoxemia and respiratory failure, typical of the acute respiratory distress syndrome (ARDS). Whether the clinical features and pathobiology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pneumonia differ from those of pneumonia secondary to other pathogens is unclear. This uncertainty has created variability in the application of historically proven therapies for ARDS to patients with COVID-19. We review the available literature and find many similarities between patients with ARDS from pneumonia attributable to SARS-CoV-2 versus other respiratory pathogens. A notable exception is the long duration of illness among patients with COVID-19, which could result from its unique pathobiology. Available data support the use of care pathways and therapies proven effective for patients with ARDS, while pointing to unique features that might be therapeutically targeted for patients with severe SARS-CoV-2 pneumonia.
Collapse
|
203
|
Grifoni A, Sidney J, Vita R, Peters B, Crotty S, Weiskopf D, Sette A. SARS-CoV-2 human T cell epitopes: Adaptive immune response against COVID-19. Cell Host Microbe 2021; 29:1076-1092. [PMID: 34237248 PMCID: PMC8139264 DOI: 10.1016/j.chom.2021.05.010] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/23/2021] [Accepted: 05/18/2021] [Indexed: 02/07/2023]
Abstract
Over the past year, numerous studies in the peer reviewed and preprint literature have reported on the virological, epidemiological and clinical characteristics of the coronavirus, SARS-CoV-2. To date, 25 studies have investigated and identified SARS-CoV-2-derived T cell epitopes in humans. Here, we review these recent studies, how they were performed, and their findings. We review how epitopes identified throughout the SARS-CoV2 proteome reveal significant correlation between number of epitopes defined and size of the antigen provenance. We also report additional analysis of SARS-CoV-2 human CD4 and CD8 T cell epitope data compiled from these studies, identifying 1,400 different reported SARS-CoV-2 epitopes and revealing discrete immunodominant regions of the virus and epitopes that are more prevalently recognized. This remarkable breadth of epitope repertoire has implications for vaccine design, cross-reactivity, and immune escape by SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - John Sidney
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Randi Vita
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Bjoern Peters
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA; Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA.
| |
Collapse
|
204
|
Sidhom JW, Baras AS. Deep learning identifies antigenic determinants of severe SARS-CoV-2 infection within T-cell repertoires. Sci Rep 2021; 11:14275. [PMID: 34253751 PMCID: PMC8275616 DOI: 10.1038/s41598-021-93608-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/24/2021] [Indexed: 12/28/2022] Open
Abstract
SARS-CoV-2 infection is characterized by a highly variable clinical course with patients experiencing asymptomatic infection all the way to requiring critical care support. This variation in clinical course has led physicians and scientists to study factors that may predispose certain individuals to more severe clinical presentations in hopes of either identifying these individuals early in their illness or improving their medical management. We sought to understand immunogenomic differences that may result in varied clinical outcomes through analysis of T-cell receptor sequencing (TCR-Seq) data in the open access ImmuneCODE database. We identified two cohorts within the database that had clinical outcomes data reflecting severity of illness and utilized DeepTCR, a multiple-instance deep learning repertoire classifier, to predict patients with severe SARS-CoV-2 infection from their repertoire sequencing. We demonstrate that patients with severe infection have repertoires with higher T-cell responses associated with SARS-CoV-2 epitopes and identify the epitopes that result in these responses. Our results provide evidence that the highly variable clinical course seen in SARS-CoV-2 infection is associated to certain antigen-specific responses.
Collapse
Affiliation(s)
- John-William Sidhom
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Alexander S Baras
- Bloomberg Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| |
Collapse
|
205
|
da Silva Antunes R, Pallikkuth S, Williams E, Dawen Yu E, Mateus J, Quiambao L, Wang E, Rawlings SA, Stadlbauer D, Jiang K, Amanat F, Arnold D, Andrews D, Fuego I, Dan JM, Grifoni A, Weiskopf D, Krammer F, Crotty S, Hoffer ME, Pahwa SG, Sette A. Differential T-Cell Reactivity to Endemic Coronaviruses and SARS-CoV-2 in Community and Health Care Workers. J Infect Dis 2021; 224:70-80. [PMID: 33822097 PMCID: PMC8083569 DOI: 10.1093/infdis/jiab176] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 03/30/2021] [Indexed: 02/06/2023] Open
Abstract
Herein we measured CD4+ T-cell responses against common cold coronaviruses (CCC) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in high-risk health care workers (HCW) and community controls. We observed higher levels of CCC-reactive T cells in SARS-CoV-2-seronegative HCW compared to community donors, consistent with potential higher occupational exposure of HCW to CCC. We further show that SARS-CoV-2 T-cell reactivity of seronegative HCW was higher than community controls and correlation between CCC and SARS-CoV-2 responses is consistent with cross-reactivity and not associated with recent in vivo activation. Surprisingly, CCC T-cell reactivity was decreased in SARS-CoV-2-infected HCW, suggesting that exposure to SARS-CoV-2 might interfere with CCC responses, either directly or indirectly. This result was unexpected, but consistently detected in independent cohorts derived from Miami and San Diego. CD4+ T-cell responses against common cold coronaviruses (CCC) are elevated in SARS-CoV-2 seronegative high-risk health care workers (HCW) compared to COVID-19 convalescent HCW, suggesting that exposure to SARS-CoV-2 might interfere with CCC responses and/or cross-reactivity associated with a protective effect.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Erin Williams
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Esther Dawen Yu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Jose Mateus
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Lorenzo Quiambao
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Eric Wang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Stephen A Rawlings
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, California, USA
| | - Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kaijun Jiang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - David Arnold
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - David Andrews
- Department of Pathology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Irma Fuego
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Jennifer M Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, California, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, California, USA
| | - Michael E Hoffer
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami, Florida, USA
- Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Savita G Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, California, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
206
|
Jung MK, Shin EC. Phenotypes and Functions of SARS-CoV-2-Reactive T Cells. Mol Cells 2021; 44:401-407. [PMID: 34120892 PMCID: PMC8245315 DOI: 10.14348/molcells.2021.0079] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/09/2021] [Accepted: 05/12/2021] [Indexed: 12/27/2022] Open
Abstract
Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), which is an ongoing pandemic disease. SARS-CoV-2-specific CD4+ and CD8+ T-cell responses have been detected and characterized not only in COVID-19 patients and convalescents, but also unexposed individuals. Here, we review the phenotypes and functions of SARS-CoV-2-specific T cells in COVID-19 patients and the relationships between SARS-CoV-2-specific T-cell responses and COVID-19 severity. In addition, we describe the phenotypes and functions of SARS-CoV-2-specific memory T cells after recovery from COVID-19 and discuss the presence of SARS-CoV-2-reactive T cells in unexposed individuals and SARS-CoV-2-specific T-cell responses elicited by COVID-19 vaccines. A better understanding of T-cell responses is important for effective control of the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Min Kyung Jung
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Eui-Cheol Shin
- Laboratory of Immunology and Infectious Diseases, Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- The Center for Epidemic Preparedness, KAIST, Daejeon 34141, Korea
| |
Collapse
|
207
|
Hassan SS, Attrish D, Ghosh S, Choudhury PP, Uversky VN, Aljabali AAA, Lundstrom K, Uhal BD, Rezaei N, Seyran M, Pizzol D, Adadi P, Soares A, Abd El-Aziz TM, Kandimalla R, Tambuwala MM, Azad GK, Sherchan SP, Baetas-da-Cruz W, Lal A, Palù G, Takayama K, Serrano-Aroca Á, Barh D, Brufsky AM. Notable sequence homology of the ORF10 protein introspects the architecture of SARS-CoV-2. Int J Biol Macromol 2021; 181:801-809. [PMID: 33862077 PMCID: PMC8051021 DOI: 10.1016/j.ijbiomac.2021.03.199] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 03/30/2021] [Accepted: 03/30/2021] [Indexed: 01/01/2023]
Abstract
The current Coronavirus Disease 19 (COVID-19) pandemic, caused by Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) shows similar pathology to MERS and SARS-CoV, with a current estimated fatality rate of 1.4%. Open reading frame 10 (ORF10) is a unique SARS-CoV-2 accessory protein, which contains eleven cytotoxic T lymphocyte (CTL) epitopes each of nine amino acids in length. Twenty-two unique SARS-CoV-2 ORF10 variants have been identified based on missense mutations found in sequence databases. Some of these mutations are predicted to decrease the stability of ORF10 in silico physicochemical and structural comparative analyses were carried out on SARS-CoV-2 and Pangolin-CoV ORF10 proteins, which share 97.37% amino acid (aa) homology. Though there is a high degree of ORF10 protein similarity of SARS-CoV-2 and Pangolin-CoV, there are differences of these two ORF10 proteins related to their sub-structure (loop/coil region), solubility, antigenicity and shift from strand to coil at aa position 26 (tyrosine). SARS-CoV-2 ORF10, which is apparently expressed in vivo since reactive T cell clones are found in convalescent patients should be monitored for changes which could correlate with the pathogenesis of COVID-19.
Collapse
Affiliation(s)
- Sk Sarif Hassan
- Department of Mathematics, Pingla Thana Mahavidyalaya, Maligram 721140, India.
| | - Diksha Attrish
- Dr. B. R. Ambedkar Centre For Biomedical Research (ACBR), University of Delhi (North Campus), Delhi 110007, India
| | - Shinjini Ghosh
- Department of Biophysics, Molecular Biology and Bioinformatics, University of Calcutta, Kolkata 700009, West Bengal, India
| | - Pabitra Pal Choudhury
- Applied Statistics Unit, Indian Statistical Institute, Kolkata 700108, West Bengal, India
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Alaa A A Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology, Yarmouk University-Faculty of Pharmacy, Irbid 566, Jordan.
| | | | - Bruce D Uhal
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran; Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Stockholm, Sweden.
| | - Murat Seyran
- Doctoral Studies in Natural and Technical Sciences (SPL 44), University of Vienna, Austria
| | - Damiano Pizzol
- Italian Agency for Development Cooperation - Khartoum, Sudan Street 33, Al Amarat, Sudan
| | - Parise Adadi
- Department of Food Science, University of Otago, Dunedin 9054, New Zealand.
| | - Antonio Soares
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229-3900, USA.
| | - Tarek Mohamed Abd El-Aziz
- Department of Cellular and Integrative Physiology, University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr, San Antonio, TX 78229-3900, USA; Zoology Department, Faculty of Science, Minia University, El-Minia 61519, Egypt.
| | - Ramesh Kandimalla
- CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India; Department of Biochemistry, Kakatiya Medical College, Warangal, Telangana State, India.
| | - Murtaza M Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Coleraine BT52 1SA, Northern Ireland, UK.
| | | | - Samendra P Sherchan
- Department of Environmental Health Sciences, Tulane University, New Orleans, LA 70112, USA.
| | - Wagner Baetas-da-Cruz
- Translational Laboratory in Molecular Physiology, Centre for Experimental Surgery, College of Medicine, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Amos Lal
- Division of Pulmonary and Critical Care Medicine, Mayo Clinic, Rochester, MN, USA
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy.
| | - Kazuo Takayama
- Center for iPS Cell Research and Application, Kyoto University, Kyoto 606-8397, Japan.
| | - Ángel Serrano-Aroca
- Biomaterials and Bioengineering Lab, Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia, San Vicente Mártir 46001, Valencia, Spain.
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Purba Medinipur, India
| | - Adam M Brufsky
- University of Pittsburgh School of Medicine, Department of Medicine, Division of Hematology/Oncology, UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|
208
|
Shrwani K, Sharma R, Krishnan M, Jones T, Mayora-Neto M, Cantoni D, Temperton NJ, Dobson SL, Subramaniam K, McNamara PS, Cunliffe NA, Turtle L, Zhang Q. Detection of Serum Cross-Reactive Antibodies and Memory Response to SARS-CoV-2 in Prepandemic and Post-COVID-19 Convalescent Samples. J Infect Dis 2021; 224:1305-1315. [PMID: 34161567 PMCID: PMC8557674 DOI: 10.1093/infdis/jiab333] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 06/22/2021] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND A notable feature of coronavirus disease 2019 (COVID-19) is that children are less susceptible to severe disease. Children are known to experience more infections with endemic human coronaviruses (HCoVs) compared to adults. Little is known whether HCoV infections lead to cross-reactive anti-severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies. METHODS We investigated the presence of cross-reactive anti-SARS-CoV-2 IgG antibodies to spike 1 (S1), S1-receptor-binding domain (S1-RBD), and nucleocapsid protein (NP) by enzyme-linked immunosorbent assays, and neutralizing activity by a SARS-CoV-2 pseudotyped virus neutralization assay, in prepandemic sera collected from children (n = 50) and adults (n = 45), and compared with serum samples from convalescent COVID-19 patients (n = 16). RESULTS A significant proportion of children (up to 40%) had detectable cross-reactive antibodies to SARS-CoV-2 S1, S1-RBD, and NP antigens, and the anti-S1 and anti-S1-RBD antibody levels correlated with anti-HCoV-HKU1 and anti-HCoV-OC43 S1 antibody titers in prepandemic samples (P < .001). There were marked increases of anti-HCoV-HKU1 and - OC43 S1 (but not anti-NL63 and -229E S1-RBD) antibody titers in serum samples from convalescent COVID-19 patients (P < .001), indicating an activation of cross-reactive immunological memory to β-coronavirus spike. CONCLUSIONS We demonstrated cross-reactive anti-SARS-CoV-2 antibodies in prepandemic serum samples from children and young adults. Promoting this cross-reactive immunity and memory response derived from common HCoV may be an effective strategy against SARS-COV-2 and future novel coronaviruses.
Collapse
Affiliation(s)
- Khalid Shrwani
- Department of Clinical Infection, Microbiology, and Immunology, Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Ravi Sharma
- Ear Nose and Throat Department, Alder Hey Children’s Hospital, Liverpool, United Kingdom
| | - Madhan Krishnan
- Ear Nose and Throat Department, Alder Hey Children’s Hospital, Liverpool, United Kingdom
| | - Terry Jones
- Liverpool Head and Neck Centre, University of Liverpool, Liverpool, United Kingdom
| | - Martin Mayora-Neto
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - Diego Cantoni
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - Nigel J Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - Susan L Dobson
- Department of Clinical Infection, Microbiology, and Immunology, Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Krishanthi Subramaniam
- Department of Clinical Infection, Microbiology, and Immunology, Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Paul S McNamara
- Institute of Child Health, Alder Hey Children’s Hospital, Liverpool, United Kingdom
| | - Nigel A Cunliffe
- Department of Clinical Infection, Microbiology, and Immunology, Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Lance Turtle
- Department of Clinical Infection, Microbiology, and Immunology, Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom
| | - Qibo Zhang
- Department of Clinical Infection, Microbiology, and Immunology, Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Liverpool, United Kingdom,Correspondence: Qibo Zhang, MD, PhD, Department of Clinical Infection, Microbiology, and Immunology, Institute of Infection, Veterinary, and Ecological Sciences, University of Liverpool, Ronald Ross Building, 8 West Derby Street, Liverpool L69 7BE, UK ()
| |
Collapse
|
209
|
Pan YG, Aiamkitsumrit B, Bartolo L, Wang Y, Lavery C, Marc A, Holec PV, Rappazzo CG, Eilola T, Gimotty PA, Hensley SE, Antia R, Zarnitsyna VI, Birnbaum ME, Su LF. Vaccination reshapes the virus-specific T cell repertoire in unexposed adults. Immunity 2021; 54:1245-1256.e5. [PMID: 34004140 PMCID: PMC8192456 DOI: 10.1016/j.immuni.2021.04.023] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 02/01/2021] [Accepted: 04/21/2021] [Indexed: 11/30/2022]
Abstract
We examined how baseline CD4+ T cell repertoire and precursor states impact responses to pathogen infection in humans using primary immunization with yellow fever virus (YFV) vaccine. YFV-specific T cells in unexposed individuals were identified by peptide-MHC tetramer staining and tracked pre- and post-vaccination by tetramers and TCR sequencing. A substantial number of YFV-reactive T cells expressed memory phenotype markers and contained expanded clones in the absence of exposure to YFV. After vaccination, pre-existing YFV-specific T cell populations with low clonal diversity underwent limited expansion, but rare populations with a reservoir of unexpanded TCRs generated robust responses. These altered dynamics reorganized the immunodominance hierarchy and resulted in an overall increase in higher avidity T cells. Thus, instead of further increasing the representation of dominant clones, YFV vaccination recruits rare and more responsive T cells. Our findings illustrate the impact of vaccines in prioritizing T cell responses and reveal repertoire reorganization as a key component of effective vaccination.
Collapse
Affiliation(s)
- Yi-Gen Pan
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Benjamas Aiamkitsumrit
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Laurent Bartolo
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yifeng Wang
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Criswell Lavery
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA; Corporal Michael J Crescenz VA Medical Center, Philadelphia, PA 19104, USA
| | - Adam Marc
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA; Corporal Michael J Crescenz VA Medical Center, Philadelphia, PA 19104, USA
| | - Patrick V Holec
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - C Garrett Rappazzo
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Theresa Eilola
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Phyllis A Gimotty
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rustom Antia
- Department of Biology, Emory University, Atlanta, GA, USA
| | | | - Michael E Birnbaum
- Department of Biological Engineering, Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Laura F Su
- Department of Medicine, Division of Rheumatology, Perelman School of Medicine, Institute for Immunology, University of Pennsylvania, Philadelphia, PA 19104, USA; Corporal Michael J Crescenz VA Medical Center, Philadelphia, PA 19104, USA.
| |
Collapse
|
210
|
Ringlander J, Martner A, Nilsson S, Westin J, Lindh M, Hellstrand K. Incidence and Severity of Covid-19 in Patients with and without Previously Verified Infections with Common Cold Coronaviruses. J Infect Dis 2021; 223:1831-1832. [PMID: 33780548 PMCID: PMC8083637 DOI: 10.1093/infdis/jiab089] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 02/09/2021] [Indexed: 12/18/2022] Open
Affiliation(s)
- Johan Ringlander
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Anna Martner
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Staffan Nilsson
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.,Department of Mathematical Sciences, Chalmers University of Technology, Gothenburg, Sweden
| | - Johan Westin
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Magnus Lindh
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Kristoffer Hellstrand
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
211
|
Dobaño C, Santano R, Jiménez A, Vidal M, Chi J, Rodrigo Melero N, Popovic M, López-Aladid R, Fernández-Barat L, Tortajada M, Carmona-Torre F, Reina G, Torres A, Mayor A, Carolis C, García-Basteiro AL, Aguilar R, Moncunill G, Izquierdo L. Immunogenicity and crossreactivity of antibodies to the nucleocapsid protein of SARS-CoV-2: utility and limitations in seroprevalence and immunity studies. Transl Res 2021; 232:60-74. [PMID: 33582244 PMCID: PMC7879156 DOI: 10.1016/j.trsl.2021.02.006] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 02/07/2021] [Accepted: 02/09/2021] [Indexed: 12/23/2022]
Abstract
COVID-19 patients elicit strong responses to the nucleocapsid (N) protein of SARS-CoV-2 but binding antibodies are also detected in prepandemic individuals, indicating potential crossreactivity with common cold human coronaviruses (HCoV) and questioning its utility in seroprevalence studies. We investigated the immunogenicity of the full-length and shorter fragments of the SARS-CoV-2 N protein, and the crossreactivity of antibodies with HCoV. We identified a C-terminus region in SARS-CoV2 N of minimal sequence homology with HCoV that was more specific for SARS-CoV-2 and highly immunogenic. IgGs to the full-length SARS-CoV-2 N also recognized N229E N, and IgGs to HKU1 N recognized SARS-CoV-2 N. Crossreactivity with SARS-CoV-2 was stronger for alpha- rather than beta-HCoV despite having less sequence identity, revealing the importance of conformational recognition. Higher preexisting IgG to OC43 N correlated with lower IgG to SARS-CoV-2 N in rRT-PCR negative individuals, reflecting less exposure and indicating a potential protective association. Antibodies to SARS-CoV-2 N were higher in patients with more severe and longer duration of symptoms and in females. IgGs remained stable for at least 3 months, while IgAs and IgMs declined faster. In conclusion, N protein is a primary target of SARS-CoV-2-specific and HCoV crossreactive antibodies, both of which may affect the acquisition of immunity to COVID-19.
Collapse
Key Words
- ade, antibody-dependent disease enhancement
- covid-19, coronavirus disease 2019
- ct, c-terminus
- fl, full-length
- hcov, common cold human coronavirus
- loess, locally estimated scatterplot smoothing
- m, month
- mfi, median fluorescence intensity
- n, nucleocapsid
- nt, n-terminus
- rbd, receptor-binding domain
- rrt-pcr, real-time reverse-transcriptase polymerase chain reaction
- s, spike
- sars-cov-2, severe acute respiratory syndrome coronavirus 2
Collapse
Affiliation(s)
- Carlota Dobaño
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain; Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Spain.
| | - Rebeca Santano
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Alfons Jiménez
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain; Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Spain
| | - Marta Vidal
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Jordi Chi
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Natalia Rodrigo Melero
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Matija Popovic
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Rubén López-Aladid
- Cellex Laboratory, CibeRes (Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, 06/06/0028), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain
| | - Laia Fernández-Barat
- Cellex Laboratory, CibeRes (Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, 06/06/0028), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain
| | - Marta Tortajada
- Occupational Health Department, Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Francisco Carmona-Torre
- Infectious Diseases Division and Clinical Microbiology, Clínica Universidad de Navarra, Pamplona, Spain; IdiSNA, Navarra Institute for Health Research, Spain
| | - Gabriel Reina
- Clínica Universidad de Navarra, Navarra Institute for Health Research, Pamplona, Spain
| | - Antoni Torres
- Cellex Laboratory, CibeRes (Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, 06/06/0028), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, Barcelona, Spain; Pneumology Service, Hospital Clinic, Barcelona, Spain
| | - Alfredo Mayor
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain; Spanish Consortium for Research in Epidemiology and Public Health (CIBERESP), Spain; Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Carlo Carolis
- Biomolecular Screening and Protein Technologies Unit, Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Alberto L García-Basteiro
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique; International Health Department, Hospital Clinic, Universitat de Barcelona, Barcelona, Spain
| | - Ruth Aguilar
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain.
| | - Luis Izquierdo
- ISGlobal, Hospital Clínic - Universitat de Barcelona, Barcelona, Catalonia, Spain
| |
Collapse
|
212
|
Cimolai N. Passive Immunity Should and Will Work for COVID-19 for Some Patients. Clin Hematol Int 2021; 3:47-68. [PMID: 34595467 PMCID: PMC8432400 DOI: 10.2991/chi.k.210328.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 03/02/2021] [Indexed: 12/12/2022] Open
Abstract
In the absence of effective antiviral chemotherapy and still in the context of emerging vaccines for severe acute respiratory syndrome-CoV-2 infections, passive immunotherapy remains a key treatment and possible prevention strategy. What might initially be conceived as a simplified donor-recipient process, the intricacies of donor plasma, IV immunoglobulins, and monoclonal antibody modality applications are becoming more apparent. Key targets of such treatment have largely focused on virus neutralization and the specific viral components of the attachment Spike protein and its constituents (e.g., receptor binding domain, N-terminal domain). The cumulative laboratory and clinical experience suggests that beneficial protective and treatment outcomes are possible. Both a dose- and a time-dependency emerge. Lesser understood are the concepts of bioavailability and distribution. Apart from direct antigen binding from protective immunoglobulins, antibody effector functions have potential roles in outcome. In attempting to mimic the natural but variable response to infection or vaccination, a strong functional polyclonal approach attracts the potential benefits of attacking antigen diversity, high antibody avidity, antibody persistence, and protection against escape viral mutation. The availability and ease of administration for any passive immunotherapy product must be considered in the current climate of need. There is never a perfect product, but yet there is considerable room for improving patient outcomes. Given the variability of human genetics, immunity, and disease, and given the nuances of the virus and its potential for change, passive immunotherapy can be developed that will be effective for some but not all patients. An understanding of such patient variability and limitations is just as important as the understanding of the direct interactions between immunotherapy and virus.
Collapse
Affiliation(s)
- Nevio Cimolai
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Pathology and Laboratory Medicine, Children’s and Women’s Health Centre of British Columbia, 4480 Oak Street, Vancouver, BC, Canada V6H 3V4
| |
Collapse
|
213
|
Cromer D, Juno JA, Khoury D, Reynaldi A, Wheatley AK, Kent SJ, Davenport MP. Prospects for durable immune control of SARS-CoV-2 and prevention of reinfection. Nat Rev Immunol 2021; 21:395-404. [PMID: 33927374 PMCID: PMC8082486 DOI: 10.1038/s41577-021-00550-x] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2021] [Indexed: 12/16/2022]
Abstract
Immunity to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is central to long-term control of the current pandemic. Despite our rapidly advancing knowledge of immune memory to SARS-CoV-2, understanding how these responses translate into protection against reinfection at both the individual and population levels remains a major challenge. An ideal outcome following infection or after vaccination would be a highly protective and durable immunity that allows for the establishment of high levels of population immunity. However, current studies suggest a decay of neutralizing antibody responses in convalescent patients, and documented cases of SARS-CoV-2 reinfection are increasing. Understanding the dynamics of memory responses to SARS-CoV-2 and the mechanisms of immune control are crucial for the rational design and deployment of vaccines and for understanding the possible future trajectories of the pandemic. Here, we summarize our current understanding of immune responses to and immune control of SARS-CoV-2 and the implications for prevention of reinfection.
Collapse
Affiliation(s)
- Deborah Cromer
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Jennifer A Juno
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - David Khoury
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, Australia
| | - Adam K Wheatley
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Stephen J Kent
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia.
| | | |
Collapse
|
214
|
Mazzoni A, Salvati L, Maggi L, Annunziato F, Cosmi L. Hallmarks of immune response in COVID-19: Exploring dysregulation and exhaustion. Semin Immunol 2021; 55:101508. [PMID: 34728121 PMCID: PMC8547971 DOI: 10.1016/j.smim.2021.101508] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/11/2021] [Accepted: 10/15/2021] [Indexed: 01/08/2023]
Abstract
One and half year following the occurrence of COVID-19 pandemic, significant efforts from laboratories all over the world generated a huge amount of data describing the prototypical features of immunity in the course of SARS-CoV-2 infection. In this Review, we rationalize and organize the main observations, trying to define a "core" signature of immunity in COVID-19. We identified six hallmarks describing the main alterations occurring in the early infection phase and in the course of the disease, which predispose to severe illness. The six hallmarks are dysregulated type I IFN activity, hyperinflammation, lymphopenia, lymphocyte impairment, dysregulated myeloid response, and heterogeneous adaptive immunity to SARS-CoV-2. Dysregulation and exhaustion came out as the trait d'union, connecting abnormalities affecting both innate and adaptive immunity, humoral and cellular responses.
Collapse
Affiliation(s)
- Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Lorenzo Salvati
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy.
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
215
|
Lehmann AA, Kirchenbaum GA, Zhang T, Reche PA, Lehmann PV. Deconvoluting the T Cell Response to SARS-CoV-2: Specificity Versus Chance and Cognate Cross-Reactivity. Front Immunol 2021; 12:635942. [PMID: 34127926 PMCID: PMC8196231 DOI: 10.3389/fimmu.2021.635942] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 03/11/2021] [Indexed: 01/08/2023] Open
Abstract
SARS-CoV-2 infection takes a mild or clinically inapparent course in the majority of humans who contract this virus. After such individuals have cleared the virus, only the detection of SARS-CoV-2-specific immunological memory can reveal the exposure, and hopefully the establishment of immune protection. With most viral infections, the presence of specific serum antibodies has provided a reliable biomarker for the exposure to the virus of interest. SARS-CoV-2 infection, however, does not reliably induce a durable antibody response, especially in sub-clinically infected individuals. Consequently, it is plausible for a recently infected individual to yield a false negative result within only a few months after exposure. Immunodiagnostic attention has therefore shifted to studies of specific T cell memory to SARS-CoV-2. Most reports published so far agree that a T cell response is engaged during SARS-CoV-2 infection, but they also state that in 20-81% of SARS-CoV-2-unexposed individuals, T cells respond to SARS-CoV-2 antigens (mega peptide pools), allegedly due to T cell cross-reactivity with Common Cold coronaviruses (CCC), or other antigens. Here we show that, by introducing irrelevant mega peptide pools as negative controls to account for chance cross-reactivity, and by establishing the antigen dose-response characteristic of the T cells, one can clearly discern between cognate T cell memory induced by SARS-CoV-2 infection vs. cross-reactive T cell responses in individuals who have not been infected with SARS-CoV-2.
Collapse
Affiliation(s)
- Alexander A Lehmann
- Research and Development, Cellular Technology Ltd., Shaker Heights, OH, United States
| | - Greg A Kirchenbaum
- Research and Development, Cellular Technology Ltd., Shaker Heights, OH, United States
| | - Ting Zhang
- Research and Development, Cellular Technology Ltd., Shaker Heights, OH, United States
| | - Pedro A Reche
- Laboratorio de Inmunomedicina & Inmunoinformatica, Departamento de Immunologia & O2, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - Paul V Lehmann
- Research and Development, Cellular Technology Ltd., Shaker Heights, OH, United States
| |
Collapse
|
216
|
Dykema AG, Zhang B, Woldemeskel BA, Garliss CC, Cheung LS, Choudhury D, Zhang J, Aparicio L, Bom S, Rashid R, Caushi JX, Hsiue EHC, Cascino K, Thompson EA, Kwaa AK, Singh D, Thapa S, Ordonez AA, Pekosz A, D'Alessio FR, Powell JD, Yegnasubramanian S, Zhou S, Pardoll DM, Ji H, Cox AL, Blankson JN, Smith KN. Functional characterization of CD4+ T cell receptors crossreactive for SARS-CoV-2 and endemic coronaviruses. J Clin Invest 2021; 131:146922. [PMID: 33830946 DOI: 10.1172/jci146922] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUNDRecent studies have reported T cell immunity to the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in unexposed donors, possibly due to crossrecognition by T cells specific for common cold coronaviruses (CCCs). True T cell crossreactivity, defined as the recognition by a single TCR of more than one distinct peptide-MHC ligand, has never been shown in the context of SARS-CoV-2.METHODSWe used the viral functional expansion of specific T cells (ViraFEST) platform to identify T cell responses crossreactive for the spike (S) glycoproteins of SARS-CoV-2 and CCCs at the T cell receptor (TCR) clonotype level in convalescent COVID-19 patients (CCPs) and SARS-CoV-2-unexposed donors. Confirmation of SARS-CoV-2/CCC crossreactivity and assessments of functional avidity were performed using a TCR cloning and transfection system.RESULTSMemory CD4+ T cell clonotypes that crossrecognized the S proteins of SARS-CoV-2 and at least one other CCC were detected in 65% of CCPs and unexposed donors. Several of these TCRs were shared among multiple donors. Crossreactive T cells demonstrated significantly impaired SARS-CoV-2-specific proliferation in vitro relative to monospecific CD4+ T cells, which was consistent with lower functional avidity of their TCRs for SARS-CoV-2 relative to CCC.CONCLUSIONSOur data confirm, for what we believe is the first time, the existence of unique memory CD4+ T cell clonotypes crossrecognizing SARS-CoV-2 and CCCs. The lower avidity of crossreactive TCRs for SARS-CoV-2 may be the result of antigenic imprinting, such that preexisting CCC-specific memory T cells have reduced expansive capacity upon SARS-CoV-2 infection. Further studies are needed to determine how these crossreactive T cell responses affect clinical outcomes in COVID-19 patients.FUNDINGNIH funding (U54CA260492, P30CA006973, P41EB028239, R01AI153349, R01AI145435-A1, R21AI149760, and U19A1088791) was provided by the National Institute of Allergy and Infectious Diseases, the National Cancer Institute, and the National Institute of Biomedical Imaging and Bioengineering. The Bloomberg~Kimmel Institute for Cancer Immunotherapy, The Johns Hopkins University Provost, and The Bill and Melinda Gates Foundation provided funding for this study.
Collapse
Affiliation(s)
- Arbor G Dykema
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | - Boyang Zhang
- Department of Biostatistics, School of Public Health
| | | | | | - Laurene S Cheung
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | - Dilshad Choudhury
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | - Jiajia Zhang
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | - Luis Aparicio
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | - Sadhana Bom
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | - Rufiaat Rashid
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | - Justina X Caushi
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | - Emily Han-Chung Hsiue
- Sidney Kimmel Comprehensive Cancer Center.,Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA.,Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center
| | | | - Elizabeth A Thompson
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | | | - Dipika Singh
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | - Sampriti Thapa
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | | | - Andrew Pekosz
- Department of Molecular Microbiology and Immunology, School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | | | - Jonathan D Powell
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | | | - Shibin Zhou
- Sidney Kimmel Comprehensive Cancer Center.,Ludwig Center, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA.,Howard Hughes Medical Institute, Chevy Chase, Maryland, USA.,Lustgarten Pancreatic Cancer Research Laboratory, Sidney Kimmel Comprehensive Cancer Center
| | - Drew M Pardoll
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| | - Hongkai Ji
- Department of Biostatistics, School of Public Health
| | - Andrea L Cox
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Department of Medicine, School of Medicine, and
| | | | - Kellie N Smith
- Bloomberg~Kimmel Institute for Cancer Immunotherapy.,Sidney Kimmel Comprehensive Cancer Center
| |
Collapse
|
217
|
Maecker HT. Immune profiling of COVID-19: preliminary findings and implications for the pandemic. J Immunother Cancer 2021; 9:jitc-2021-002550. [PMID: 33963016 PMCID: PMC8108128 DOI: 10.1136/jitc-2021-002550] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2021] [Indexed: 12/14/2022] Open
Abstract
SARS-CoV-2 infection can have widely diverse clinical outcomes, from asymptomatic infection to death, with many possible clinical symptoms and syndromes. It is thus essential to understand how the virus interacts with the host immune system to bring about these varied outcomes and to inform vaccine development. We now know that both antibody and T cell responses are induced in the majority of infected individuals, and that cross-reactive responses from other coronaviruses also exist in the uninfected population. Innate immune responses are a key focus of research and may influence the course of disease and the character of subsequent adaptive responses. Finally, baseline immune profiles and changes during early acute infection may be key to predicting the course of disease. Understanding all these aspects can help to create better immune monitoring tools for COVID-19, including tools for predicting disease severity or specific sequelae, perhaps even prior to infection.
Collapse
Affiliation(s)
- Holden T Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
218
|
Repeated Exposure to Subinfectious Doses of SARS-CoV-2 May Promote T Cell Immunity and Protection against Severe COVID-19. Viruses 2021; 13:v13060961. [PMID: 34067349 PMCID: PMC8224680 DOI: 10.3390/v13060961] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/14/2021] [Accepted: 05/19/2021] [Indexed: 12/21/2022] Open
Abstract
Europe is experiencing a third wave of COVID-19 due to the spread of highly transmissible SARS-CoV-2 variants. A number of positive and negative factors constantly shape the rates of COVID-19 infections, hospitalization, and mortality. Among these factors, the rise in increasingly transmissible variants on one side and the effect of vaccinations on the other side create a picture deeply different from that of the first pandemic wave. Starting from the observation that in several European countries the number of COVID-19 infections in the second and third pandemic wave increased without a proportional rise in disease severity and mortality, we hypothesize the existence of an additional factor influencing SARS-CoV-2 dynamics. This factor consists of an immune defence against severe COVID-19, provided by SARS-CoV-2-specific T cells progressively developing upon natural exposure to low virus doses present in populated environments. As suggested by recent studies, low-dose viral particles entering the respiratory and intestinal tracts may be able to induce T cell memory in the absence of inflammation, potentially resulting in different degrees of immunization. In this scenario, non-pharmaceutical interventions would play a double role, one in the short term by reducing the detrimental spreading of SARS-CoV-2 particles, and one in the long term by allowing the development of a widespread (although heterogeneous and uncontrollable) form of immune protection.
Collapse
|
219
|
Grau-Expósito J, Sánchez-Gaona N, Massana N, Suppi M, Astorga-Gamaza A, Perea D, Rosado J, Falcó A, Kirkegaard C, Torrella A, Planas B, Navarro J, Suanzes P, Álvarez-Sierra D, Ayora A, Sansano I, Esperalba J, Andrés C, Antón A, Ramón Y Cajal S, Almirante B, Pujol-Borrell R, Falcó V, Burgos J, Buzón MJ, Genescà M. Peripheral and lung resident memory T cell responses against SARS-CoV-2. Nat Commun 2021; 12:3010. [PMID: 34021148 PMCID: PMC8140108 DOI: 10.1038/s41467-021-23333-3] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/24/2021] [Indexed: 12/13/2022] Open
Abstract
Resident memory T cells (TRM) positioned within the respiratory tract are probably required to limit SARS-CoV-2 spread and COVID-19. Importantly, TRM are mostly non-recirculating, which reduces the window of opportunity to examine these cells in the blood as they move to the lung parenchyma. Here, we identify circulating virus-specific T cell responses during acute infection with functional, migratory and apoptotic patterns modulated by viral proteins and associated with clinical outcome. Disease severity is associated predominantly with IFNγ and IL-4 responses, increased responses against S peptides and apoptosis, whereas non-hospitalized patients have increased IL-12p70 levels, degranulation in response to N peptides and SARS-CoV-2-specific CCR7+ T cells secreting IL-10. In convalescent patients, lung-TRM are frequently detected even 10 months after initial infection, in which contemporaneous blood does not reflect tissue-resident profiles. Our study highlights a balanced anti-inflammatory antiviral response associated with a better outcome and persisting TRM cells as important for future protection against SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Judith Grau-Expósito
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Nerea Sánchez-Gaona
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Núria Massana
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Marina Suppi
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Antonio Astorga-Gamaza
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - David Perea
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Joel Rosado
- Thoracic Surgery and Lung Transplantation Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Anna Falcó
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Cristina Kirkegaard
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Ariadna Torrella
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Bibiana Planas
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Jordi Navarro
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Paula Suanzes
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Daniel Álvarez-Sierra
- Diagnostic Immunology Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Alfonso Ayora
- Occupational Risk Prevention Unit, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Irene Sansano
- Pathology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Departament de Ciències morfològiques, Universitat Autònoma de Barcelona, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Juliana Esperalba
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Cristina Andrés
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Andrés Antón
- Respiratory Viruses Unit, Microbiology Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Santiago Ramón Y Cajal
- Pathology Department, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Departament de Ciències morfològiques, Universitat Autònoma de Barcelona, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Benito Almirante
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Ricardo Pujol-Borrell
- Diagnostic Immunology Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,FOCIS Center of Excellence, Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Vicenç Falcó
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Joaquín Burgos
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - María J Buzón
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.
| | - Meritxell Genescà
- Infectious Diseases Department, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.
| |
Collapse
|
220
|
Low JS, Vaqueirinho D, Mele F, Foglierini M, Jerak J, Perotti M, Jarrossay D, Jovic S, Perez L, Cacciatore R, Terrot T, Pellanda AF, Biggiogero M, Garzoni C, Ferrari P, Ceschi A, Lanzavecchia A, Sallusto F, Cassotta A. Clonal analysis of immunodominance and cross-reactivity of the CD4 T cell response to SARS-CoV-2. Science 2021; 372:1336-1341. [PMID: 34006597 PMCID: PMC8168615 DOI: 10.1126/science.abg8985] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 05/13/2021] [Indexed: 12/14/2022]
Abstract
The identification of CD4+ T cell epitopes is instrumental for the design of subunit vaccines for broad protection against coronaviruses. Here we demonstrate in COVID-19-recovered individuals a robust CD4+ T cell response to naturally processed SARS-CoV-2 spike (S) and nucleoprotein (N), including effector, helper, and memory T cells. By characterizing 2943 S-reactive T cell clones from 34 individuals, we found that 34% of clones and 93% of individuals recognized a conserved immunodominant S346-365 region within the RBD comprising nested HLA-DR- and HLA-DP-restricted epitopes. Using pre- and post-COVID-19 samples and S proteins from endemic coronaviruses, we identify cross-reactive T cells targeting multiple S protein sites. The immunodominant and cross-reactive epitopes identified can inform vaccination strategies to counteract emerging SARS-CoV-2 variants.
Collapse
Affiliation(s)
- Jun Siong Low
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Daniela Vaqueirinho
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Federico Mele
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Mathilde Foglierini
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Josipa Jerak
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Michela Perotti
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - David Jarrossay
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Sandra Jovic
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Laurent Perez
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland
| | - Rosalia Cacciatore
- Laboratory of Immunogenetics, Department of Transfusion Medicine and Immuno-Hematology, Fondazione I.R.C.C.S. Policlinico S. Matteo, 27100 Pavia, Italy
| | - Tatiana Terrot
- Clinical Trial Unit, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland
| | | | - Maira Biggiogero
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco, 6900 Lugano, Switzerland
| | - Christian Garzoni
- Clinic of Internal Medicine and Infectious Diseases, Clinica Luganese Moncucco, 6900 Lugano, Switzerland
| | - Paolo Ferrari
- Faculty of Biomedical Sciences, Università della Svizzera italiana, 6900 Lugano, Switzerland.,Department of Internal Medicine, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland.,Prince of Wales Hospital Clinical School, University of New South Wales, Sydney, New South Wales 2052, Australia
| | - Alessandro Ceschi
- Clinical Trial Unit, Ente Ospedaliero Cantonale, 6500 Bellinzona, Switzerland.,Faculty of Biomedical Sciences, Università della Svizzera italiana, 6900 Lugano, Switzerland.,Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, 6900 Lugano, Switzerland.,Department of Clinical Pharmacology and Toxicology, University Hospital Zurich, 8091 Zurich, Switzerland
| | | | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland. .,Institute of Microbiology, ETH Zürich, 8093 Zurich, Switzerland
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera italiana, 6500 Bellinzona, Switzerland.
| |
Collapse
|
221
|
Woldemeskel BA, Garliss CC, Blankson JN. SARS-CoV-2 mRNA vaccines induce broad CD4+ T cell responses that recognize SARS-CoV-2 variants and HCoV-NL63. J Clin Invest 2021; 131:149335. [PMID: 33822770 PMCID: PMC8121504 DOI: 10.1172/jci149335] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 03/31/2021] [Indexed: 12/13/2022] Open
Abstract
Recent studies have shown T cell cross-recognition of SARS-CoV-2 and common cold coronavirus spike proteins. However, the effect of SARS-CoV-2 vaccines on T cell responses to common cold coronaviruses (CCCs) remains unknown. In this study, we analyzed CD4+ T cell responses to spike peptides from SARS-CoV-2 and 3 CCCs (HCoV-229E, HCoV-NL63, and HCoV-OC43) before and after study participants received Pfizer-BioNTech (BNT162b2) or Moderna (mRNA-1273) mRNA-based COVID-19 vaccines. Vaccine recipients showed broad T cell responses to the SARS-CoV-2 spike protein, and we identified 23 distinct targeted peptides in 9 participants, including 1 peptide that was targeted in 6 individuals. Only 4 of these 23 targeted peptides would potentially be affected by mutations in the UK (B.1.1.7) and South African (B.1.351) variants, and CD4+ T cells from vaccine recipients recognized the 2 variant spike proteins as effectively as they recognized the spike protein from the ancestral virus. Interestingly, we observed a 3-fold increase in the CD4+ T cell responses to HCoV-NL63 spike peptides after vaccination. Our results suggest that T cell responses elicited or enhanced by SARS-CoV-2 mRNA vaccines may be able to control SARS-CoV-2 variants and lead to cross-protection against some endemic coronaviruses.
Collapse
MESH Headings
- Adult
- BNT162 Vaccine
- CD4-Positive T-Lymphocytes/immunology
- COVID-19 Vaccines/immunology
- Coronavirus 229E, Human/genetics
- Coronavirus 229E, Human/immunology
- Coronavirus NL63, Human/genetics
- Coronavirus NL63, Human/immunology
- Coronavirus OC43, Human/genetics
- Coronavirus OC43, Human/immunology
- Cross Reactions
- Female
- Humans
- Male
- Middle Aged
- RNA, Messenger/genetics
- RNA, Messenger/immunology
- SARS-CoV-2/genetics
- SARS-CoV-2/immunology
- Spike Glycoprotein, Coronavirus/genetics
- Spike Glycoprotein, Coronavirus/immunology
Collapse
|
222
|
Lineburg KE, Grant EJ, Swaminathan S, Chatzileontiadou DSM, Szeto C, Sloane H, Panikkar A, Raju J, Crooks P, Rehan S, Nguyen AT, Lekieffre L, Neller MA, Tong ZWM, Jayasinghe D, Chew KY, Lobos CA, Halim H, Burrows JM, Riboldi-Tunnicliffe A, Chen W, D'Orsogna L, Khanna R, Short KR, Smith C, Gras S. CD8 + T cells specific for an immunodominant SARS-CoV-2 nucleocapsid epitope cross-react with selective seasonal coronaviruses. Immunity 2021; 54:1055-1065.e5. [PMID: 33945786 PMCID: PMC8043652 DOI: 10.1016/j.immuni.2021.04.006] [Citation(s) in RCA: 137] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/26/2021] [Accepted: 04/09/2021] [Indexed: 01/16/2023]
Abstract
Efforts are being made worldwide to understand the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus responsible for the coronavirus disease 2019 (COVID-19) pandemic, including the impact of T cell immunity and cross-recognition with seasonal coronaviruses. Screening of SARS-CoV-2 peptide pools revealed that the nucleocapsid (N) protein induced an immunodominant response in HLA-B7+ COVID-19-recovered individuals that was also detectable in unexposed donors. A single N-encoded epitope that was highly conserved across circulating coronaviruses drove this immunodominant response. In vitro peptide stimulation and crystal structure analyses revealed T cell-mediated cross-reactivity toward circulating OC43 and HKU-1 betacoronaviruses but not 229E or NL63 alphacoronaviruses because of different peptide conformations. T cell receptor (TCR) sequencing indicated that cross-reactivity was driven by private TCR repertoires with a bias for TRBV27 and a long CDR3β loop. Our findings demonstrate the basis of selective T cell cross-reactivity for an immunodominant SARS-CoV-2 epitope and its homologs from seasonal coronaviruses, suggesting long-lasting protective immunity.
Collapse
Affiliation(s)
- Katie E Lineburg
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Emma J Grant
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Srividhya Swaminathan
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Demetra S M Chatzileontiadou
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Christopher Szeto
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Hannah Sloane
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Archana Panikkar
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Jyothy Raju
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Pauline Crooks
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Sweera Rehan
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Andrea T Nguyen
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Lea Lekieffre
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Michelle A Neller
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Zhen Wei Marcus Tong
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Dhilshan Jayasinghe
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Keng Yih Chew
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Christian A Lobos
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Hanim Halim
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Jacqueline M Burrows
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | | | - Weisan Chen
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Lloyd D'Orsogna
- Department of Clinical Immunology, PathWest Laboratory Medicine, Fiona Stanley Hospital, Murdoch, WA 6150, Australia; School of Medicine, University of Western Australia, Nedlands, WA 6009, Australia
| | - Rajiv Khanna
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia
| | - Kirsty R Short
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD 4072, Australia; Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Corey Smith
- QIMR Berghofer Centre for Immunotherapy and Vaccine Development and Translational and Human Immunology Laboratory, Department of Immunology, QIMR Berghofer Medical Research Institute, Brisbane, QLD 4006, Australia; Faculty of Medicine, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Stephanie Gras
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia; Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia.
| |
Collapse
|
223
|
Root-Bernstein R. Pneumococcal and Influenza Vaccination Rates and Pneumococcal Invasive Disease Rates Set Geographical and Ethnic Population Susceptibility to Serious COVID-19 Cases and Deaths. Vaccines (Basel) 2021; 9:474. [PMID: 34066697 PMCID: PMC8151685 DOI: 10.3390/vaccines9050474] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/01/2021] [Accepted: 05/05/2021] [Indexed: 12/21/2022] Open
Abstract
This study examines the relationship of pneumococcal vaccination rates, influenza, measles-mumps-rubella (MMR) diphtheria-tetanus-pertussis vaccinations (DTP), polio, Haemophilus influenzae type B (Hib), and Bacillus Calmette-Guerin (tuberculosis) vaccination rates to COVID-19 case and death rates for 51 nations that have high rates of COVID-19 testing and for which nearly complete childhood, at-risk adult and elderly pneumococcal vaccination data were available. The study is unique in a large number of nations examined, the range of vaccine controls, in testing effects of combinations of vaccinations, and in examining the relationship of COVID-19 and vaccination rates to invasive pneumococcal disease (IPD). Analysis of Italian regions and the states of the United States were also performed. Significant positive correlations were found between IPD (but not lower respiratory infections) and COVID-19 rates, while significant negative correlations were found between pneumococcal vaccination and COVID-19 rates. Influenza and MMR vaccination rates were negatively correlated with lower respiratory infection (LRI) rates and may synergize with pneumococcal vaccination rates to protect against COVID-19. Pneumococcal and influenza vaccination rates were independent of other vaccination rates. These results suggest that endemic rates of bacterial pneumonias, for which pneumococci are a sentinel, may set regional and national susceptibility to severe COVID-19 disease and death.
Collapse
Affiliation(s)
- Robert Root-Bernstein
- Department of Physiology, 567 Wilson Road, Room 1104 Biomedical and Physical Sciences Building, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
224
|
Ameratunga R, Woon ST, Jordan A, Longhurst H, Leung E, Steele R, Lehnert K, Snell R, Brooks AES. Perspective: diagnostic laboratories should urgently develop T cell assays for SARS-CoV-2 infection. Expert Rev Clin Immunol 2021; 17:421-430. [PMID: 33745411 DOI: 10.1080/1744666x.2021.1905525] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Introduction: Diagnostic tests play a critical role in the management of Sars-CoV-2, the virus responsible for COVID-19. There are two groups of tests, which are in widespread use to identify patients who have contracted the virus. The commonly used reverse transcriptase quantitative polymerase chain reaction (RT-qPCR) test becomes negative once viral shedding ceases by approximately 2-3weeks. Antibody tests directed to viral antigens become positive after the second week of infection. IgG antibody responses to the virus are muted in children, pregnant females, and those with mild symptoms. IgA and IgM antibodies rapidly wane, although IgG antibodies directed to the receptor-binding domain (RBD) of the spike (S) glycoprotein are more durable. Current data show variability in the sensitivity of commercial and in-house antibody tests to SARS-CoV-2.Areas covered: The role of T cells in acute illness is uncertain, but long-term protection against the virus may rely on memory T cell responses. Measuring memory T cell responses is important for retrospective confirmation of cases, who may have been infected early in the pandemic before reliable RT-qPCR tests were available and whose SARS-CoV-2 antibodies may have become undetectable. Relevant peer-reviewed published references from PubMed are included up to 15 March 2021.Expert opinion: After surveying the literature, the authors present the case for urgent development of diagnostic T cell assays for SARS-CoV-2 by accredited laboratories.
Collapse
Affiliation(s)
- Rohan Ameratunga
- Department of Clinical Immunology, Auckland Hospital, Grafton Auckland, New Zealand.,Department of Virology and Immunology, Auckland Hospital, Grafton, Auckland, New Zealand.,Department of Molecular Medicine and Pathology, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, New Zealand
| | - See-Tarn Woon
- Department of Virology and Immunology, Auckland Hospital, Grafton, Auckland, New Zealand
| | - Anthony Jordan
- Department of Clinical Immunology, Auckland Hospital, Grafton Auckland, New Zealand
| | - Hilary Longhurst
- Department of Clinical Immunology, Auckland Hospital, Grafton Auckland, New Zealand.,Department of Medicine, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, New Zeland
| | - Euphemia Leung
- Auckland Cancer Society Research Centre, School of Medicine, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Richard Steele
- Department of Virology and Immunology, Auckland Hospital, Grafton, Auckland, New Zealand.,Department of Respiratory Medicine, Wellington Hospital, Wellington, New Zealand
| | - Klaus Lehnert
- School of Biological Sciences and Center for Brain Research, University of Auckland, Symonds St, Auckland, New Zealand
| | - Russell Snell
- School of Biological Sciences and Center for Brain Research, University of Auckland, Symonds St, Auckland, New Zealand
| | - Anna E S Brooks
- School of Biological Sciences and Center for Brain Research, University of Auckland, Symonds St, Auckland, New Zealand.,Maurice Wilkins Centre, University of Auckland, New Zealand
| |
Collapse
|
225
|
Majdoubi A, Michalski C, O'Connell SE, Dada S, Narpala S, Gelinas J, Mehta D, Cheung C, Winkler DF, Basappa M, Liu AC, Görges M, Barakauskas VE, Irvine M, Mehalko J, Esposito D, Sekirov I, Jassem AN, Goldfarb DM, Pelech S, Douek DC, McDermott AB, Lavoie PM. A majority of uninfected adults show preexisting antibody reactivity against SARS-CoV-2. JCI Insight 2021; 6:146316. [PMID: 33720905 PMCID: PMC8119195 DOI: 10.1172/jci.insight.146316] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 03/12/2021] [Indexed: 12/24/2022] Open
Abstract
Preexisting cross-reactivity to SARS-CoV-2 occurs in the absence of prior viral exposure. However, this has been difficult to quantify at the population level due to a lack of reliably defined seroreactivity thresholds. Using an orthogonal antibody testing approach, we estimated that about 0.6% of nontriaged adults from the greater Vancouver, Canada, area between May 17 and June 19, 2020, showed clear evidence of a prior SARS-CoV-2 infection, after adjusting for false-positive and false-negative test results. Using a highly sensitive multiplex assay and positive/negative thresholds established in infants in whom maternal antibodies have waned, we determined that more than 90% of uninfected adults showed antibody reactivity against the spike protein, receptor-binding domain (RBD), N-terminal domain (NTD), or the nucleocapsid (N) protein from SARS-CoV-2. This seroreactivity was evenly distributed across age and sex, correlated with circulating coronaviruses' reactivity, and was partially outcompeted by soluble circulating coronaviruses' spike. Using a custom SARS-CoV-2 peptide mapping array, we found that this antibody reactivity broadly mapped to spike and to conserved nonstructural viral proteins. We conclude that most adults display preexisting antibody cross-reactivity against SARS-CoV-2, which further supports investigation of how this may impact the clinical severity of COVID-19 or SARS-CoV-2 vaccine responses.
Collapse
Affiliation(s)
- Abdelilah Majdoubi
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Christina Michalski
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sarah E O'Connell
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Sarah Dada
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Sandeep Narpala
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Jean Gelinas
- Department of Anesthesiology, Surrey Memorial Hospital (SMH), Surrey, British Columbia, Canada.,Department of Anesthesiology & Pain Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Disha Mehta
- Department of Anesthesiology, Surrey Memorial Hospital (SMH), Surrey, British Columbia, Canada.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Claire Cheung
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Dirk Fh Winkler
- Kinexus Bioinformatics Corporation, Vancouver, British Columbia, Canada
| | - Manjula Basappa
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Aaron C Liu
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,Vaccine Evaluation Centre, BC Children's Hospital Research Institute, Vancouver, British Columbia
| | - Matthias Görges
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Vilte E Barakauskas
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mike Irvine
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada
| | - Jennifer Mehalko
- National Cancer Institute RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | - Dominic Esposito
- National Cancer Institute RAS Initiative, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Leidos Biomedical Research Inc., Frederick, Maryland, USA
| | - Inna Sekirov
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,British Columbia Centre for Disease Control (CDC) Public Health Laboratory, Vancouver, British Columbia, Canada
| | - Agatha N Jassem
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada.,British Columbia Centre for Disease Control (CDC) Public Health Laboratory, Vancouver, British Columbia, Canada
| | - David M Goldfarb
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada.,Division of Medical Microbiology, Department of Pathology and Laboratory Medicine, and
| | - Steven Pelech
- Kinexus Bioinformatics Corporation, Vancouver, British Columbia, Canada.,Department of Medicine, University of British Columbia, Vancouver, Canada
| | - Daniel C Douek
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Adrian B McDermott
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Pascal M Lavoie
- BC Children's Hospital Research Institute, Vancouver, British Columbia, Canada.,Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
226
|
Bamidis AD, Koehler P, di Cristanziano V, Rasche K, Demirel B, Bacher P, Hallek M, Kochanek M, Klein F, Hofmann SC, Wesselmann U, Kofler DM. First manifestation of adult-onset Still's disease after COVID-19. LANCET RHEUMATOLOGY 2021; 3:e319-e321. [PMID: 33817663 PMCID: PMC7997647 DOI: 10.1016/s2665-9913(21)00072-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Anna D Bamidis
- Center for Dermatology, Allergy and Dermatosurgery, Helios University Hospital Wuppertal, University Witten/Herdecke, Wuppertal, Germany
| | - Philipp Koehler
- Department I of Internal Medicine, University of Cologne, Cologne 50931, Germany.,CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne 50931, Germany.,Centre for Molecular Medicine Cologne, University of Cologne, Cologne 50931, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany
| | | | - Kurt Rasche
- Department of Pulmonology, Allergology, Sleep and Respiratory Medicine, Helios University Hospital Wuppertal, University Witten/Herdecke, Wuppertal, Germany
| | | | - Petra Bacher
- Institute of Immunology, Christian-Albrechts-University of Kiel, Kiel, Germany.,Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Michael Hallek
- Department I of Internal Medicine, University of Cologne, Cologne 50931, Germany.,CECAD Center of Excellence on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne 50931, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany
| | - Matthias Kochanek
- Department I of Internal Medicine, University of Cologne, Cologne 50931, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany
| | - Florian Klein
- Centre for Molecular Medicine Cologne, University of Cologne, Cologne 50931, Germany.,Institut of Virology, University of Cologne, Cologne 50931, Germany.,German Center for Infection Research, Partner Site Bonn-Cologne, Cologne, Germany
| | - Silke C Hofmann
- Center for Dermatology, Allergy and Dermatosurgery, Helios University Hospital Wuppertal, University Witten/Herdecke, Wuppertal, Germany
| | - Ulrich Wesselmann
- Center for Dermatology, Allergy and Dermatosurgery, Helios University Hospital Wuppertal, University Witten/Herdecke, Wuppertal, Germany
| | - David M Kofler
- Department I of Internal Medicine, University of Cologne, Cologne 50931, Germany.,Center for Integrated Oncology Aachen Bonn Cologne Duesseldorf, Germany
| |
Collapse
|
227
|
Tan H, Lee WS, Wragg KM, Nelson C, Esterbauer R, Kelly HG, Amarasena T, Jones R, Starkey G, Wang BZ, Yoshino O, Tiang T, Grayson ML, Opdam H, D'Costa R, Vago A, Mackay LK, Gordon CL, Wheatley AK, Kent SJ, Juno JA. Adaptive immunity to human coronaviruses is widespread but low in magnitude. Clin Transl Immunology 2021; 10:e1264. [PMID: 33747512 PMCID: PMC7968850 DOI: 10.1002/cti2.1264] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES Endemic human coronaviruses (hCoVs) circulate worldwide but cause minimal mortality. Although seroconversion to hCoV is near ubiquitous during childhood, little is known about hCoV-specific T-cell memory in adults. METHODS We quantified CD4 T-cell and antibody responses to hCoV spike antigens in 42 SARS-CoV-2-uninfected individuals. Antigen-specific memory T cells and circulating T follicular helper (cTFH) cells were identified using an activation-induced marker assay and characterised for memory phenotype and chemokine receptor expression. RESULTS T-cell responses were widespread within conventional memory and cTFH compartments but did not correlate with IgG titres. SARS-CoV-2 cross-reactive T cells were observed in 48% of participants and correlated with HKU1 memory. hCoV-specific T cells exhibited a CCR6+ central memory phenotype in the blood, but were enriched for frequency and CXCR3 expression in human lung-draining lymph nodes. CONCLUSION Overall, hCoV-specific humoral and cellular memory are independently maintained, with a shared phenotype existing among coronavirus-specific CD4 T cells. This understanding of endemic coronavirus immunity provides insight into the homeostatic maintenance of immune responses that are likely to be critical components of protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Hyon‐Xhi Tan
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
| | - Wen Shi Lee
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
| | - Kathleen M Wragg
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
| | - Christina Nelson
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
| | - Robyn Esterbauer
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
| | - Hannah G Kelly
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
- Australian Research Council Centre for Excellence in Convergent Bio‐Nano Science and TechnologyUniversity of MelbourneMelbourneVICAustralia
| | - Thakshila Amarasena
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
| | - Robert Jones
- Department of SurgeryAustin HealthHeidelbergVICAustralia
| | - Graham Starkey
- Department of SurgeryAustin HealthHeidelbergVICAustralia
| | - Bao Zhong Wang
- Department of SurgeryAustin HealthHeidelbergVICAustralia
| | - Osamu Yoshino
- Department of SurgeryAustin HealthHeidelbergVICAustralia
| | - Thomas Tiang
- Department of SurgeryAustin HealthHeidelbergVICAustralia
| | | | - Helen Opdam
- DonateLifeThe Australian Organ and Tissue AuthorityCarltonVICAustralia
- Department of Intensive CareAustin HealthHeidelbergVICAustralia
| | - Rohit D'Costa
- DonateLife VictoriaCarltonVICAustralia
- Intensive Care UnitThe Royal Melbourne HospitalParkvilleVICAustralia
| | - Angela Vago
- Department of SurgeryAustin HealthHeidelbergVICAustralia
| | - Laura K Mackay
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
| | - Claire L Gordon
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
- Department of Infectious DiseasesAustin HealthHeidelbergVICAustralia
| | - Adam K Wheatley
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
| | - Stephen J Kent
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
- Australian Research Council Centre for Excellence in Convergent Bio‐Nano Science and TechnologyUniversity of MelbourneMelbourneVICAustralia
- Melbourne Sexual Health Centre and Department of Infectious DiseasesAlfred Hospital and Central Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Jennifer A Juno
- Department of Microbiology and ImmunologyUniversity of Melbourne, at the Peter Doherty institute for Infection and ImmunityMelbourneVICAustralia
| |
Collapse
|
228
|
Dijkstra JM, Frenette AP, Dixon B. Most Japanese individuals are genetically predisposed to recognize an immunogenic protein fragment shared between COVID-19 and common cold coronaviruses. F1000Res 2021; 10:196. [PMID: 34026045 PMCID: PMC8108557 DOI: 10.12688/f1000research.51479.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/25/2021] [Indexed: 12/15/2022] Open
Abstract
In the spring of 2020, we and others hypothesized that T cells in COVID-19 patients may recognize identical protein fragments shared between the coronaviruses of the common cold and COVID-19 and thereby confer cross-virus immune memory. Here, we look at this issue by screening studies that, since that time, have experimentally addressed COVID-19 associated T cell specificities. Currently, the identical T cell epitope shared between COVID-19 and common cold coronaviruses most convincingly identified as immunogenic is the CD8 + T cell epitope VYIGDPAQL if presented by the MHC class I allele HLA-A*24:02. The HLA-A*24:02 allele is found in the majority of Japanese individuals and several indigenous populations in Asia, Oceania, and the Americas. In combination with histories of common cold infections, HLA-A*24:02 may affect their protection from COVID-19.
Collapse
Affiliation(s)
- Johannes M. Dijkstra
- Institute for Comprehensive Medical Science, Fujita Health Universit, Toyoake-shi, 470-1192, Japan
| | - Aaron P. Frenette
- Department of Biology, University of Waterlo, Waterloo, ON, N2L 3G1, Canada
| | - Brian Dixon
- Department of Biology, University of Waterlo, Waterloo, ON, N2L 3G1, Canada
| |
Collapse
|
229
|
Bertoletti A, Tan AT, Le Bert N. The T-cell response to SARS-CoV-2: kinetic and quantitative aspects and the case for their protective role. OXFORD OPEN IMMUNOLOGY 2021; 2:iqab006. [PMID: 38626271 PMCID: PMC7928654 DOI: 10.1093/oxfimm/iqab006] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Revised: 02/10/2021] [Accepted: 02/17/2021] [Indexed: 12/23/2022] Open
Abstract
Severe Acute Respiratory Syndrome-Coronavirus-2 (SARS-CoV-2), the etiological agent of Coronavirus Diseases 2019 (COVID-19), triggers an adaptive immunity in the infected host that results in the production of virus-specific antibodies and T cells. Although kinetic and quantitative aspects of antibodies have been analyzed in large patient cohorts, similar information about SARS-CoV-2-specific T cells are scarce. We summarize the available knowledge of quantitative and temporal features of the SARS-CoV-2 T-cell response in this review. Currently, most of the data are derived only from the analysis of the circulatory compartment. Despite this limitation, early appearance, multi-specificity and functionality of SARS-CoV-2-specific T cells are associated with accelerated viral clearance and with protection from severe COVID-19.
Collapse
Affiliation(s)
- Antonio Bertoletti
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
- Singapore Immunology Network, A*STAR, Singapore
| | - Anthony T Tan
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| | - Nina Le Bert
- Programme in Emerging Infectious Diseases, Duke-NUS Medical School, Singapore
| |
Collapse
|
230
|
Zhao Y, Kilian C, Turner JE, Bosurgi L, Roedl K, Bartsch P, Gnirck AC, Cortesi F, Schultheiß C, Hellmig M, Enk LUB, Hausmann F, Borchers A, Wong MN, Paust HJ, Siracusa F, Scheibel N, Herrmann M, Rosati E, Bacher P, Kylies D, Jarczak D, Lütgehetmann M, Pfefferle S, Steurer S, Zur-Wiesch JS, Puelles VG, Sperhake JP, Addo MM, Lohse AW, Binder M, Huber S, Huber TB, Kluge S, Bonn S, Panzer U, Gagliani N, Krebs CF. Clonal expansion and activation of tissue-resident memory-like Th17 cells expressing GM-CSF in the lungs of severe COVID-19 patients. Sci Immunol 2021; 6:eabf6692. [PMID: 33622974 PMCID: PMC8128299 DOI: 10.1126/sciimmunol.abf6692] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 02/18/2021] [Indexed: 01/08/2023]
Abstract
Hyperinflammation contributes to lung injury and subsequent acute respiratory distress syndrome (ARDS) with high mortality in patients with severe coronavirus disease 2019 (COVID-19). To understand the underlying mechanisms involved in lung pathology, we investigated the role of the lung-specific immune response. We profiled immune cells in bronchoalveolar lavage fluid and blood collected from COVID-19 patients with severe disease and bacterial pneumonia patients not associated with viral infection. By tracking T cell clones across tissues, we identified clonally expanded tissue-resident memory-like Th17 cells (Trm17 cells) in the lungs even after viral clearance. These Trm17 cells were characterized by a a potentially pathogenic cytokine expression profile of IL17A and CSF2 (GM-CSF). Interactome analysis suggests that Trm17 cells can interact with lung macrophages and cytotoxic CD8+ T cells, which have been associated with disease severity and lung damage. High IL-17A and GM-CSF protein levels in the serum of COVID-19 patients were associated with a more severe clinical course. Collectively, our study suggests that pulmonary Trm17 cells are one potential orchestrator of the hyperinflammation in severe COVID-19.
Collapse
Affiliation(s)
- Yu Zhao
- III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Germany
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Germany
| | - Christoph Kilian
- III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Germany
| | - Jan-Eric Turner
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Lidia Bosurgi
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
- Protozoa Immunology, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Kevin Roedl
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Patricia Bartsch
- III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Germany
| | - Ann-Christin Gnirck
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Filippo Cortesi
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Christoph Schultheiß
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Malte Hellmig
- III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Germany
| | - Leon U B Enk
- III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Germany
| | - Fabian Hausmann
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Germany
| | - Alina Borchers
- III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Germany
| | - Milagros N Wong
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Hans-Joachim Paust
- III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Germany
| | - Francesco Siracusa
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Nicola Scheibel
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Marissa Herrmann
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Elisa Rosati
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
| | - Petra Bacher
- Institute of Clinical Molecular Biology, Christian-Albrechts-University of Kiel, Kiel, Germany
- Institute of Immunology, Christian-Albrechts-University of Kiel & UKSH Schleswig-Holstein, Kiel, Germany
| | - Dominik Kylies
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
| | - Dominik Jarczak
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Marc Lütgehetmann
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Germany
| | - Susanne Pfefferle
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Germany
| | - Stefan Steurer
- Institute for Pathology, University Medical Center Hamburg-Eppendorf, Germany
| | | | - Victor G Puelles
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Jan-Peter Sperhake
- Department of Legal Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Marylyn M Addo
- I. Department of Medicine, Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf, Germany
- German Center for Infection Research, Partner Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Ansgar W Lohse
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Mascha Binder
- Department of Internal Medicine IV, Oncology/Hematology, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Samuel Huber
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Tobias B Huber
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Stefan Kluge
- Department of Intensive Care Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Stefan Bonn
- Institute of Medical Systems Biology, University Medical Center Hamburg-Eppendorf, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
- Center for Biomedical AI, University Medical Center Hamburg-Eppendorf, Germany
| | - Ulf Panzer
- III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
| | - Nicola Gagliani
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany.
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Germany
- Department for General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Germany
- Immunology and Allergy Unit, Department of Medicine, Solna, Karolinska Institute and University Hospital, Stockholm, Sweden
| | - Christian F Krebs
- III. Department of Medicine, Division of Translational Immunology, University Medical Center Hamburg-Eppendorf, Germany.
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
| |
Collapse
|
231
|
Abstract
Understanding the precise nature and durability of protective immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is essential in order to gain insight into the pathophysiology of coronavirus disease 2019 (COVID-19) and to develop novel treatment strategies to this disease. Here I succinctly summarize what is currently known and unknown about the immune response during COVID-19 and discuss whether natural infections can lead to herd immunity.
Collapse
Affiliation(s)
- Masayuki Miyasaka
- Immunology Frontier Research Center (IFReC), Osaka University, Japan
| |
Collapse
|
232
|
Cohen CA, Li APY, Hachim A, Hui DSC, Kwan MYW, Tsang OTY, Chiu SS, Chan WH, Yau YS, Kavian N, Ma FNL, Lau EHY, Cheng SMS, Poon LLM, Peiris JSM, Valkenburg SA. SARS-CoV-2 specific T cell responses are lower in children and increase with age and time after infection. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.02.02.21250988. [PMID: 33564773 PMCID: PMC7872365 DOI: 10.1101/2021.02.02.21250988] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
SARS-CoV-2 infection of children leads to a mild illness and the immunological differences with adults remains unclear. We quantified the SARS-CoV-2 specific T cell responses in adults and children (<13 years of age) with RT-PCR confirmed asymptomatic and symptomatic infection for long-term memory, phenotype and polyfunctional cytokines. Acute and memory CD4+ T cell responses to structural SARS-CoV-2 proteins significantly increased with age, whilst CD8+ T cell responses increased with time post infection. Infected children had significantly lower CD4+ and CD8+ T cell responses to SARS-CoV-2 structural and ORF1ab proteins compared to infected adults. SARS-CoV-2-specific CD8+ T cell responses were comparable in magnitude to uninfected negative adult controls. In infected adults CD4+ T cell specificity was skewed towards structural peptides, whilst children had increased contribution of ORF1ab responses. This may reflect differing T cell compartmentalisation for antigen processing during antigen exposure or lower recruitment of memory populations. T cell polyfunctional cytokine production was comparable between children and adults, but children had a lower proportion of SARS-CoV-2 CD4+ T cell effector memory. Compared to adults, children had significantly lower levels of antibodies to β-coronaviruses, indicating differing baseline immunity. Total T follicular helper responses was increased in children during acute infection indicating rapid co-ordination of the T and B cell responses. However total monocyte responses were reduced in children which may be reflective of differing levels of inflammation between children and adults. Therefore, reduced prior β-coronavirus immunity and reduced activation and recruitment of de novo responses in children may drive milder COVID-19 pathogenesis.
Collapse
Affiliation(s)
- Carolyn A Cohen
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Athena PY Li
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Asmaa Hachim
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - David SC Hui
- Department of Medicine and Therapeutics, Prince of Wales Hospital, Chinese University of Hong Kong, Hong Kong SAR, China
| | - Mike YW Kwan
- Department of Paediatric and Adolescent Medicine, Hong Kong Hospital Authority Infectious Disease Center, Princess Margaret Hospital, Special Administrative Region of Hong Kong, China
| | - Owen TY Tsang
- Infectious Diseases Centre, Princess Margaret Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Susan S Chiu
- Department of Paediatric and Adolescent Medicine, The University of Hong Kong and Queen Mary Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Wai Hung Chan
- Department of Paediatrics, Queen Elizabeth Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Yat Sun Yau
- Department of Paediatrics, Queen Elizabeth Hospital, Hospital Authority of Hong Kong, Special Administrative Region of Hong Kong, China
| | - Niloufar Kavian
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Fionn NL Ma
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Eric HY Lau
- WHO Collaborating Centre for Infectious Disease Epidemiology and Control, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Samuel MS Cheng
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Leo LM Poon
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - JS Malik Peiris
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Division of Public Health Laboratory Sciences, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sophie A Valkenburg
- HKU-Pasteur Research Pole, School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
233
|
Circuits between infected macrophages and T cells in SARS-CoV-2 pneumonia. Nature 2021; 590:635-641. [PMID: 33429418 PMCID: PMC7987233 DOI: 10.1038/s41586-020-03148-w] [Citation(s) in RCA: 523] [Impact Index Per Article: 130.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 12/21/2020] [Indexed: 01/29/2023]
Abstract
Some patients infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) develop severe pneumonia and acute respiratory distress syndrome1 (ARDS). Distinct clinical features in these patients have led to speculation that the immune response to virus in the SARS-CoV-2-infected alveolus differs from that in other types of pneumonia2. Here we investigate SARS-CoV-2 pathobiology by characterizing the immune response in the alveoli of patients infected with the virus. We collected bronchoalveolar lavage fluid samples from 88 patients with SARS-CoV-2-induced respiratory failure and 211 patients with known or suspected pneumonia from other pathogens, and analysed them using flow cytometry and bulk transcriptomic profiling. We performed single-cell RNA sequencing on 10 bronchoalveolar lavage fluid samples collected from patients with severe coronavirus disease 2019 (COVID-19) within 48 h of intubation. In the majority of patients with SARS-CoV-2 infection, the alveolar space was persistently enriched in T cells and monocytes. Bulk and single-cell transcriptomic profiling suggested that SARS-CoV-2 infects alveolar macrophages, which in turn respond by producing T cell chemoattractants. These T cells produce interferon-γ to induce inflammatory cytokine release from alveolar macrophages and further promote T cell activation. Collectively, our results suggest that SARS-CoV-2 causes a slowly unfolding, spatially limited alveolitis in which alveolar macrophages containing SARS-CoV-2 and T cells form a positive feedback loop that drives persistent alveolar inflammation.
Collapse
|
234
|
Koch CM, Prigge AD, Anekalla KR, Shukla A, Do-Umehara HC, Setar L, Chavez J, Abdala-Valencia H, Politanska Y, Markov NS, Hahn GR, Heald-Sargent T, Sanchez-Pinto LN, Muller WJ, Misharin AV, Ridge KM, Coates BM. Immune response to SARS-CoV-2 in the nasal mucosa in children and adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.01.26.21250269. [PMID: 33532801 PMCID: PMC7852252 DOI: 10.1101/2021.01.26.21250269] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
RATIONALE Despite similar viral load and infectivity rates between children and adults infected with SARS-CoV-2, children rarely develop severe illness. Differences in the host response to the virus at the primary infection site are among the proposed mechanisms. OBJECTIVES To investigate the host response to SARS-CoV-2, respiratory syncytial virus (RSV), and influenza virus (IV) in the nasal mucosa in children and adults. METHODS Clinical outcomes and gene expression in the nasal mucosa were analyzed in 36 children hospitalized with SARS-CoV-2 infection, 24 children with RSV infection, 9 children with IV infection, 16 adults with mild to moderate SARS-CoV-2 infection, and 7 healthy pediatric and 13 healthy adult controls. RESULTS In both children and adults, infection with SARS-CoV-2 leads to an interferon response in the nasal mucosa. The magnitude of the interferon response correlated with the abundance of viral reads and was comparable between symptomatic children and adults infected with SARS-CoV-2 and symptomatic children infected with RSV and IV. Cell type deconvolution identified an increased abundance of immune cells in the samples from children and adults with a viral infection. Expression of ACE2 and TMPRSS2 - key entry factors for SARS-CoV-2 - did not correlate with age or presence or absence of viral infection. CONCLUSIONS Our findings support the hypothesis that differences in the immune response to SARS-CoV-2 determine disease severity, independent of viral load and interferon response at the primary infection primary site.
Collapse
Affiliation(s)
| | - Andrew D Prigge
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | | | - Avani Shukla
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | | | - Leah Setar
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | - Jairo Chavez
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | | | | | | | - Grant R Hahn
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | - Taylor Heald-Sargent
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | - L Nelson Sanchez-Pinto
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
- Department of Preventive Medicine, Northwestern University
| | - William J Muller
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| | | | - Karen M Ridge
- Department of Medicine, Northwestern University
- Department of Cell and Developmental Biology, Northwestern University
| | - Bria M Coates
- Department of Pediatrics, Northwestern University
- Ann & Robert H. Lurie Children's Hospital of Chicago
| |
Collapse
|
235
|
da Silva Antunes R, Pallikkuth S, Williams E, Yu ED, Mateus J, Quiambao L, Wang E, Rawlings SA, Stadlbauer D, Jiang K, Amanat F, Arnold D, Andrews D, Fuego I, Dan JM, Grifoni A, Weiskopf D, Krammer F, Crotty S, Hoffer ME, Pahwa SG, Sette A. Differential T cell reactivity to seasonal coronaviruses and SARS-CoV-2 in community and health care workers. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2021:2021.01.12.21249683. [PMID: 33469594 PMCID: PMC7814840 DOI: 10.1101/2021.01.12.21249683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Herein we measured CD4+ T cell responses against common cold corona (CCC) viruses and SARS-CoV-2 in high-risk health care workers (HCW) and community controls. We observed higher levels of CCC reactive T cells in SARS-CoV-2 seronegative HCW compared to community donors, consistent with potential higher occupational exposure of HCW to CCC. We further show that SARS-CoV-2 reactivity of seronegative HCW was higher than community controls and correlation between CCC and SARS-CoV-2 responses is consistent with cross-reactivity and not associated with recent in vivo activation. Surprisingly, CCC reactivity was decreased in SARS-CoV-2 infected HCW, suggesting that exposure to SARS-CoV-2 might interfere with CCC responses, either directly or indirectly. This result was unexpected, but consistently detected in independent cohorts derived from Miami and San Diego.
Collapse
Affiliation(s)
- Ricardo da Silva Antunes
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Suresh Pallikkuth
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Erin Williams
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Esther Dawen Yu
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Jose Mateus
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Lorenzo Quiambao
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Eric Wang
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Stephen A. Rawlings
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, CA 92037, USA
| | - Daniel Stadlbauer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kaijun Jiang
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fatima Amanat
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David Arnold
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - David Andrews
- Department of Pathology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Irma Fuego
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Jennifer M. Dan
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Daniela Weiskopf
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shane Crotty
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| | - Michael E. Hoffer
- Department of Otolaryngology, University of Miami, Miller School of Medicine, Miami, FL, USA
- Department of Neurological Surgery, University of Miami, Miller School of Medicine, Miami, FL, USA
| | - Savita G. Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology (LJI), La Jolla, CA 92037, USA
- Department of Medicine, Division of Infectious Diseases and Global Public Health, University of California, San Diego (UCSD), La Jolla, CA 92037, USA
| |
Collapse
|
236
|
Flemming A. Crossreactivity not so helpful after all? Nat Rev Immunol 2021; 21:70. [PMID: 33402725 PMCID: PMC7783696 DOI: 10.1038/s41577-020-00494-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
237
|
Minervina AA, Komech EA, Titov A, Bensouda Koraichi M, Rosati E, Mamedov IZ, Franke A, Efimov GA, Chudakov DM, Mora T, Walczak AM, Lebedev YB, Pogorelyy MV. Longitudinal high-throughput TCR repertoire profiling reveals the dynamics of T-cell memory formation after mild COVID-19 infection. eLife 2021; 10:e63502. [PMID: 33399535 PMCID: PMC7806265 DOI: 10.7554/elife.63502] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 01/05/2021] [Indexed: 12/12/2022] Open
Abstract
COVID-19 is a global pandemic caused by the SARS-CoV-2 coronavirus. T cells play a key role in the adaptive antiviral immune response by killing infected cells and facilitating the selection of virus-specific antibodies. However, neither the dynamics and cross-reactivity of the SARS-CoV-2-specific T-cell response nor the diversity of resulting immune memory is well understood. In this study, we use longitudinal high-throughput T-cell receptor (TCR) sequencing to track changes in the T-cell repertoire following two mild cases of COVID-19. In both donors, we identified CD4+ and CD8+ T-cell clones with transient clonal expansion after infection. We describe characteristic motifs in TCR sequences of COVID-19-reactive clones and show preferential occurrence of these motifs in publicly available large dataset of repertoires from COVID-19 patients. We show that in both donors, the majority of infection-reactive clonotypes acquire memory phenotypes. Certain T-cell clones were detected in the memory fraction at the pre-infection time point, suggesting participation of pre-existing cross-reactive memory T cells in the immune response to SARS-CoV-2.
Collapse
Affiliation(s)
| | - Ekaterina A Komech
- Shemyakin-Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
- Pirogov Russian National Research Medical UniversityMoscowRussian Federation
| | - Aleksei Titov
- National Research Center for HematologyMoscowRussian Federation
| | - Meriem Bensouda Koraichi
- Laboratoire de physique de l'École Normale Supérieure, ENS, PSL, Sorbonne Universite, Universite de Paris, and CNRSParisFrance
| | - Elisa Rosati
- Institute of Clinical Molecular Biology, Kiel UniversityKielGermany
| | - Ilgar Z Mamedov
- Shemyakin-Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
- Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Masaryk University, Central European Institute of TechnologyBrnoCzech Republic
- V.I. Kulakov National Medical Research Center for Obstetrics, Gynecology and PerinatologyMoscowRussian Federation
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel UniversityKielGermany
| | | | - Dmitriy M Chudakov
- Shemyakin-Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
- Pirogov Russian National Research Medical UniversityMoscowRussian Federation
- Masaryk University, Central European Institute of TechnologyBrnoCzech Republic
| | - Thierry Mora
- Laboratoire de physique de l'École Normale Supérieure, ENS, PSL, Sorbonne Universite, Universite de Paris, and CNRSParisFrance
| | - Aleksandra M Walczak
- Laboratoire de physique de l'École Normale Supérieure, ENS, PSL, Sorbonne Universite, Universite de Paris, and CNRSParisFrance
| | - Yuri B Lebedev
- Shemyakin-Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
- Moscow State UniversityMoscowRussian Federation
| | - Mikhail V Pogorelyy
- Shemyakin-Ovchinnikov Institute of Bioorganic ChemistryMoscowRussian Federation
- Pirogov Russian National Research Medical UniversityMoscowRussian Federation
| |
Collapse
|
238
|
Moi ML. [Dengue amidst COVID-19: challenges & control measures for the double burden]. Uirusu 2021; 71:1-10. [PMID: 35526989 DOI: 10.2222/jsv.71.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Dengue, an arbovirus, is a public health treat in the tropics and sub-tropical climates worldwide. The disease incidence has grown dramatically worldwide, with an estimated 390 million dengue virus infection per year. Dengue has distinct epidemiological patterns which are associated with the four virus serotypes. All four serotypes can co-circulate within a region, in which a number of regions are hyperendemic for all 4 serotypes. Currently, there are no specific treatment or vaccine for the disease. Dengue prevention depends on vector control measures and early interventions. The COVID-19 pandemic has placed immense pressure on health care and management systems worldwide. During the COVID-19 pandemic, the situation was aggravated by the simultaneous dengue outbreaks, that has led to a double burden which has further impacted the healthcare sector, particularly in resource limited settings. This review article will focus on dengue epidemics during the COVID-19 pandemic and discuss on recent findings on immunological cascades between dengue and COVID-19 and, the impact on vaccine development.
Collapse
|
239
|
da Silva LT, Ortega MM, Tiyo BT, Viana IFT, de Lima TE, Tozetto-Mendoza TR, Oliveira LMDS, Teixeira FME, Lins RD, de Almeida A, Mendes-Correa MC, da Silva Duarte AJ, Oshiro TM. SARS-CoV-2 recombinant proteins stimulate distinct cellular and humoral immune response profiles in samples from COVID-19 convalescent patients. Clinics (Sao Paulo) 2021; 76:e3548. [PMID: 34878034 PMCID: PMC8610223 DOI: 10.6061/clinics/2021/e3548] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Accepted: 10/27/2021] [Indexed: 01/11/2023] Open
Abstract
OBJECTIVES In this preliminary study we investigated cellular and humoral immune responses to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antigens in blood samples from 14 recovered coronavirus disease 2019 (COVID-19) patients and compared them to those in samples from 12 uninfected/unvaccinated volunteers. METHODS Cellular immunity was assessed by intracellular detection of IFN-γ in CD3+ T lymphocytes after stimulation with SARS-CoV-2 spike (S1), nucleocapsid (NC), or receptor-binding domain (RBD) recombinant proteins or overlapping peptide pools covering the sequence of SARS-CoV-2 spike, membrane and nucleocapsid regions. The humoral response was examined by ELISAs and/or chemiluminescence assays for the presence of serum IgG antibodies directed to SARS-CoV-2 proteins. RESULTS We observed differences between humoral and cellular immune profiles in response to stimulation with the same proteins. Assays of IgG antibodies directed to SARS-CoV-2 NC, RBD and S1/S2 recombinant proteins were able to differentiate convalescent from uninfected/unvaccinated groups. Cellular immune responses to SARS-CoV-2 protein stimuli did not exhibit a specific response, as T cells from both individuals with no history of contact with SARS-CoV-2 and from recovered donors were able to produce IFN-γ. CONCLUSIONS Determination of the cellular immune response to stimulation with a pool of SARS-CoV-2 peptides but not with SARS-CoV-2 proteins is able to distinguish convalescent individuals from unexposed individuals. Regarding the humoral immune response, the screening for serum IgG antibodies directed to SARS-CoV-2 proteins has been shown to be specific for the response of recovered individuals.
Collapse
Affiliation(s)
- Laís Teodoro da Silva
- Laboratorio de Investigacao Medica em Dermatologia e Imunodeficiencias (LIM 56), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Corresponding author. E-mail:
| | - Marina Mazzilli Ortega
- Laboratorio de Investigacao Medica em Dermatologia e Imunodeficiencias (LIM 56), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Bruna Tiaki Tiyo
- Laboratorio de Investigacao Medica em Dermatologia e Imunodeficiencias (LIM 56), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | | | - Tayná Evily de Lima
- Departamento de Virologia, Instituto Aggeu Magalhaes, Fundacao Oswaldo Cruz, Recife, PE, BR
| | - Tania Regina Tozetto-Mendoza
- Laboratorio de Virologia (LIM-52), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Luanda Mara da Silva Oliveira
- Laboratorio de Investigacao Medica em Dermatologia e Imunodeficiencias (LIM 56), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Franciane Mouradian Emidio Teixeira
- Laboratorio de Investigacao Medica em Dermatologia e Imunodeficiencias (LIM 56), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Roberto Dias Lins
- Departamento de Virologia, Instituto Aggeu Magalhaes, Fundacao Oswaldo Cruz, Recife, PE, BR
| | - Alexandre de Almeida
- Laboratorio de Investigacao Medica em Dermatologia e Imunodeficiencias (LIM 56), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Maria Cassia Mendes-Correa
- Laboratorio de Virologia (LIM-52), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Alberto Jose da Silva Duarte
- Laboratorio de Investigacao Medica em Dermatologia e Imunodeficiencias (LIM 56), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
- Divisao de Laboratorio Central, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| | - Telma Miyuki Oshiro
- Laboratorio de Investigacao Medica em Dermatologia e Imunodeficiencias (LIM 56), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, BR
| |
Collapse
|