1
|
Xu L, He R, Ye X, Wang Y, Hui S, Li H, Chen H, Huang P. Leveraging transcriptome-wide association studies identifies the relationship between upper respiratory flora and cell type-specific gene expression in severe respiratory disease. PLoS One 2025; 20:e0322864. [PMID: 40343915 PMCID: PMC12063895 DOI: 10.1371/journal.pone.0322864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/30/2025] [Indexed: 05/11/2025] Open
Abstract
ObjectivesThe upper respiratory tract flora may influence host immunity and modulate susceptibility to viral respiratory infections. This study aimed to investigate the associations between upper respiratory tract flora and immune cells in severe ILI, identify specific microbial taxa and immune response pathways contributing to disease severity, and elucidate how flora influences ILI progression by modulating immune cell functions.MethodsHeritability of GWAS summary data was estimated using LDSC (v1.0.1). Gene-level genetic associations were analyzed with MAGMA. scRNA-seq data were integrated with genetic association data using scDRS. FUSION was used to construct cell type-specific expression quantitative trait locus models based on genotypes and scRNA-seq data from the onek1k project, which were combined with flora abundance-related GWAS data for a transcriptome-wide association study.ResultsFrom the LDSC analysis, data from 1195 severe ILI-associated GWASs with upper respiratory flora(h2 > 0.1) were included in subsequent analysis. TWAS identified 19 significant association pairs (Padj < 0.05), and 1226 differentially expressed genes between mild and severe ILI patients (Padj < 0.05 and | log2FC|>0.25). Functional enrichment analyses using GO, KEGG, and Reactome databases revealed that immune cells,such as CD4 + T effector memory cells, cDCs, NK cells, were enriched in multiple biological processes or pathways.ConclusionsThis study identified associations between severe ILI-related upper respiratory tract flora and cell type-specific gene expression, potentially explaining how differential flora influences ILI progression. CD16 + monocytes exhibited the most differentially expressed genes, followed by proliferating cells and cDCs, highlighting the significant role of immune cell-enriched pathways in ILI progression.
Collapse
Affiliation(s)
- Lei Xu
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Ran He
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Xiangyu Ye
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Yifan Wang
- Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, Jiangsu, China
| | - Shirong Hui
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Haochang Li
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| | - Hongbo Chen
- Department of Infectious Disease, Jurong Hospital Affiliated to Jiangsu University, Jurong, Jiangsu, China
| | - Peng Huang
- Depatment of Epidemiology, Center for Global Health, School of Public Health, National Vaccine Innovation Platiorm, Nanjing Medical University, Nanjing, China
| |
Collapse
|
2
|
Salou M, Paiva RA, Lantz O. Development and Functions of MAIT Cells. Annu Rev Immunol 2025; 43:253-283. [PMID: 39879553 DOI: 10.1146/annurev-immunol-082323-025943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Mucosal-associated invariant T (MAIT) cells are evolutionarily conserved T cells that recognize microbial metabolites. They are abundant in humans and conserved during mammalian evolution, which suggests that they have important nonredundant functions. In this article, we discuss the evolutionary conservation of MAIT cells and describe their original developmental process. MAIT cells exert a wide variety of effector functions, from killing infected cells and promoting inflammation to repairing tissues. We provide insights into these functions and discuss how they result from the context of stimulation encountered by MAIT cells in different tissues and pathological settings. We describe how MAIT cell numbers and features are modified in disease states, focusing mainly on in vivo models. Lastly, we discuss emerging strategies to manipulate MAIT cells for therapeutic purposes.
Collapse
Affiliation(s)
- Marion Salou
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
| | - Rafael A Paiva
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
| | - Olivier Lantz
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
- Laboratoire d'Immunologie Clinique, Institut Curie, Paris, France
- Centre d'Investigation Clinique en Biothérapie, Gustave-Roussy and Institut Curie (CIC-BT1428), Paris, France
| |
Collapse
|
3
|
Fang Y, Chen Y, Niu S, Lyu Z, Tian Y, Shen X, Li YR, Yang L. Biological functions and therapeutic applications of human mucosal-associated invariant T cells. J Biomed Sci 2025; 32:32. [PMID: 40025566 PMCID: PMC11871619 DOI: 10.1186/s12929-025-01125-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/18/2025] [Indexed: 03/04/2025] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are a unique subset of innate-like T lymphocytes that bridge innate and adaptive immunity. Characterized by their semi-invariant T cell receptor (TCR) and abundant localization in mucosal tissues, MAIT cells recognize microbial metabolites, primarily derived from the riboflavin biosynthesis pathway, presented by the major histocompatibility complex (MHC)-related protein 1 (MR1). This interaction, along with co-stimulatory signals, triggers rapid immune responses, including cytokine secretion and cytotoxic activity, highlighting their importance in maintaining immune homeostasis and combating infections. This review provides an in-depth overview of MAIT cell biology, including development, activation pathways, and functional diversity, highlighting their protective roles in immunity, contributions to diseases like cancer and inflammatory bowel disease (IBD), and context-dependent dual functions in health and pathology. This review also highlights the emerging therapeutic potential of MAIT cells in immunotherapy. Their unique TCR specificity, abundance, and tissue-homing properties make them ideal candidates for engineering novel therapies, such as chimeric antigen receptor (CAR)-MAIT cells, targeting infections, cancers, and autoimmune diseases. Challenges like antigen escape, T cell exhaustion, and CAR design optimization must be addressed to enhance clinical efficacy. In summary, MAIT cells are integral to immune function, and their therapeutic potential presents exciting opportunities for the treatment of a wide range of diseases. Further research is essential to unlock the full potential of these versatile immune cells.
Collapse
Affiliation(s)
- Ying Fang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yuning Chen
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Siyue Niu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Zibai Lyu
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yanxin Tian
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Xinyuan Shen
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yan-Ruide Li
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| | - Lili Yang
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Parker Institute for Cancer Immunotherapy, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
4
|
Sugimoto C, Wakao H. The Role of Mucosal-Associated Invariant T Cells in Viral Infections and Their Function in Vaccine Development. Vaccines (Basel) 2025; 13:155. [PMID: 40006702 PMCID: PMC11860804 DOI: 10.3390/vaccines13020155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/29/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
Mucosal-Associated Invariant T (MAIT) cells, which bridge innate and adaptive immunity, have emerged as an important player in viral infections despite their inability to directly recognize viral antigens. This review provides a comprehensive analysis of MAIT cell responses across different viral infections, revealing consistent patterns in their behavior and function. We discuss the dynamics of MAIT cells during various viral infections, including changes in their frequency, activation status, and functional characteristics. Particular attention is given to emerging strategies for MAIT-cell-targeted vaccine development, including the use of MR1 ligands as mucosal adjuvants and the activation of MAIT cells through viral vectors and mRNA vaccines. Current knowledge of MAIT cell biology in viral infections provides promising approaches for harnessing their functions in vaccine development.
Collapse
Affiliation(s)
- Chie Sugimoto
- Host Defense Division, Research Center for Advanced Medical Science, Dokkyo Medical University, Mibu 321-0293, Japan;
| | | |
Collapse
|
5
|
Meggyes M, Nagy DU, Toth I, Feik T, Polgar B, Deen ISA, Sipos D, Szereday L, Peterfalvi A. Immune Checkpoint Receptor Expression Profiles of MAIT Cells in Moderate and Severe COVID-19. Scand J Immunol 2025; 101:e70008. [PMID: 39980239 PMCID: PMC11842947 DOI: 10.1111/sji.70008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
MAIT cells are one of the largest unconventional T cell populations and, recruited to the site of infection, play both protective and pathogenic roles during pulmonary viral infections. MAIT cell activation patterns change significantly during COVID-19, with a notable decrease in their frequency in peripheral blood of severe cases. In the present study, we aimed to investigate the expression profiles of various immune checkpoint pathways on MAIT, MAIT-like and non-MAIT cells in moderate and severe COVID-19 patients undergoing cytokine storm. Despite numerous studies comparing MAIT cell characteristics based on COVID-19 disease severity, none have delved into the critical differences in MAIT cell immune checkpoint profiles between moderate and severe COVID-19 patients, all experiencing a cytokine storm. Flow cytometry was used to analyse peripheral blood mononuclear cells from a cohort of 35 patients, comprising 18 moderate and 17 severe cases, alongside 14 healthy controls. Our investigation specifically focuses on severe COVID-19 presentations, revealing a marked deletion of MAIT cells. Further exploration into the regulatory dynamics of MAIT cell functionality reveals shifts in the expression profiles of critical immune checkpoint receptors, notably PD-1 and CD226. In severe COVID-19 patients, MAIT cells showed a significant decrease in the expression of CD226, whereas MAIT-like and non-MAIT cells demonstrated a significant increase in the expression of PD-1 compared to healthy individuals. The expression of the TIGIT receptor remained unaltered across all investigated groups. Our findings contribute to the existing knowledge by elucidating the changes in MAIT cell subpopulations and their potential role in COVID-19 disease severity.
Collapse
Affiliation(s)
- Matyas Meggyes
- Department of Medical Microbiology and ImmunologyMedical School, University of PecsPecsHungary
- Janos Szentagothai Research CentrePecsHungary
| | - David U. Nagy
- Institute of Geobotany/Plant Ecology, Martin‐Luther‐UniversityHalle (Saale)Germany
| | - Ildiko Toth
- Department of Anesthesiology and Intensive TherapyMedical School, University of PecsPecsHungary
| | - Timoteus Feik
- Department of Medical Microbiology and ImmunologyMedical School, University of PecsPecsHungary
| | - Beata Polgar
- Department of Medical Microbiology and ImmunologyMedical School, University of PecsPecsHungary
| | - Iyad Saad Al Deen
- Department of Medical Microbiology and ImmunologyMedical School, University of PecsPecsHungary
| | - David Sipos
- 1st Department of Medicine, Division of Infectious DiseasesMedical School, University of PecsPecsHungary
| | - Laszlo Szereday
- Department of Medical Microbiology and ImmunologyMedical School, University of PecsPecsHungary
- Janos Szentagothai Research CentrePecsHungary
| | - Agnes Peterfalvi
- Department of Laboratory MedicineMedical School, University of PecsPecsHungary
| |
Collapse
|
6
|
Shi Q, Zhang P, Hu Q, Zhang T, Hou R, Yin S, Zou Y, Chen F, Jiao S, Si L, Zheng B, Chen Y, Zhan T, Liu Y, Zhu W, Qi N. Role of TOMM34 on NF-κB activation-related hyperinflammation in severely ill patients with COVID-19 and influenza. EBioMedicine 2024; 108:105343. [PMID: 39276680 PMCID: PMC11418153 DOI: 10.1016/j.ebiom.2024.105343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 08/21/2024] [Accepted: 09/02/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND Highly pathogenic respiratory RNA viruses such as SARS-CoV-2 and its associated syndrome COVID-19 pose a tremendous threat to the global public health. Innate immune responses to SARS-CoV-2 depend mainly upon the NF-κB-mediated inflammation. Identifying unknown host factors driving the NF-κB activation and inflammation is crucial for the development of immune intervention strategies. METHODS Published single-cell RNA sequencing (scRNA-seq) data was used to analyze the differential transcriptome profiles of bronchoalveolar lavage (BAL) cells between healthy individuals (n = 27) and patients with severe COVID-19 (n = 21), as well as the differential transcriptome profiles of peripheral blood mononuclear cells (PBMCs) between healthy individuals (n = 22) and severely ill patients with COVID-19 (n = 45) or influenza (n = 16). Loss-of-function and gain-of-function assays were performed in diverse viruses-infected cells and male mice models to identify the role of TOMM34 in antiviral innate immunity. FINDINGS TOMM34, together with a list of genes encoding pro-inflammatory cytokines and antiviral immune proteins, was transcriptionally upregulated in circulating monocytes, lung epithelium and innate immune cells from individuals with severe COVID-19 or influenza. Deficiency of TOMM34/Tomm34 significantly impaired the type I interferon responses and NF-κB-mediated inflammation in various human/murine cell lines, murine bone marrow-derived macrophages (BMDMs) and in vivo. Mechanistically, TOMM34 recruits TRAF6 to facilitate the K63-linked polyubiquitination of NEMO upon viral infection, thus promoting the downstream NF-κB activation. INTERPRETATION In this study, viral induction of TOMM34 is positively correlated with the hyperinflammation in severely ill patients with COVID-19 and influenza. Our findings also highlight the physiological role of TOMM34 in the innate antiviral signallings. FUNDING A full list of funding sources can be found in the acknowledgements section.
Collapse
Affiliation(s)
- Qiwen Shi
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Pengfei Zhang
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Qingtao Hu
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Tianxin Zhang
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Ruixia Hou
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Shengxiang Yin
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Yilin Zou
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Fenghua Chen
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Shuang Jiao
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Lanlan Si
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Bangjin Zheng
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Yichao Chen
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China
| | - Tingzhu Zhan
- Collaborative Innovation Center of Yangtze River Delta Region Green Pharmaceuticals, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, 310014, China
| | - Yongxiang Liu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China.
| | - Wenting Zhu
- Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China.
| | - Nan Qi
- State Key Laboratory of Respiratory Disease, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China; Guangzhou National Laboratory, Guangzhou International Bio-Island, Guangzhou, 510005, China.
| |
Collapse
|
7
|
Amini A, Klenerman P, Provine NM. Role of mucosal-associated invariant T cells in coronavirus disease 2019 vaccine immunogenicity. Curr Opin Virol 2024; 67:101412. [PMID: 38838550 PMCID: PMC11511680 DOI: 10.1016/j.coviro.2024.101412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 05/07/2024] [Accepted: 05/08/2024] [Indexed: 06/07/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells are an unconventional T cell population that are highly abundant in humans. They possess a semi-invariant T cell receptor (TCR) that recognises microbial metabolites formed during riboflavin biosynthesis, presented on a nonpolymorphic MHC-like molecule MR1. MAIT cells possess an array of effector functions, including type 1, type 17, and tissue repair activity. Deployment of these functions depends on the stimuli they receive through their TCR and/or cytokine receptors. Strong cytokine signalling, such as in response to vaccination, can bypass TCR triggering and provokes a strong proinflammatory response. Although data are still emerging, multiple aspects of MAIT cell biology are associated with modulation of immunity induced by the coronavirus disease 2019 mRNA and adenovirus vector vaccines. In this review, we will address how MAIT cells may play a role in immunogenicity of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and how these cells can be harnessed as cellular adjuvants.
Collapse
Affiliation(s)
- Ali Amini
- Translational Gastroenterology Unit, Nuffield Department of Medicine - Experimental Medicine, University of Oxford, UK
| | - Paul Klenerman
- Translational Gastroenterology Unit, Nuffield Department of Medicine - Experimental Medicine, University of Oxford, UK; Peter Medawar Building for Pathogen Research, Nuffield Department of Medicine, University of Oxford, UK; Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, UK.
| | - Nicholas M Provine
- Pandemic Sciences Institute, Nuffield Department of Medicine, University of Oxford, UK; Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, UK.
| |
Collapse
|
8
|
Pena NM, Santana LC, Hunter JR, Blum VF, Vergara T, Gouvea C, Leal E, Bellei N, Schechter M, Diaz RS. T cell-mediated Immune response and correlates of inflammation and their relationship with COVID-19 clinical severity: not an intuitive guess. BMC Infect Dis 2024; 24:612. [PMID: 38902613 PMCID: PMC11191252 DOI: 10.1186/s12879-024-09490-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/10/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND Predictors of the outcome of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) infection remain to be fully determined. We evaluated selected viral characteristics and immunological responses that might predict and/or correlate to the clinical outcome of COVID-19. METHODS For individuals developing divergent clinical outcomes, the magnitude and breadth of T cell-mediated responses were measured within 36 h of symptom onset. Peripheral Blood Mononuclear Cells (PBMCs) were subjected to in vitro stimulation with SARS-CoV-2-based peptides. In addition, SARS-CoV-2 sequences were generated by metagenome, and HLA typing was performed using Luminex technology. FINDINGS CD4+ T cell activation was negatively correlated with SARS-CoV-2 basal viral load in patients with severe COVID-19 (p = 0·043). The overall cellular immune response, as inferred by the IFN-γ signal, was higher at baseline for patients who progressed to mild disease compared to patients who progressed to severe disease (p = 0·0044). Subjects with milder disease developed higher T cell responses for MHC class I and II-restricted peptides (p = 0·033). INTERPRETATION Mounting specific cellular immune responses in the first days after symptom onset, as inferred by IFN-γ magnitude in the ELISPOT assay, may efficiently favor a positive outcome. In contrast, progression to severe COVID-19 was accompanied by stronger cellular immune responses, higher CD4 + T cell activation, and a higher number of in silico predicted high-affinity class I HLA alleles.
Collapse
Affiliation(s)
- Nathalia Mantovani Pena
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
- Weill Cornell Medicine, New York, United States of America
| | - Luiz Claudio Santana
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
| | - James R Hunter
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
| | - Vinicius Fontanesi Blum
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
| | - Tania Vergara
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
- Oncohiv, Rio de Janeiro, Brazil
| | - Celso Gouvea
- Centro de Hematologia e Hemoterapia do Ceará, Fortaleza, CE, Brazil
| | - Elcio Leal
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belem, Pará, Brazil
| | - Nancy Bellei
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
| | - Mauro Schechter
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil
- Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Ricardo Sobhie Diaz
- Infectious Diseases Division, Federal University of São Paulo (UNIFESP), Pedro de Toledo, 669, Vila Clementino, Sao Paulo, SP, 04039-032, Brazil.
| |
Collapse
|
9
|
Maaß H, Ynga-Durand M, Milošević M, Krstanović F, Matešić MP, Žuža I, Jonjić S, Brizić I, Šustić A, Bloos F, Protić A, Čičin-Šain L. Serum cytokine dysregulation signatures associated with COVID-19 outcomes in high mortality intensive care unit cohorts across pandemic waves and variants. Sci Rep 2024; 14:13605. [PMID: 38871772 DOI: 10.1038/s41598-024-64384-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 06/07/2024] [Indexed: 06/15/2024] Open
Abstract
The aim of this study was to characterize the systemic cytokine signature of critically ill COVID-19 patients in a high mortality setting aiming to identify biomarkers of severity, and to explore their associations with viral loads and clinical characteristics. We studied two COVID-19 critically ill patient cohorts from a referral centre located in Central Europe. The cohorts were recruited during the pre-alpha/alpha (November 2020 to April 2021) and delta (end of 2021) period respectively. We determined both the serum and bronchoalveolar SARS-CoV-2 viral load and identified the variant of concern (VoC) involved. Using a cytokine multiplex assay, we quantified systemic cytokine concentrations and analyzed their relationship with clinical findings, routine laboratory workup and pulmonary function data obtained during the ICU stay. Patients who did not survive had a significantly higher systemic and pulmonary viral load. Patients infected with the pre-alpha VoC showed a significantly lower viral load in comparison to those infected with the alpha- and delta-variants. Levels of systemic CTACK, M-CSF and IL-18 were significantly higher in non-survivors in comparison to survivors. CTACK correlated directly with APACHE II scores. We observed differences in lung compliance and the association between cytokine levels and pulmonary function, dependent on the VoC identified. An intra-cytokine analysis revealed a loss of correlation in the non-survival group in comparison to survivors in both cohorts. Critically ill COVID-19 patients exhibited a distinct systemic cytokine profile based on their survival outcomes. CTACK, M-CSF and IL-18 were identified as mortality-associated analytes independently of the VoC involved. The Intra-cytokine correlation analysis suggested the potential role of a dysregulated systemic network of inflammatory mediators in severe COVID-19 mortality.
Collapse
Affiliation(s)
- Henrike Maaß
- Department of Viral Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine (CiiM), a joint venture of Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany
| | - Mario Ynga-Durand
- Department of Viral Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany
- Centre for Individualized Infection Medicine (CiiM), a joint venture of Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany
| | - Marko Milošević
- Department of Anesthesiology, Faculty of Medicine, Reanimation, Intensive Care and Emergency Medicine, University of Rijeka, Rijeka, Croatia
| | - Fran Krstanović
- Faculty of Medicine, Center for Proteomics, University of Rijeka, Rijeka, Croatia
| | | | - Iva Žuža
- Department of Radiology, Clinical Hospital Centre Rijeka, Rijeka, Croatia
| | - Stipan Jonjić
- Faculty of Medicine, Center for Proteomics, University of Rijeka, Rijeka, Croatia
| | - Ilija Brizić
- Faculty of Medicine, Center for Proteomics, University of Rijeka, Rijeka, Croatia
| | - Alan Šustić
- Department of Anesthesiology, Faculty of Medicine, Reanimation, Intensive Care and Emergency Medicine, University of Rijeka, Rijeka, Croatia
- Department of Clinical Medical Science II, Faculty of Health Studies, University of Rijeka, Rijeka, Croatia
| | - Frank Bloos
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Jena, Germany
| | - Alen Protić
- Department of Anesthesiology, Faculty of Medicine, Reanimation, Intensive Care and Emergency Medicine, University of Rijeka, Rijeka, Croatia
| | - Luka Čičin-Šain
- Department of Viral Immunology, Helmholtz Center for Infection Research, Braunschweig, Germany.
- Centre for Individualized Infection Medicine (CiiM), a joint venture of Helmholtz Centre for Infection Research and Hannover Medical School, Hannover, Germany.
- German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, Braunschweig, Germany.
| |
Collapse
|
10
|
Opsteen S, Fram T, Files JK, Levitan EB, Goepfert P, Erdmann N. Impact of Chronic HIV Infection on Acute Immune Responses to SARS-CoV-2. J Acquir Immune Defic Syndr 2024; 96:92-100. [PMID: 38408318 PMCID: PMC11009054 DOI: 10.1097/qai.0000000000003399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 01/29/2024] [Indexed: 02/28/2024]
Abstract
ABSTRACT There is mounting evidence that HIV infection is a risk factor for severe presentations of COVID-19. We hypothesized that the persistent immune activation associated with chronic HIV infection contributes to worsened outcomes during acute COVID-19. The goals of this study were to provide an in-depth analysis of immune response to acute COVID-19 and investigate relationships between immune responses and clinical outcomes in an unvaccinated, sex- and race-matched cohort of people with HIV (PWH, n = 20) and people without HIV (PWOH, n = 41). We performed flow cytometric analyses on peripheral blood mononuclear cells from PWH and PWOH experiencing acute COVID-19 (≤21-day postsymptom onset). PWH were younger (median 52 vs 65 years) and had milder COVID-19 (40% vs 88% hospitalized) compared with PWOH. Flow cytometry panels included surface markers for immune cell populations, activation and exhaustion surface markers (with and without SARS-CoV-2-specific antigen stimulation), and intracellular cytokine staining. We observed that PWH had increased expression of activation (eg, CD137 and OX40) and exhaustion (eg, PD1 and TIGIT) markers as compared to PWOH during acute COVID-19. When analyzing the impact of COVID-19 severity, we found that hospitalized PWH had lower nonclassical (CD16 + ) monocyte frequencies, decreased expression of TIM3 on CD4 + T cells, and increased expression of PDL1 and CD69 on CD8 + T cells. Our findings demonstrate that PWH have increased immune activation and exhaustion as compared to a cohort of predominately older, hospitalized PWOH and raises questions on how chronic immune activation affects acute disease and the development of postacute sequelae.
Collapse
Affiliation(s)
- Skye Opsteen
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Tim Fram
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Jacob K. Files
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Emily B. Levitan
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL
| | - Paul Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| | - Nathaniel Erdmann
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL; and
| |
Collapse
|
11
|
Huang X, Kantonen J, Nowlan K, Nguyen NA, Jokiranta ST, Kuivanen S, Heikkilä N, Mahzabin S, Kantele A, Vapalahti O, Myllykangas L, Heinonen S, Mäyränpää MI, Strandin T, Kekäläinen E. Mucosal-Associated Invariant T Cells are not susceptible in vitro to SARS-CoV-2 infection but accumulate into the lungs of COVID-19 patients. Virus Res 2024; 341:199315. [PMID: 38211733 PMCID: PMC10826420 DOI: 10.1016/j.virusres.2024.199315] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/15/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
Prolonged T cell lymphopenia is common in COVID-19, caused by SARS-CoV-2. While the mechanisms of lymphopenia during COVID-19 remain elusive, it is especially pronounced in a specialized innate-like T cell population called Mucosal Associated Invariant T cells (MAITs). MAITs has been suggested to express Angiotensin-Converting Enzyme 2 (ACE2), which is the well-known cellular receptor for SARS-CoV-2. However, it is still unclear if SARS-CoV-2 can infect or affect MAIT cells directly. In this study, we performed multicolor flow cytometry on peripheral blood mononuclear cells obtained from COVID-19 patients to assess the frequencies of CD8+Vα7.2+CD161+ MAIT subsets at acute and convalescent disease phases. The susceptibility of MAITs and T cells to direct exposure by SARS-CoV-2 was analysed using cells isolated from healthy donor buffy coats by viability assays, virus-specific RT-PCR, and flow cytometry. In situ lung immunofluorescence was used to evaluate retention of T cells, especially MAIT cells, in lung tissues during acute COVID-19. Our study confirms previous reports indicating that circulating MAITs are activated, and their frequency is declined in patients with acute SARS-CoV-2 infection, whereas an accumulation of MAITs and T cells was seen in the lung tissue of individuals with fatal COVID-19. However, despite a fraction of MAITs found to express ACE2, no evidence for the susceptibility of MAITs for direct infection or activation by SARS-CoV-2 particles was observed. Thus, their activation and decline in the circulation is most likely explained by indirect mechanisms involving other immune cells and cytokine-induced pro-inflammatory environment but not by direct exposure to viral particles at the infection site.
Collapse
Affiliation(s)
- Xiaobo Huang
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland.
| | - Jonas Kantonen
- Department of Pathology, University of Helsinki, Helsinki, Finland; Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Kirsten Nowlan
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Ngoc Anh Nguyen
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Suvi T Jokiranta
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland
| | - Suvi Kuivanen
- Department of Virology, Medicum, University of Helsinki, Helsinki, Finland; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Virology, Berlin, Germany
| | - Nelli Heikkilä
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Center of Vaccinology, University of Geneva, Geneva, Switzerland
| | - Shamita Mahzabin
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Anu Kantele
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Meilahti Vaccine Research Center, MeVac, Infectious Diseases, Helsinki University and Helsinki University Hospital, Helsinki, Finland
| | - Olli Vapalahti
- Division of Virology and Immunology, HUS Diagnostic Center, HUSLAB Clinical Microbiology, Helsinki, Finland; Zoonosis Unit, Department of Virology, Medicum, University of Helsinki, Helsinki, Finland; Department of Veterinary Biosciences, University of Helsinki, Helsinki, Finland
| | - Liisa Myllykangas
- Department of Pathology, University of Helsinki, Helsinki, Finland; Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Santtu Heinonen
- New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mikko I Mäyränpää
- Department of Pathology, University of Helsinki, Helsinki, Finland; Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Tomas Strandin
- Zoonosis Unit, Department of Virology, Medicum, University of Helsinki, Helsinki, Finland
| | - Eliisa Kekäläinen
- Translational Immunology Research Program, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; Department of Bacteriology and Immunology, University of Helsinki, Helsinki, Finland; Division of Virology and Immunology, HUS Diagnostic Center, HUSLAB Clinical Microbiology, Helsinki, Finland
| |
Collapse
|
12
|
Samer C, McWilliam HE, McSharry BP, Velusamy T, Burchfield JG, Stanton RJ, Tscharke DC, Rossjohn J, Villadangos JA, Abendroth A, Slobedman B. Multi-targeted loss of the antigen presentation molecule MR1 during HSV-1 and HSV-2 infection. iScience 2024; 27:108801. [PMID: 38303725 PMCID: PMC10831258 DOI: 10.1016/j.isci.2024.108801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 09/18/2023] [Accepted: 01/02/2024] [Indexed: 02/03/2024] Open
Abstract
The major histocompatibility complex (MHC), Class-I-related (MR1) molecule presents microbiome-synthesized metabolites to Mucosal-associated invariant T (MAIT) cells, present at sites of herpes simplex virus (HSV) infection. During HSV type 1 (HSV-1) infection there is a profound and rapid loss of MR1, in part due to expression of unique short 3 protein. Here we show that virion host shutoff RNase protein downregulates MR1 protein, through loss of MR1 transcripts. Furthermore, a third viral protein, infected cell protein 22, also downregulates MR1, but not classical MHC-I molecules. This occurs early in the MR1 trafficking pathway through proteasomal degradation. Finally, HSV-2 infection results in the loss of MR1 transcripts, and intracellular and surface MR1 protein, comparable to that seen during HSV-1 infection. Thus HSV coordinates a multifaceted attack on the MR1 antigen presentation pathway, potentially protecting infected cells from MAIT cell T cell receptor-mediated detection at sites of primary infection and reactivation.
Collapse
Affiliation(s)
- Carolyn Samer
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, and the Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Hamish E.G. McWilliam
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Brian P. McSharry
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, and the Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
- School of Dentistry and Medical Sciences, Faculty of Science and Health, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Thilaga Velusamy
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - James G. Burchfield
- Charles Perkins Centre, The University of Sydney, Sydney, NSW, Australia
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW, Australia
| | - Richard J. Stanton
- Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, Wales
| | - David C. Tscharke
- John Curtin School of Medical Research, Australian National University, Canberra, ACT, Australia
| | - Jamie Rossjohn
- Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, Wales
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jose A. Villadangos
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC, Australia
| | - Allison Abendroth
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, and the Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| | - Barry Slobedman
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, and the Charles Perkins Centre, The University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|
13
|
Kammann T, Gorin JB, Parrot T, Gao Y, Ponzetta A, Emgård J, Maleki KT, Sekine T, Rivera-Ballesteros O, Karolinska COVID-19 Study Group, Gredmark-Russ S, Rooyackers O, Skagerberg M, Eriksson LI, Norrby-Teglund A, Mak JY, Fairlie DP, Björkström NK, Klingström J, Ljunggren HG, Aleman S, Buggert M, Strålin K, Sandberg JK. Dynamic MAIT Cell Recovery after Severe COVID-19 Is Transient with Signs of Heterogeneous Functional Anomalies. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:389-396. [PMID: 38117799 PMCID: PMC10784727 DOI: 10.4049/jimmunol.2300639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/16/2023] [Indexed: 12/22/2023]
Abstract
Mucosal-associated invariant T (MAIT) cells are an abundant population of unconventional T cells in humans and play important roles in immune defense against microbial infections. Severe COVID-19 is associated with strong activation of MAIT cells and loss of these cells from circulation. In the present study, we investigated the capacity of MAIT cells to recover after severe COVID-19. In longitudinal paired analysis, MAIT cells initially rebounded numerically and phenotypically in most patients at 4 mo postrelease from the hospital. However, the rebounding MAIT cells displayed signs of persistent activation with elevated expression of CD69, CD38, and HLA-DR. Although MAIT cell function was restored in many patients, a subgroup displayed a predominantly PD-1high functionally impaired MAIT cell pool. This profile was associated with poor expression of IFN-γ and granzyme B in response to IL-12 + L-18 and low levels of polyfunctionality. Unexpectedly, although the overall T cell counts recovered, normalization of the MAIT cell pool failed at 9-mo follow-up, with a clear decline in MAIT cell numbers and a further increase in PD-1 levels. Together, these results indicate an initial transient period of inconsistent recovery of MAIT cells that is not sustained and eventually fails. Persisting MAIT cell impairment in previously hospitalized patients with COVID-19 may have consequences for antimicrobial immunity and inflammation and could potentially contribute to post-COVID-19 health problems.
Collapse
Affiliation(s)
- Tobias Kammann
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jean-Baptiste Gorin
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Tiphaine Parrot
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Yu Gao
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Andrea Ponzetta
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Johanna Emgård
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Kimia T. Maleki
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Takuya Sekine
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Olga Rivera-Ballesteros
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Sara Gredmark-Russ
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Olav Rooyackers
- Department of Clinical Interventions and Technology, Karolinska Institutet, Stockholm, Sweden
- Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
| | - Magdalena Skagerberg
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Lars I. Eriksson
- Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Anna Norrby-Teglund
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jeffrey Y.W. Mak
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David P. Fairlie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Niklas K. Björkström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Jonas Klingström
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Soo Aleman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases and Dermatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Kristoffer Strålin
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Division of Infectious Diseases and Dermatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Johan K. Sandberg
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
14
|
Hurt W, Youngs J, Ball J, Edgeworth J, Hopkins P, Jenkins DR, Leaver S, Mazzella A, Molloy SF, Schelenz S, Wise MP, White PL, Yusuff H, Wyncoll D, Bicanic T. COVID-19-associated pulmonary aspergillosis in mechanically ventilated patients: a prospective, multicentre UK study. Thorax 2023; 79:75-82. [PMID: 37657925 PMCID: PMC10804023 DOI: 10.1136/thorax-2023-220002] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 06/22/2023] [Indexed: 09/03/2023]
Abstract
BACKGROUND Invasive pulmonary aspergillosis is a complication of severe COVID-19, with regional variation in reported incidence and mortality. We describe the incidence, risk factors and mortality associated with COVID-19-associated pulmonary aspergillosis (CAPA) in a prospective, multicentre UK cohort. METHODS From March 2020 to March 2021, 266 mechanically ventilated adults with COVID-19 were enrolled across 5 UK hospital intensive care units (ICUs). CAPA was defined using European Confederation for Medical Mycology and the International Society for Human and Animal Mycology criteria and fungal diagnostics performed on respiratory and serum samples. RESULTS Twenty-nine of 266 patients (10.9%) had probable CAPA, 14 (5.2%) possible CAPA and none proven CAPA. Probable CAPA was diagnosed a median of 9 (IQR 7-16) days after ICU admission. Factors associated with probable CAPA after multivariable logistic regression were cumulative steroid dose given within 28 days prior to ICU admission (adjusted OR (aOR) 1.16; 95% CI 1.01 to 1.43 per 100 mg prednisolone-equivalent), receipt of an interleukin (IL)-6 inhibitor (aOR 2.79; 95% CI 1.22 to 6.48) and chronic obstructive pulmonary disease (COPD) (aOR 4.78; 95% CI 1.13 to 18.13). Mortality in patients with probable CAPA was 55%, vs 46% in those without. After adjustment for immortal time bias, CAPA was associated with an increased risk of 90-day mortality (HR 1.85; 95% CI 1.07 to 3.19); however, this association did not remain statistically significant after further adjustment for confounders (adjusted HR 1.57; 95% CI 0.88 to 2.80). There was no difference in mortality between patients with CAPA prescribed antifungals (9 of 17; 53%) and those who were not (7 of 12; 58%) (p=0.77). INTERPRETATION In this first prospective UK study, probable CAPA was associated with corticosteroid use, receipt of IL-6 inhibitors and pre-existing COPD. CAPA did not impact mortality following adjustment for prognostic variables.
Collapse
Affiliation(s)
- William Hurt
- Institute of Infection and Immunity, St George's University of London, London, UK
- Clinical Infection Unit, St George's University Hospitals NHS Foundation Trust, London, UK
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Jonathan Youngs
- Institute of Infection and Immunity, St George's University of London, London, UK
- Clinical Infection Unit, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Jonathan Ball
- Adult Critical Care, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Jonathan Edgeworth
- Clinical Infection and Microbiology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Philip Hopkins
- Adult Critical Care, King's College Hospital NHS Foundation Trust, London, UK
| | - David R Jenkins
- Clinical Microbiology, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Susannah Leaver
- Adult Critical Care, St George's University Hospitals NHS Foundation Trust, London, UK
| | - Andrea Mazzella
- Institute of Infection and Immunity, St George's University of London, London, UK
| | - Síle F Molloy
- Institute of Infection and Immunity, St George's University of London, London, UK
| | - Silke Schelenz
- Medical Microbiology, King's College Hospital NHS Foundation Trust, London, UK
| | - Matt P Wise
- Adult Critical Care, University of Wales Hospital, Cardiff, UK
| | | | - Hakeem Yusuff
- Adult Critical Care, University Hospitals of Leicester NHS Trust, Leicester, UK
| | - Duncan Wyncoll
- Adult Critical Care, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Tihana Bicanic
- Institute of Infection and Immunity, St George's University of London, London, UK
- Clinical Infection Unit, St George's University Hospitals NHS Foundation Trust, London, UK
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| |
Collapse
|
15
|
Burt P, Thurley K. Distribution modeling quantifies collective T H cell decision circuits in chronic inflammation. SCIENCE ADVANCES 2023; 9:eadg7668. [PMID: 37703364 PMCID: PMC10881075 DOI: 10.1126/sciadv.adg7668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 08/11/2023] [Indexed: 09/15/2023]
Abstract
Immune responses are tightly regulated by a diverse set of interacting immune cell populations. Alongside decision-making processes such as differentiation into specific effector cell types, immune cells initiate proliferation at the beginning of an inflammation, forming two layers of complexity. Here, we developed a general mathematical framework for the data-driven analysis of collective immune cell dynamics. We identified qualitative and quantitative properties of generic network motifs, and we specified differentiation dynamics by analysis of kinetic transcriptome data. Furthermore, we derived a specific, data-driven mathematical model for T helper 1 versus T follicular helper cell-fate decision dynamics in acute and chronic lymphocytic choriomeningitis virus infections in mice. The model recapitulates important dynamical properties without model fitting and solely by using measured response-time distributions. Model simulations predict different windows of opportunity for perturbation in acute and chronic infection scenarios, with potential implications for optimization of targeted immunotherapy.
Collapse
Affiliation(s)
- Philipp Burt
- Systems Biology of Inflammation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Institute for Theoretical Biophysics, Humboldt University, Berlin, Germany
| | - Kevin Thurley
- Systems Biology of Inflammation, German Rheumatism Research Center (DRFZ), a Leibniz Institute, Berlin, Germany
- Biomathematics Division, Institute of Experimental Oncology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
16
|
Tian L, Xu J, Chen C, Lin J, Ju L, Chen L, Zhang Y, Han X, Liu L. HLA-DR + mucosal-associated invariant T cells predict poor prognosis in patients with sepsis: A prospective observational study. Scand J Immunol 2023; 98:e13286. [PMID: 37163215 DOI: 10.1111/sji.13286] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 02/09/2023] [Accepted: 04/22/2023] [Indexed: 05/11/2023]
Abstract
Mucosal-associated invariant T (MAIT) cells are important in antibacterial immune responses; however, during sepsis, they are few in number and exhibit highly activated phenotypes. The relationship between MAIT cells in peripheral blood and the prognosis of sepsis is not well understood. Thus, this study aimed to examine the levels and phenotypes of MAIT cells in early sepsis, evaluate their clinical relevance, and investigate their association with patient prognosis. This prospective observational study enrolled 72 septic patients defined according to the Sepsis 3.0 criteria and 21 healthy controls matched for age and sex. Their peripheral blood samples were used to assay the expression of immune activation (CD69 and HLA-DR) and immune checkpoint (PD-1 and PD-L1) markers on MAIT cells. The systemic inflammatory response syndrome, acute physiology and chronic health evaluation (APACHE) II, and sequential organ failure assessment scores were recorded. Subsequently, the association between MAIT cell characteristics and clinical indicators was assessed using Spearman's rank correlation analysis, and binary logistic regression analysis with a forward stepwise approach assessed independent risk factors for 28-day mortality. We noted a decrease in the percentage of MAIT cells in the patients' peripheral blood, which exhibited an activated phenotype. Besides, HLA-DR+ MAIT cell percentage and the APACHE II score were independently associated with the 28-day mortality and, in combination, were the best indicators of mortality. Thus, the percentage of HLA-DR+ MAIT cells in early sepsis serves as a novel prognostic biomarker for predicting mortality and improves the predictive capacity of the APACHE II score.
Collapse
Affiliation(s)
- Lijun Tian
- Department of Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of Critical Care Medicine, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Junxian Xu
- Department of Critical Care Medicine, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Cong Chen
- Physical Examination Center, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Jinfeng Lin
- Department of Critical Care Medicine, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Linling Ju
- Nantong Institute of Liver Disease, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Lin Chen
- Nantong Institute of Liver Disease, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Yufeng Zhang
- Department of Respiratory Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, China
| | - Xudong Han
- Department of Critical Care Medicine, Nantong Third People's Hospital, Affiliated Nantong Hospital 3 of Nantong University, Nantong, China
| | - Lijun Liu
- Department of Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
17
|
Kurioka A, Klenerman P. Aging unconventionally: γδ T cells, iNKT cells, and MAIT cells in aging. Semin Immunol 2023; 69:101816. [PMID: 37536148 PMCID: PMC10804939 DOI: 10.1016/j.smim.2023.101816] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 07/19/2023] [Accepted: 07/20/2023] [Indexed: 08/05/2023]
Abstract
Unconventional T cells include γδ T cells, invariant Natural Killer T cells (iNKT) cells and Mucosal Associated Invariant T (MAIT) cells, which are distinguished from conventional T cells by their recognition of non-peptide ligands presented by non-polymorphic antigen presenting molecules and rapid effector functions that are pre-programmed during their development. Here we review current knowledge of the effect of age on unconventional T cells, from early life to old age, in both mice and humans. We then discuss the role of unconventional T cells in age-associated diseases and infections, highlighting the similarities between members of the unconventional T cell family in the context of aging.
Collapse
Affiliation(s)
- Ayako Kurioka
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | - Paul Klenerman
- Nuffield Department of Medicine, University of Oxford, Oxford, UK; Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| |
Collapse
|
18
|
Sandberg JK, Leeansyah E, Eller MA, Shacklett BL, Paquin-Proulx D. The Emerging Role of MAIT Cell Responses in Viral Infections. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:511-517. [PMID: 37549397 PMCID: PMC10421619 DOI: 10.4049/jimmunol.2300147] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 05/08/2023] [Indexed: 08/09/2023]
Abstract
Mucosal-associated invariant T (MAIT) cells are unconventional T cells with innate-like antimicrobial responsiveness. MAIT cells are known for MR1 (MHC class I-related protein 1)-restricted recognition of microbial riboflavin metabolites giving them the capacity to respond to a broad range of microbes. However, recent progress has shown that MAIT cells can also respond to several viral infections in humans and in mouse models, ranging from HIV-1 and hepatitis viruses to influenza virus and SARS-CoV-2, in a primarily cognate Ag-independent manner. Depending on the disease context MAIT cells can provide direct or indirect antiviral protection for the host and may help recruit other immune cells, but they may also in some circumstances amplify inflammation and aggravate immunopathology. Furthermore, chronic viral infections are associated with varying degrees of functional and numerical MAIT cell impairment, suggesting secondary consequences for host defense. In this review, we summarize recent progress and highlight outstanding questions regarding the emerging role of MAIT cells in antiviral immunity.
Collapse
Affiliation(s)
- Johan K. Sandberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Edwin Leeansyah
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
- Precision Medicine and Healthcare Research Centre, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen, China
| | - Michael A. Eller
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Barbara L. Shacklett
- Department of Medical Microbiology and Immunology, School of Medicine, University of California Davis, Davis, CA
| | - Dominic Paquin-Proulx
- U.S. Military HIV Research Program, Walter Reed Army Institute of Research, Silver Spring, MD
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD
| |
Collapse
|
19
|
Beitari S, Duque D, Bavananthasivam J, Hewitt M, Sandhu JK, Hadžisejdić I, Tran A. Cross protection to SARS-CoV-2 variants in hamsters with naturally-acquired immunity. Virol J 2023; 20:167. [PMID: 37507719 PMCID: PMC10386765 DOI: 10.1186/s12985-023-02136-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/21/2023] [Indexed: 07/30/2023] Open
Abstract
Since SARS-CoV-2 was first reported in late 2019, multiple variations of the original virus have emerged. Each variant harbors accumulations of mutations, particularly within the spike glycoprotein, that are associated with increased viral transmissibility and escape immunity. The different mutations in the spike protein of different variants shape the subsequent antibody and T cell responses, such that exposure to different spike proteins can result in reduced or enhanced responses to heterologous variants further down the line. Globally, people have been exposed and re-exposed to multiple variations of the Ancestral strain, including the five variants of concerns. Studies have shown that the protective immune response of an individual is influenced by which strain or combination of strains they are exposed to. The initial exposure to a specific strain may also shape their subsequent immune patterns and response to later infections with a heterologous virus. Most immunological observations were carried out early during the pandemic when the Ancestral strain was circulating. However, SARS-CoV-2 variants exhibit varying patterns of disease severity, waning immunity, immune evasion and sensitivity to therapeutics. Here we investigated the cross-protection in hamsters previously infected with a variant of concern (VOC) and subsequently re-infected with a heterologous variant. We also determined if cross-protection and immunity were dependent on the specific virus to which the hamster was first exposed. We further profiled the host cytokine response induced by each SARS-CoV-2 variants as well as subsequent to re-infection. A comparative analysis of the three VOCs revealed that Alpha variant was the most pathogenic VOC to emerge. We showed that naturally acquired immunity protected hamsters from subsequent re-infection with heterologous SARS-CoV-2 variant, regardless which variant the animal was first exposed to. Our study supports observations that heterologous infection of different SARS-CoV-2 variants do not exacerbate disease in subsequent re-infections. The continual emergence of new SARS-CoV-2 variants mandates a better understanding of cross-protection and immune imprinting in infected individuals. Such information is essential to guide vaccine strategy and public policy to emerging SARS-CoV-2 VOCs and future novel pandemic coronaviruses.
Collapse
Affiliation(s)
- Saina Beitari
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, ON, Ottawa, Canada
| | - Diana Duque
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, ON, Ottawa, Canada
| | - Jegarubee Bavananthasivam
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, ON, Ottawa, Canada
| | - Melissa Hewitt
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, ON, Ottawa, Canada
| | - Jagdeep K Sandhu
- Preclinical Imaging, Human Health Therapeutics Research Centre, National Research Council Canada, ON, Ottawa, Canada
| | - Ita Hadžisejdić
- Clinical Department of Pathology and Cytology, University of Rijeka, Rijeka, Croatia
| | - Anh Tran
- Infectious Diseases, Human Health Therapeutics Research Centre, National Research Council Canada, ON, Ottawa, Canada.
| |
Collapse
|
20
|
Rashu R, Ninkov M, Wardell CM, Benoit JM, Wang NI, Meilleur CE, D'Agostino MR, Zhang A, Feng E, Saeedian N, Bell GI, Vahedi F, Hess DA, Barr SD, Troyer RM, Kang CY, Ashkar AA, Miller MS, Haeryfar SMM. Targeting the MR1-MAIT cell axis improves vaccine efficacy and affords protection against viral pathogens. PLoS Pathog 2023; 19:e1011485. [PMID: 37384813 DOI: 10.1371/journal.ppat.1011485] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 06/14/2023] [Indexed: 07/01/2023] Open
Abstract
Mucosa-associated invariant T (MAIT) cells are MR1-restricted, innate-like T lymphocytes with tremendous antibacterial and immunomodulatory functions. Additionally, MAIT cells sense and respond to viral infections in an MR1-independent fashion. However, whether they can be directly targeted in immunization strategies against viral pathogens is unclear. We addressed this question in multiple wild-type and genetically altered but clinically relevant mouse strains using several vaccine platforms against influenza viruses, poxviruses and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). We demonstrate that 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil (5-OP-RU), a riboflavin-based MR1 ligand of bacterial origin, can synergize with viral vaccines to expand MAIT cells in multiple tissues, reprogram them towards a pro-inflammatory MAIT1 phenotype, license them to bolster virus-specific CD8+ T cell responses, and potentiate heterosubtypic anti-influenza protection. Repeated 5-OP-RU administration did not render MAIT cells anergic, thus allowing for its inclusion in prime-boost immunization protocols. Mechanistically, tissue MAIT cell accumulation was due to their robust proliferation, as opposed to altered migratory behavior, and required viral vaccine replication competency and Toll-like receptor 3 and type I interferon receptor signaling. The observed phenomenon was reproducible in female and male mice, and in both young and old animals. It could also be recapitulated in a human cell culture system in which peripheral blood mononuclear cells were exposed to replicating virions and 5-OP-RU. In conclusion, although viruses and virus-based vaccines are devoid of the riboflavin biosynthesis machinery that supplies MR1 ligands, targeting MR1 enhances the efficacy of vaccine-elicited antiviral immunity. We propose 5-OP-RU as a non-classic but potent and versatile vaccine adjuvant against respiratory viruses.
Collapse
Affiliation(s)
- Rasheduzzaman Rashu
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Marina Ninkov
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Christine M Wardell
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Jenna M Benoit
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Nicole I Wang
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Courtney E Meilleur
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Michael R D'Agostino
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Ali Zhang
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Emily Feng
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - Nasrin Saeedian
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Gillian I Bell
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, Canada
| | - Fatemeh Vahedi
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
| | - David A Hess
- Krembil Centre for Stem Cell Biology, Molecular Medicine Research Laboratories, Robarts Research Institute, London, Ontario, Canada
- Department of Physiology and Pharmacology, Western University, London, Ontario, Canada
| | - Stephen D Barr
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Ryan M Troyer
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Chil-Yong Kang
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Ali A Ashkar
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Matthew S Miller
- McMaster Immunology Research Centre, McMaster University, Hamilton, Ontario, Canada
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
- Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, Ontario, Canada
| | - S M Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Division of Clinical Immunology and Allergy, Department of Medicine, Western University, London, Ontario, Canada
- Division of General Surgery, Department of Surgery, Western University, London, Ontario, Canada
- Lawson Health Research Institute, London, Ontario, Canada
| |
Collapse
|
21
|
Tarantino N, Litvinova E, Samri A, Soulié C, Morin V, Rousseau A, Dorgham K, Parizot C, Bonduelle O, Beurton A, Miyara M, Ghillani P, Mayaux J, Lhote R, Lacorte JM, Marcelin AG, Amoura Z, Luyt CE, Gorochov G, Guihot A, Vieillard V. Identification of natural killer markers associated with fatal outcome in COVID-19 patients. Front Cell Infect Microbiol 2023; 13:1165756. [PMID: 37342247 PMCID: PMC10277643 DOI: 10.3389/fcimb.2023.1165756] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 05/18/2023] [Indexed: 06/22/2023] Open
Abstract
Introduction Increasing evidence has shown that coronavirus disease 19 (COVID-19) severity is driven by a dysregulated immunological response. Previous studies have demonstrated that natural killer (NK) cell dysfunction underpins severe illness in COVID-19 patients, but have lacked an in-depth analysis of NK cell markers as a driver of death in the most critically ill patients. Methods We enrolled 50 non-vaccinated hospitalized patients infected with the initial virus or the alpha variant of SARS-CoV-2 with moderate or severe illness, to evaluate phenotypic and functional features of NK cells. Results Here, we show that, consistent with previous studies, evolution NK cells from COVID-19 patients are more activated, with the decreased activation of natural cytotoxicity receptors and impaired cytotoxicity and IFN-γ production, in association with disease regardless of the SARS-CoV-2 strain. Fatality was observed in 6 of 17 patients with severe disease; NK cells from all of these patients displayed a peculiar phenotype of an activated memory-like phenotype associated with massive TNF-α production. Discussion These data suggest that fatal COVID-19 infection is driven by an uncoordinated inflammatory response in part mediated by a specific subset of activated NK cells.
Collapse
Affiliation(s)
- Nadine Tarantino
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
| | - Elena Litvinova
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Département d’Immunologie, Paris, France
| | - Assia Samri
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
| | - Cathia Soulié
- Sorbonne Université, Inserm, Institut Pierre Louis d’Epidémiologie et de Santé Publique (iPLESP), Assistance Publique – Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Laboratoire de Virologie, Paris, France
| | - Véronique Morin
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
| | - Alice Rousseau
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
| | - Karim Dorgham
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
| | - Christophe Parizot
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Département d’Immunologie, Paris, France
| | - Olivia Bonduelle
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
| | - Alexandra Beurton
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Service de Médecine Intensive-Réanimation et Pneumologie, Paris, France
- Sorbonne Université, Inserm UMRS Neurophysiologie Respiratoire Expérimentale et Clinique, Assistance Publique – Hôpitaux de Paris (AP-HP), Paris, France
| | - Makoto Miyara
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Département d’Immunologie, Paris, France
| | - Pascale Ghillani
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Département d’Immunologie, Paris, France
| | - Julien Mayaux
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Service de Médecine Intensive-Réanimation et Pneumologie, Paris, France
| | - Raphael Lhote
- Service de Médecine Interne 2, Institut E3M, Assistance Publique – Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Jean-Marc Lacorte
- Sorbonne Université, Inserm, UMRS1166-ICAN Institute of Cardiometabolism and Nutrition, Paris, France
- Service de Biochimie Endocrinienne et Oncologique, Assistance Publique – Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Anne-Geneviève Marcelin
- Sorbonne Université, Inserm, Institut Pierre Louis d’Epidémiologie et de Santé Publique (iPLESP), Assistance Publique – Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Laboratoire de Virologie, Paris, France
| | - Zahir Amoura
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
- Service de Médecine Interne 2, Institut E3M, Assistance Publique – Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Charles-Edouard Luyt
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Service de Médecine Intensive-Réanimation et Pneumologie, Paris, France
- Service de Médecine Interne 2, Institut E3M, Assistance Publique – Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Paris, France
| | - Guy Gorochov
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Département d’Immunologie, Paris, France
| | - Amélie Guihot
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
- Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Pitié-Salpêtrière, Département d’Immunologie, Paris, France
| | - Vincent Vieillard
- Sorbonne Université, Inserm, CNRS, Centre d’Immunologie et des Maladies Infectieuses (CIMI-Paris), Hôpital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
22
|
Opsteen S, Files JK, Fram T, Erdmann N. The role of immune activation and antigen persistence in acute and long COVID. J Investig Med 2023; 71:545-562. [PMID: 36879504 PMCID: PMC9996119 DOI: 10.1177/10815589231158041] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 01/09/2023] [Accepted: 01/24/2023] [Indexed: 03/08/2023]
Abstract
In late 2019, severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) triggered the global coronavirus disease 2019 (COVID-19) pandemic. Although most infections cause a self-limited syndrome comparable to other upper respiratory viral pathogens, a portion of individuals develop severe illness leading to substantial morbidity and mortality. Furthermore, an estimated 10%-20% of SARS-CoV-2 infections are followed by post-acute sequelae of COVID-19 (PASC), or long COVID. Long COVID is associated with a wide variety of clinical manifestations including cardiopulmonary complications, persistent fatigue, and neurocognitive dysfunction. Severe acute COVID-19 is associated with hyperactivation and increased inflammation, which may be an underlying cause of long COVID in a subset of individuals. However, the immunologic mechanisms driving long COVID development are still under investigation. Early in the pandemic, our group and others observed immune dysregulation persisted into convalescence after acute COVID-19. We subsequently observed persistent immune dysregulation in a cohort of individuals experiencing long COVID. We demonstrated increased SARS-CoV-2-specific CD4+ and CD8+ T-cell responses and antibody affinity in patients experiencing long COVID symptoms. These data suggest a portion of long COVID symptoms may be due to chronic immune activation and the presence of persistent SARS-CoV-2 antigen. This review summarizes the COVID-19 literature to date detailing acute COVID-19 and convalescence and how these observations relate to the development of long COVID. In addition, we discuss recent findings in support of persistent antigen and the evidence that this phenomenon contributes to local and systemic inflammation and the heterogeneous nature of clinical manifestations seen in long COVID.
Collapse
Affiliation(s)
- Skye Opsteen
- Division of Infectious Diseases, Department
of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jacob K Files
- Division of Infectious Diseases, Department
of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Tim Fram
- Division of Infectious Diseases, Department
of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Nathan Erdmann
- Division of Infectious Diseases, Department
of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
23
|
Sakulpaisal M, Sothornwit J, Somboonporn W. The effects of exogenous estrogen in women with SAR-CoV-2 infection: a systematic review and meta-analysis. Hum Reprod 2023:7128247. [PMID: 37071891 DOI: 10.1093/humrep/dead074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 03/22/2023] [Indexed: 04/20/2023] Open
Abstract
STUDY QUESTION Does exogenous estrogen use affect COVID-19-related mortality in women? SUMMARY ANSWER Menopausal hormone therapy (MHT) was associated with a lower likelihood of all-cause fatality related to COVID-19 in postmenopausal women (odds ratio (OR) 0.28, 95% CI 0.18, 0.44; 4 studies, 21 517 women) but the combined oral contraceptive pill in premenopausal women did not have a significant effect (OR 1.00, 95% CI 0.42-2.41; 2 studies, 5099 women). WHAT IS KNOWN ALREADY Men are much more likely to die from COVID-19 than women. STUDY DESIGN, SIZE, DURATION In this systematic meta-analysis, a literature search was conducted using the following search terms related toCOVID-19 and estrogen, sex hormones, hormonal replacement, menopause, or contraception. The PubMed, Scopus, Cochrane Library, and EMBASE databases were searched to identify relevant studies published between December 2019 and December 2021. We also searched MedRxiv as a preprint database and reviewed the reference lists of all included studies and clinical trial registries for ongoing clinical studies until December 2021. PARTICIPANTS/MATERIALS, SETTING, METHODS All comparative studies that compared the rates mortality and morbidity (hospitalization, intensive care unit (ICU) admission, and ventilation support) due to COVID-19 in women using exogenous estrogen to a control group of women (nonusers) were included. A review of the studies for inclusion, extraction of data, and assessment of the risk of bias was performed independently by two reviewers. The ROBINS-I tool and the RoB 2 tool were used for bias assessment of the included studies. Pooled odds ratios (ORs) with 95% CIs were calculated using Review Manager V5.4.1. The I2 statistic was used to quantify heterogeneity. The quality of the evidence was assessed using GRADE criteria. MAIN RESULTS AND THE ROLE OF CHANCE After searching the databases, we identified a total of 5310 studies. After removing duplicate records, ineligible studies, and ongoing studies, a total of four cohort studies and one randomized controlled trial comprising 177 809 participants were included in this review. There was a moderate certainty of evidence that MHT was associated with a lower likelihood of all-cause fatality related to COVID-19 (OR 0.28, 95% CI 0.18, 0.44; I2 = 0%; 4 studies, 21 517 women). The review indicated a low certainty of evidence for other outcomes. The mortality rate of premenopausal women in the combined oral contraceptive pill group did not differ significantly from the control group (OR 1.00, 95% CI 0.42-2.41; 2 studies, 5099 women). MHT marginally increased the rate of hospitalization and ICU admission (OR 1.37, 95% CI 1.18-1.61; 3 studies, 151 485 women), but there was no significant difference in the need for respiratory support between MHT users and nonusers (OR 0.91, 95% CI 0.52-1.59; 3 studies, 151 485 women). Overall, the tendency and magnitude of the effects of MHT in postmenopausal women with COVID-19 were consistent across the included studies. LIMITATIONS, REASONS FOR CAUTION The certainty of the evidence for other outcomes of this review may be limited, as all included studies were cohort studies. In addition, the dosages and durations of exogenous estrogen used by postmenopausal women varied from study to study, and combined progestogen administration may have had some effect on the outcomes. WIDER IMPLICATIONS OF THE FINDINGS The findings of this study can aid in counseling postmenopausal women taking MHT when they are diagnosed with COVID, as they have a lower chance of death than those not taking MHT. STUDY FUNDING/COMPETING INTEREST(S) Khon Kaen University provided financial support for this review and had no involvement at any stage of the study. The authors have no conflicts of interest to declare. REGISTRATION NUMBER PROSPERO, CRD42021271882.
Collapse
Affiliation(s)
- Maytha Sakulpaisal
- Reproductive Medicine Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Jen Sothornwit
- Reproductive Medicine Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Woraluk Somboonporn
- Reproductive Medicine Unit, Department of Obstetrics and Gynecology, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
24
|
Sun Y, Tao Q, Cao Y, Yang T, Zhang L, Luo Y, Wang L. Kaempferol has potential anti-coronavirus disease 2019 (COVID-19) targets based on bioinformatics analyses and pharmacological effects on endotoxin-induced cytokine storm. Phytother Res 2023. [PMID: 36726236 DOI: 10.1002/ptr.7740] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 02/03/2023]
Abstract
COVID-19 has infected 272 million patients and caused 5.33 million deaths around the world, and it remains the main global threat. Previous studies revealed that Chinese traditional medicine is an effective treatment for COVID-19 infection. This study aims to reveal the pharmacological effects of kaempferol, which is the active component of Radix Bupleuri and Tripterygii Radix, and potential mechanisms for the treatment of COVID-19. Here, we employed the bioinformatics methods to filter the anti-COVID-19 candidate genes of kaempferol, which mainly enriched in inflammation (TNF, JUN, etc.) and virus infection (AKT1, JNK, etc.). The Transcription levels of AKT1, JNK and JUN were significantly reduced by kaempferol treatment in the LPS-activated macrophages. In addition, kaempferol reduced the secretion of inflammatory factors by LPS-stimulated macrophages, inhibited MAPK/NF-κB signaling and regulated macrophage polarization to M2 type in vitro, and suppressed endotoxin-induced cytokine storm and improved survival in mice. Molecular docking analysis demonstrated that kaempferol was probable to bind the COVID-19 protein 5R84 and formatted hydrogen bond with the residues, the free binding energy of which was lower than the original ligand. In summary, our current work indicates that kaempferol has anti-COVID-19 potential through the reduction of COVID-19-induced body dysfunction and molecule-protein interaction, and bioinformatics results clarify that some of these key target genes might serve as potential molecular markers for detecting COVID-19.
Collapse
Affiliation(s)
- Yaoxiang Sun
- Department of Clinical Laboratory, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Qing Tao
- Center for Translational Medicine and Jiangsu Key Laboratory of Molecular Medicine, Medical School of Nanjing University, Nanjing, China
| | - Yang Cao
- College of Arts & Science, Vanderbilt University, Nashville, Tennessee, USA
| | - Tingting Yang
- Department of Clinical Laboratory, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Ling Zhang
- Department of Clinical Laboratory, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Yifeng Luo
- Department of Clinical Laboratory, The Affiliated Yixing Hospital of Jiangsu University, Yixing, China
| | - Lei Wang
- Department of Clinical Laboratory, Jiangsu Province hospital on Integration of Chinese and Western Medicine, Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing, China
| |
Collapse
|
25
|
dos Santos AAC, Rodrigues LE, Alecrim-Zeza AL, de Araújo Ferreira L, Trettel CDS, Gimenes GM, da Silva AF, Sousa-Filho CPB, Serdan TDA, Levada-Pires AC, Hatanaka E, Borges FT, de Barros MP, Cury-Boaventura MF, Bertolini GL, Cassolla P, Marzuca-Nassr GN, Vitzel KF, Pithon-Curi TC, Masi LN, Curi R, Gorjao R, Hirabara SM. Molecular and cellular mechanisms involved in tissue-specific metabolic modulation by SARS-CoV-2. Front Microbiol 2022; 13:1037467. [PMID: 36439786 PMCID: PMC9684198 DOI: 10.3389/fmicb.2022.1037467] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 10/26/2022] [Indexed: 09/09/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is triggered by the SARS-CoV-2, which is able to infect and cause dysfunction not only in lungs, but also in multiple organs, including central nervous system, skeletal muscle, kidneys, heart, liver, and intestine. Several metabolic disturbances are associated with cell damage or tissue injury, but the mechanisms involved are not yet fully elucidated. Some potential mechanisms involved in the COVID-19-induced tissue dysfunction are proposed, such as: (a) High expression and levels of proinflammatory cytokines, including TNF-α IL-6, IL-1β, INF-α and INF-β, increasing the systemic and tissue inflammatory state; (b) Induction of oxidative stress due to redox imbalance, resulting in cell injury or death induced by elevated production of reactive oxygen species; and (c) Deregulation of the renin-angiotensin-aldosterone system, exacerbating the inflammatory and oxidative stress responses. In this review, we discuss the main metabolic disturbances observed in different target tissues of SARS-CoV-2 and the potential mechanisms involved in these changes associated with the tissue dysfunction.
Collapse
Affiliation(s)
| | - Luiz Eduardo Rodrigues
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Amanda Lins Alecrim-Zeza
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Liliane de Araújo Ferreira
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Caio dos Santos Trettel
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Gabriela Mandú Gimenes
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Adelson Fernandes da Silva
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | | | - Tamires Duarte Afonso Serdan
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
- Department of Molecular Pathobiology, University of New York, New York, NY, United States
| | - Adriana Cristina Levada-Pires
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Elaine Hatanaka
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Fernanda Teixeira Borges
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
- Divisão de Nefrologia, Departamento de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Marcelo Paes de Barros
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Maria Fernanda Cury-Boaventura
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Gisele Lopes Bertolini
- Department of Physiological Sciences, Biological Science Center, State University of Londrina, Londrina, PR, Brazil
| | - Priscila Cassolla
- Department of Physiological Sciences, Biological Science Center, State University of Londrina, Londrina, PR, Brazil
| | | | - Kaio Fernando Vitzel
- School of Health Sciences, College of Health, Massey University, Auckland, New Zealand
| | - Tania Cristina Pithon-Curi
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Laureane Nunes Masi
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Rui Curi
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
- Instituto Butantan, São Paulo, Brazil
| | - Renata Gorjao
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| | - Sandro Massao Hirabara
- Programa de Pós-graduação Interdisciplinar em Ciências da Saúde, Universidade Cruzeiro do Sul, São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Hackstein CP, Klenerman P. Emerging features of MAIT cells and other unconventional T cell populations in human viral disease and vaccination. Semin Immunol 2022; 61-64:101661. [PMID: 36374780 PMCID: PMC10933818 DOI: 10.1016/j.smim.2022.101661] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 09/30/2022] [Accepted: 10/03/2022] [Indexed: 12/14/2022]
Abstract
MAIT cells are one representative of a group of related unconventional or pre-set T cells, and are particularly abundant in humans. While these unconventional T cell types, which also include populations of Vδ2 cells and iNKT cells, recognise quite distinct ligands, they share functional features including the ability to sense "danger" by integration of cytokine signals. Since such signals are common to many human pathologies, activation of MAIT cells in particular has been widely observed. In this review we will discuss recent trends in these data, for example the findings from patients with Covid-19 and responses to novel vaccines. Covid-19 is an example where MAIT cell activation has been correlated with disease severity by several groups, and the pathways leading to activation are being clarified, but the overall role of the cells in vivo requires further exploration. Given the potential wide functional responsiveness of these cells, which ranges from tissue repair to cytotoxicity, and likely impacts on the activity of many other cell populations, defining the role of these cells - not only as sensitive biomarkers but also as mediators - across human disease remains an important task.
Collapse
Affiliation(s)
- Carl-Philipp Hackstein
- Peter Medawar Building for Pathogen Research, Nuffield Dept of Medicine, University of Oxford, Oxford OX1 3SY, UK; Translational Gastroenterology Unit, Nuffield Dept of Medicine, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Nuffield Dept of Medicine, University of Oxford, Oxford OX1 3SY, UK; Translational Gastroenterology Unit, Nuffield Dept of Medicine, John Radcliffe Hospital, Oxford OX3 9DU, UK.
| |
Collapse
|
27
|
Role of Innate and Adaptive Cytokines in the Survival of COVID-19 Patients. Int J Mol Sci 2022; 23:ijms231810344. [PMID: 36142255 PMCID: PMC9499609 DOI: 10.3390/ijms231810344] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 09/02/2022] [Accepted: 09/02/2022] [Indexed: 11/16/2022] Open
Abstract
SARS-CoV-2 is a new coronavirus characterized by a high infection and transmission capacity. A significant number of patients develop inadequate immune responses that produce massive releases of cytokines that compromise their survival. Soluble factors are clinically and pathologically relevant in COVID-19 survival but remain only partially characterized. The objective of this work was to simultaneously study 62 circulating soluble factors, including innate and adaptive cytokines and their soluble receptors, chemokines and growth and wound-healing/repair factors, in severe COVID-19 patients who survived compared to those with fatal outcomes. Serum samples were obtained from 286 COVID-19 patients and 40 healthy controls. The 62 circulating soluble factors were quantified using a Luminex Milliplex assay. Results. The patients who survived had decreased levels of the following 30 soluble factors of the 62 studied compared to those with fatal outcomes, therefore, these decreases were observed for cytokines and receptors predominantly produced by the innate immune system—IL-1α, IL-1α, IL-18, IL-15, IL-12p40, IL-6, IL-27, IL-1Ra, IL-1RI, IL-1RII, TNFα, TGFα, IL-10, sRAGE, sTNF-RI and sTNF-RII—for the chemokines IL-8, IP-10, MCP-1, MCP-3, MIG and fractalkine; for the growth factors M-CSF and the soluble receptor sIL2Ra; for the cytokines involved in the adaptive immune system IFNγ, IL-17 and sIL-4R; and for the wound-repair factor FGF2. On the other hand, the patients who survived had elevated levels of the soluble factors TNFβ, sCD40L, MDC, RANTES, G-CSF, GM-CSF, EGF, PDGFAA and PDGFABBB compared to those who died. Conclusions. Increases in the circulating levels of the sCD40L cytokine; MDC and RANTES chemokines; the G-CSF and GM-CSF growth factors, EGF, PDGFAA and PDGFABBB; and tissue-repair factors are strongly associated with survival. By contrast, large increases in IL-15, IL-6, IL-18, IL-27 and IL-10; the sIL-1RI, sIL1RII and sTNF-RII receptors; the MCP3, IL-8, MIG and IP-10 chemokines; the M-CSF and sIL-2Ra growth factors; and the wound-healing factor FGF2 favor fatal outcomes of the disease.
Collapse
|
28
|
Brlić PK, Pavletić M, Lerga M, Krstanović F, Matešić MP, Miklić K, Malić S, Mikša L, Pajcur M, Peruč D, Schubert M, Bertoglio F, Arapović J, Protić A, Šustić A, Milošević M, Šain LČ, Jonjić S, Lisnić VJ, Brizić I. SARS-CoV-2 Spike and Nucleocapsid Antibody Response in Vaccinated Croatian Healthcare Workers and Infected Hospitalized Patients: A Single Center Cohort Study. Viruses 2022; 14:1966. [PMID: 36146773 PMCID: PMC9503044 DOI: 10.3390/v14091966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 08/31/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
Studies assessing the dynamics and duration of antibody responses following SARS-CoV-2 infection or vaccination are an invaluable tool for vaccination schedule planning, assessment of risk groups and management of pandemics. In this study, we developed and employed ELISA assays to analyze the humoral responses to Nucleocapsid and Spike proteins in vaccinated health-care workers (HCW) and critically ill COVID-19 patients. Sera of more than 1000 HCWs and critically ill patients from the Clinical Hospital Center Rijeka were tested across a one-year period, encompassing the spread of major SARS-CoV-2 variants of concern (VOCs). We observed 97% of seroconversion in HCW cohort as well as sustained anti-Spike antibody response in vaccinees for more than 6 months. In contrast, the infection-induced anti-Nucleocapsid response was waning significantly in a six-month period. Furthermore, a substantial decrease in vaccinees' anti-Spike antibodies binding to Spike protein of Omicron VOC was also observed. Critically ill COVID-19 patients had higher levels of anti-Spike and anti-Nucleocapsid antibodies compared to HCWs. No significant differences in anti-Spike and anti-Nucleocapsid antibody levels between the critically ill COVID-19 patients that were on non-invasive oxygen supplementation and those on invasive ventilation support were observed. However, stronger anti-Spike, but not anti-Nucleocapsid, antibody response correlated with a better disease outcome in the cohort of patients on invasive ventilation support. Altogether, our results contribute to the growing pool of data on humoral responses to SARS-CoV-2 infection and vaccination.
Collapse
Affiliation(s)
- Paola Kučan Brlić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Martina Pavletić
- Emergency Department, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Mate Lerga
- Emergency Department, Clinical Hospital Center Rijeka, 51000 Rijeka, Croatia
| | - Fran Krstanović
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Marina Pribanić Matešić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Karmela Miklić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Suzana Malić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Leonarda Mikša
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Maja Pajcur
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Dolores Peruč
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Maren Schubert
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Federico Bertoglio
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Biotechnologie, Technische Universität Braunschweig, Spielmannstr. 7, 38106 Braunschweig, Germany
| | - Jurica Arapović
- Faculty of Medicine, University of Mostar, Bijeli Brijeg b.b., 88000 Mostar, Bosnia and Herzegovina
| | - Alen Protić
- Department of Anesthesiology, Reanimation, Intensive Care and Emergency Medicine, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Alan Šustić
- Department of Anesthesiology, Reanimation, Intensive Care and Emergency Medicine, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
- Department of Clinical Medical Science II, Faculty of Health Studies, University of Rijeka, 51000 Rijeka, Croatia
| | - Marko Milošević
- Department of Anesthesiology, Reanimation, Intensive Care and Emergency Medicine, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Luka Čičin Šain
- Helmholtz Center for Infection Research, Department of Viral Immunology, 38124 Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover/Braunschweig, 38124 Braunschweig, Germany
- Centre for Individualised Infection Medicine (CiiM), Joint Venture of Helmholtz Centre for Infection Research and Hannover Medical School, 30625 Hannover, Germany
| | - Stipan Jonjić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Vanda Juranić Lisnić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| | - Ilija Brizić
- Center for Proteomics, Faculty of Medicine, University of Rijeka, Braće Branchetta 20, 51000 Rijeka, Croatia
| |
Collapse
|
29
|
Olea B, Albert E, Giménez E, Torres I, Amat P, Remigia MJ, Alberola J, Carbonell N, Ferreres J, Blasco ML, Navarro D. SARS-CoV-2-reactive IFN-γ-producing CD4 + and CD8 + T cells in blood do not correlate with clinical severity in unvaccinated critically ill COVID-19 patients. Sci Rep 2022; 12:14271. [PMID: 35995830 PMCID: PMC9395536 DOI: 10.1038/s41598-022-18659-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 08/17/2022] [Indexed: 11/18/2022] Open
Abstract
We examined the relationship between peripheral blood levels of SARS-CoV-2 S (Spike protein)1/M (Membrane protein)-reactive IFN-γ-producing CD4+ and CD8+ T cells, serum levels of biomarkers of clinical severity, and mortality in critically ill COVID-19 patients. The potential association between SARS-CoV-2-S-Receptor Binding Domain (RBD)-specific IgG levels in sera and mortality was also investigated. SARS-CoV-2 T cells and anti-RBD IgG levels were monitored in 71 non-consecutive patients (49 male and 22 female; median age, 65 years) by whole-blood flow cytometry and Enzyme-linked immunosorbent assay (ELISA), respectively (326 specimens). SARS-CoV-2 RNA loads in paired tracheal aspirates [TA] (n = 147) were available from 54 patients. Serum levels of interleukin-6, ferritin, D-Dimer, lactose dehydrogenase and C-reactive protein in paired sera were known. SARS-CoV-2 T cells (either CD4+, CD8+ or both) were detectable in 70 patients. SARS-CoV-2 IFN-γ CD4+ T-cell responses were documented more frequently than their CD8+ counterparts (62 vs. 56 patients) and were of greater magnitude overall. Detectable SARS-CoV-2 S1/M-reactive CD8+ and CD4+ T-cell responses were associated with higher SARS-CoV-2 RNA loads in TA. SARS-CoV-2 RNA load in TA decreased over time, irrespective of the dynamics of SARS-CoV-2-reactive CD8+ and CD4+ T cells. No correlation was found between SARS-CoV-2 IFN-γ T-cell counts, anti-RBD IgG concentrations and biomarker serum levels (Rho ≤ 0.3). The kinetics of both T cell subsets was comparable between those who died or survived, whereas anti-RBD IgG levels were higher across different time points in deceased patients than in survivors. Enumeration of peripheral blood levels of SARS-CoV-2-S1/M-reactive IFN-γ CD4+ and CD8+ T cells does not predict viral clearance from the lower respiratory tract or poor clinical outcomes in critically ill COVID-19 patients. In contrast, anti-RBD IgG levels were directly associated with increased mortality.
Collapse
Affiliation(s)
- Beatriz Olea
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Eliseo Albert
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Estela Giménez
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Ignacio Torres
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Paula Amat
- Hematology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - María José Remigia
- Hematology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - Juan Alberola
- Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 17, 46010, Valencia, Spain
| | - Nieves Carbonell
- Medical Intensive Care Unit, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - José Ferreres
- Medical Intensive Care Unit, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - María Luisa Blasco
- Medical Intensive Care Unit, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain
| | - David Navarro
- Microbiology Service, Clinic University Hospital, INCLIVA Health Research Institute, Valencia, Spain. .,Department of Microbiology, School of Medicine, University of Valencia, Av. Blasco Ibáñez 17, 46010, Valencia, Spain.
| |
Collapse
|
30
|
Agrati C, Carsetti R, Bordoni V, Sacchi A, Quintarelli C, Locatelli F, Ippolito G, Capobianchi MR. The immune response as a double-edged sword: the lesson learnt during the COVID-19 pandemic. Immunology 2022; 167:287-302. [PMID: 35971810 PMCID: PMC9538066 DOI: 10.1111/imm.13564] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 08/06/2022] [Indexed: 11/29/2022] Open
Abstract
The COVID‐19 pandemic has represented an unprecedented challenge for the humanity, and scientists around the world provided a huge effort to elucidate critical aspects in the fight against the pathogen, useful in designing public health strategies, vaccines and therapeutic approaches. One of the first pieces of evidence characterizing the SARS‐CoV‐2 infection has been its breadth of clinical presentation, ranging from asymptomatic to severe/deadly disease, and the indication of the key role played by the immune response in influencing disease severity. This review is aimed at summarizing what the SARS‐CoV‐2 infection taught us about the immune response, highlighting its features of a double‐edged sword mediating both protective and pathogenic processes. We will discuss the protective role of soluble and cellular innate immunity and the detrimental power of a hyper‐inflammation‐shaped immune response, resulting in tissue injury and immunothrombotic events. We will review the importance of B‐ and T‐cell immunity in reducing the clinical severity and their ability to cross‐recognize viral variants.
Collapse
Affiliation(s)
- Chiara Agrati
- Laboratory of Cellular Immunology, INMI L. Spallanzani, IRCCS
| | - Rita Carsetti
- B cell laboratory, Immunology Research Area, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | | | - Alessandra Sacchi
- Molecular Virology and antimicrobial immunity Laboratory, Department of Science, Roma Tre University, Rome, Italy
| | - Concetta Quintarelli
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy.,Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS
| | - Franco Locatelli
- Department of Hematology/Oncology and Cell and Gene Therapy, Bambino Gesù Children Hospital, IRCCS.,Department of Pediatrics, Catholic University of Sacred Heart, Rome, Italy
| | - Giuseppe Ippolito
- General Directorate for Research and Health Innovation, Italian Ministry of Health
| | - Maria R Capobianchi
- Sacro Cuore Don Calabria Hospital IRCCS, Negrar di Valpolicella (Verona).,Saint Camillus International University of Health Sciences, Rome
| |
Collapse
|
31
|
Giannella A, Riccetti S, Sinigaglia A, Piubelli C, Razzaboni E, Di Battista P, Agostini M, Dal Molin E, Manganelli R, Gobbi F, Ceolotto G, Barzon L. Circulating microRNA signatures associated with disease severity and outcome in COVID-19 patients. Front Immunol 2022; 13:968991. [PMID: 36032130 PMCID: PMC9403711 DOI: 10.3389/fimmu.2022.968991] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 07/20/2022] [Indexed: 12/14/2022] Open
Abstract
Background SARS-CoV-2 induces a spectrum of clinical conditions ranging from asymptomatic infection to life threatening severe disease. Host microRNAs have been involved in the cytokine storm driven by SARS-CoV-2 infection and proposed as candidate biomarkers for COVID-19. Methods To discover signatures of circulating miRNAs associated with COVID-19, disease severity and mortality, small RNA-sequencing was performed on serum samples collected from 89 COVID-19 patients (34 severe, 29 moderate, 26 mild) at hospital admission and from 45 healthy controls (HC). To search for possible sources of miRNAs, investigation of differentially expressed (DE) miRNAs in relevant human cell types in vitro. Results COVID-19 patients showed upregulation of miRNAs associated with lung disease, vascular damage and inflammation and downregulation of miRNAs that inhibit pro-inflammatory cytokines and chemokines, angiogenesis, and stress response. Compared with mild/moderate disease, patients with severe COVID-19 had a miRNA signature indicating a profound impairment of innate and adaptive immune responses, inflammation, lung fibrosis and heart failure. A subset of the DE miRNAs predicted mortality. In particular, a combination of high serum miR-22-3p and miR-21-5p, which target antiviral response genes, and low miR-224-5p and miR-155-5p, targeting pro-inflammatory factors, discriminated severe from mild/moderate COVID-19 (AUROC 0.88, 95% CI 0.80-0.95, p<0.0001), while high leukocyte count and low levels of miR-1-3p, miR-23b-3p, miR-141-3p, miR-155-5p and miR-4433b-5p predicted mortality with high sensitivity and specificity (AUROC 0.95, 95% CI 0.89-1.00, p<0.0001). In vitro experiments showed that some of the DE miRNAs were modulated directly by SARS-CoV-2 infection in permissive lung epithelial cells. Conclusions We discovered circulating miRNAs associated with COVID-19 severity and mortality. The identified DE miRNAs provided clues on COVID-19 pathogenesis, highlighting signatures of impaired interferon and antiviral responses, inflammation, organ damage and cardiovascular failure as associated with severe disease and death.
Collapse
Affiliation(s)
| | - Silvia Riccetti
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Chiara Piubelli
- Department of Infectious-Tropical Diseases and Microbiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Sacro Cuore Don Calabria Hospital, Verona, Italy
| | - Elisa Razzaboni
- Department of Infectious-Tropical Diseases and Microbiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Sacro Cuore Don Calabria Hospital, Verona, Italy
| | - Piero Di Battista
- Maternal and Child Health Department, University of Padova, Padova, Italy
| | - Matteo Agostini
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | | | - Riccardo Manganelli
- Department of Molecular Medicine, University of Padova, Padova, Italy
- Microbiology and Virology Unit, Padova University Hospital, Padova, Italy
| | - Federico Gobbi
- Department of Infectious-Tropical Diseases and Microbiology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Sacro Cuore Don Calabria Hospital, Verona, Italy
| | | | - Luisa Barzon
- Department of Molecular Medicine, University of Padova, Padova, Italy
- Microbiology and Virology Unit, Padova University Hospital, Padova, Italy
| |
Collapse
|
32
|
Singh K, Cogan S, Elekes S, Murphy DM, Cummins S, Curran R, Najda Z, Dunne MR, Jameson G, Gargan S, Martin S, Long A, Doherty DG. SARS-CoV-2 spike and nucleocapsid proteins fail to activate human dendritic cells or γδ T cells. PLoS One 2022; 17:e0271463. [PMID: 35834480 PMCID: PMC9282473 DOI: 10.1371/journal.pone.0271463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/30/2022] [Indexed: 11/20/2022] Open
Abstract
γδ T cells are thought to contribute to immunity against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), but the mechanisms by which they are activated by the virus are unknown. Using flow cytometry, we investigated if the two most abundant viral structural proteins, spike and nucleocapsid, can activate human γδ T cell subsets, directly or in the presence of dendritic cells (DC). Both proteins failed to induce interferon-γ production by Vδ1 or Vδ2 T cells within fresh mononuclear cells or lines of expanded γδ T cells generated from healthy donors, but the same proteins stimulated CD3+ cells from COVID-19 patients. The nucleocapsid protein stimulated interleukin-12 production by DC and downstream interferon-γ production by co-cultured Vδ1 and Vδ2 T cells, but protease digestion and use of an alternative nucleocapsid preparation indicated that this activity was due to contaminating non-protein material. Thus, SARS-CoV-2 spike and nucleocapsid proteins do not have stimulatory activity for DC or γδ T cells. We propose that γδ T cell activation in COVID-19 patients is mediated by immune recognition of viral RNA or other structural proteins by γδ T cells, or by other immune cells, such as DC, that produce γδ T cell-stimulatory ligands or cytokines.
Collapse
Affiliation(s)
- Kiran Singh
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Sita Cogan
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Stefan Elekes
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Dearbhla M. Murphy
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Sinead Cummins
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Rory Curran
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Zaneta Najda
- Molecular Cell Biology Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Margaret R. Dunne
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Gráinne Jameson
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Siobhan Gargan
- Discipline of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Seamus Martin
- Molecular Cell Biology Laboratory, Smurfit Institute of Genetics, Trinity College Dublin, Dublin, Ireland
| | - Aideen Long
- Discipline of Clinical Medicine, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| | - Derek G. Doherty
- Discipline of Immunology, Trinity Translational Medicine Institute, Trinity College Dublin, St. James’s Hospital, Dublin, Ireland
| |
Collapse
|
33
|
McNaughton AL, Paton RS, Edmans M, Youngs J, Wellens J, Phalora P, Fyfe A, Belij-Rammerstorfer S, Bolton JS, Ball J, Carnell GW, Dejnirattisai W, Dold C, Eyre DW, Hopkins P, Howarth A, Kooblall K, Klim H, Leaver S, Lee LN, López-Camacho C, Lumley SF, Macallan DC, Mentzer AJ, Provine NM, Ratcliff J, Slon-Compos J, Skelly D, Stolle L, Supasa P, Temperton N, Walker C, Wang B, Wyncoll D, Oxford Protective T Cell Immunology for COVID-19 (OPTIC) consortium, Scottish National Blood Transfusion Service (SNBTS) consortium, Simmonds P, Lambe T, Baillie JK, Semple MG, Openshaw PJ, International Severe Acute Respiratory and emerging Infection Consortium Coronavirus Clinical Characterisation Consortium (ISARIC4C) investigators, Obolski U, Turner M, Carroll M, Mongkolsapaya J, Screaton G, Kennedy SH, Jarvis L, Barnes E, Dunachie S, Lourenço J, Matthews PC, Bicanic T, Klenerman P, Gupta S, Thompson CP. Fatal COVID-19 outcomes are associated with an antibody response targeting epitopes shared with endemic coronaviruses. JCI Insight 2022; 7:156372. [PMID: 35608920 PMCID: PMC9310533 DOI: 10.1172/jci.insight.156372] [Citation(s) in RCA: 18] [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: 11/09/2021] [Accepted: 05/18/2022] [Indexed: 11/17/2022] Open
Abstract
The role of immune responses to previously seen endemic coronavirus epitopes in severe acute respiratory coronavirus 2 (SARS-CoV-2) infection and disease progression has not yet been determined. Here, we show that a key characteristic of fatal outcomes with coronavirus disease 2019 (COVID-19) is that the immune response to the SARS-CoV-2 spike protein is enriched for antibodies directed against epitopes shared with endemic beta-coronaviruses and has a lower proportion of antibodies targeting the more protective variable regions of the spike. The magnitude of antibody responses to the SARS-CoV-2 full-length spike protein, its domains and subunits, and the SARS-CoV-2 nucleocapsid also correlated strongly with responses to the endemic beta-coronavirus spike proteins in individuals admitted to an intensive care unit (ICU) with fatal COVID-19 outcomes, but not in individuals with nonfatal outcomes. This correlation was found to be due to the antibody response directed at the S2 subunit of the SARS-CoV-2 spike protein, which has the highest degree of conservation between the beta-coronavirus spike proteins. Intriguingly, antibody responses to the less cross-reactive SARS-CoV-2 nucleocapsid were not significantly different in individuals who were admitted to an ICU with fatal and nonfatal outcomes, suggesting an antibody profile in individuals with fatal outcomes consistent with an "original antigenic sin" type response.
Collapse
Affiliation(s)
- Anna L. McNaughton
- Peter Medawar Building for Pathogen Research
- Nuffield Department of Medicine, and
| | - Robert S. Paton
- Peter Medawar Building for Pathogen Research
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Matthew Edmans
- Peter Medawar Building for Pathogen Research
- Nuffield Department of Medicine, and
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Jonathan Youngs
- Institute of Infection & Immunity, St George’s University of London, London, United Kingdom
| | - Judith Wellens
- Peter Medawar Building for Pathogen Research
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
- Translational Research for Gastrointestinal Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Prabhjeet Phalora
- Peter Medawar Building for Pathogen Research
- Nuffield Department of Medicine, and
| | - Alex Fyfe
- Peter Medawar Building for Pathogen Research
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | | | - Jai S. Bolton
- Peter Medawar Building for Pathogen Research
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Jonathan Ball
- General Intensive Care service, St George’s University Hospital National Health Service (NHS) Trust, London, United Kingdom
| | - George W. Carnell
- Department of Veterinary Medicine, University of Cambridge, Cambridge, United Kingdom
| | | | | | - David W. Eyre
- Nuffield Department of Population Health, University of Oxford, Oxford, United Kingdom
| | - Philip Hopkins
- Centre for Human & Applied Physiological Sciences, School of Basic & Medical Biosciences, Faculty of Life Sciences & Medicine, King’s College, London, United Kingdom
| | - Alison Howarth
- Department of Microbiology/Infectious Diseases, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Kreepa Kooblall
- Oxford Centre for Diabetes, Endocrinology and Metabolism, Churchill Hospital, and
| | - Hannah Klim
- Peter Medawar Building for Pathogen Research
- Department of Zoology, University of Oxford, Oxford, United Kingdom
- Future of Humanity Institute, Department of Philosophy, and
| | - Susannah Leaver
- General Intensive Care service, St George’s University Hospital National Health Service (NHS) Trust, London, United Kingdom
| | - Lian Ni Lee
- Peter Medawar Building for Pathogen Research
- Nuffield Department of Medicine, and
| | | | - Sheila F. Lumley
- Peter Medawar Building for Pathogen Research
- Nuffield Department of Medicine, and
- Department of Microbiology/Infectious Diseases, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Derek C. Macallan
- Institute of Infection & Immunity, St George’s University of London, London, United Kingdom
| | | | - Nicholas M. Provine
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Jeremy Ratcliff
- Peter Medawar Building for Pathogen Research
- Nuffield Department of Medicine, and
| | - Jose Slon-Compos
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine
| | - Donal Skelly
- Peter Medawar Building for Pathogen Research
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
- Oxford University Hospitals NHS Foundation Trust, Oxford, United Kingdom
| | - Lucas Stolle
- Department of Biochemistry, University of Oxford, Oxford, United Kingdom
| | - Piyada Supasa
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, University of Kent, Chatham, United Kingdom
| | - Chris Walker
- Meso Scale Diagnostics, Rockville, Maryland, USA
| | - Beibei Wang
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine
| | - Duncan Wyncoll
- Intensive Care Medicine, Guy’s and St Thomas’ Hospital NHS Foundation Trust, London, United Kingdom
| | | | | | - Peter Simmonds
- Peter Medawar Building for Pathogen Research
- Nuffield Department of Medicine, and
| | - Teresa Lambe
- The Jenner Institute Laboratories, University of Oxford, Oxford, United Kingdom
| | | | - Malcolm G. Semple
- NIHR Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, Faculty of Health and Life Sciences, University of Liverpool, Liverpool, United Kingdom
| | | | | | - Uri Obolski
- School of Public Health, Faculty of Medicine, and
- Porter School of the Environment and Earth Sciences, Faculty of Exact Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Marc Turner
- National Microbiology Reference Unit, Scottish National Blood Transfusion Service, Edinburgh, United Kingdom
| | - Miles Carroll
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine
- National Infection Service, Public Health England (PHE), Salisbury, United Kingdom
| | - Juthathip Mongkolsapaya
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine
- Siriraj Center of Research for Excellence in Dengue & Emerging Pathogens, Faculty of Medicine Siriraj Hospital, Mahidol University, Thailand
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Gavin Screaton
- Wellcome Centre for Human Genetics, Nuffield Department of Medicine
- Chinese Academy of Medical Science (CAMS) Oxford Institute (COI), University of Oxford, Oxford, United Kingdom
| | - Stephen H. Kennedy
- Nuffield Department of Women’s & Reproductive Health, University of Oxford, John Radcliffe Hospital, Oxford, United Kingdom
| | - Lisa Jarvis
- National Microbiology Reference Unit, Scottish National Blood Transfusion Service, Edinburgh, United Kingdom
| | - Eleanor Barnes
- Peter Medawar Building for Pathogen Research
- Nuffield Department of Medicine, and
- Translational Gastroenterology Unit, Experimental Medicine Division, Nuffield Department of Medicine, John Radcliffe Hospital, Oxford, United Kingdom
| | - Susanna Dunachie
- Peter Medawar Building for Pathogen Research
- Department of Microbiology/Infectious Diseases, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - José Lourenço
- Peter Medawar Building for Pathogen Research
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Philippa C. Matthews
- Peter Medawar Building for Pathogen Research
- Nuffield Department of Medicine, and
- Department of Microbiology/Infectious Diseases, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, United Kingdom
| | - Tihana Bicanic
- Institute of Infection & Immunity, St George’s University of London, London, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research
- Nuffield Department of Medicine, and
- Translational Research for Gastrointestinal Diseases, University Hospitals Leuven, Leuven, Belgium
| | - Sunetra Gupta
- Peter Medawar Building for Pathogen Research
- Department of Zoology, University of Oxford, Oxford, United Kingdom
| | - Craig P. Thompson
- Peter Medawar Building for Pathogen Research
- Department of Zoology, University of Oxford, Oxford, United Kingdom
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| |
Collapse
|
34
|
Rovito R, Augello M, Ben-Haim A, Bono V, d'Arminio Monforte A, Marchetti G. Hallmarks of Severe COVID-19 Pathogenesis: A Pas de Deux Between Viral and Host Factors. Front Immunol 2022; 13:912336. [PMID: 35757770 PMCID: PMC9231592 DOI: 10.3389/fimmu.2022.912336] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 05/02/2022] [Indexed: 12/15/2022] Open
Abstract
Two years into Coronavirus Disease 2019 (COVID-19) pandemic, a comprehensive characterization of the pathogenesis of severe and critical forms of COVID-19 is still missing. While a deep dysregulation of both the magnitude and functionality of innate and adaptive immune responses have been described in severe COVID-19, the mechanisms underlying such dysregulations are still a matter of scientific debate, in turn hampering the identification of new therapies and of subgroups of patients that would most benefit from individual clinical interventions. Here we review the current understanding of viral and host factors that contribute to immune dysregulation associated with COVID-19 severity in the attempt to unfold and broaden the comprehension of COVID-19 pathogenesis and to define correlates of protection to further inform strategies of targeted therapeutic interventions.
Collapse
Affiliation(s)
- Roberta Rovito
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Matteo Augello
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Assaf Ben-Haim
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Valeria Bono
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Antonella d'Arminio Monforte
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| | - Giulia Marchetti
- Clinic of Infectious Diseases and Tropical Medicine, Azienda Socio Sanitaria Territoriale (ASST) Santi Paolo e Carlo, Department of Health Sciences, University of Milan, Milan, Italy
| |
Collapse
|
35
|
Ljunggren H, Heggernes Ask E, Cornillet M, Strunz B, Chen P, Rao Muvva J, Akber M, Buggert M, Chambers BJ, Cuapio Gomez A, Dzidic M, Filipovic I, Flodström‐Tullberg M, Garcia M, Gorin J, Gredmark‐Russ S, Hertwig L, Klingström J, Kokkinou E, Kvedaraite E, Lourda M, Mjösberg J, Maucourant C, Norrby‐Teglund A, Palma Medina LM, Parrot T, Perez‐Potti A, Ponzetta A, Ringqvist E, Rivera‐Ballesteros O, Rooyackers O, Sandberg JK, Sandberg JT, Sekine T, Svensson M, Varnaite R, Wullimann D, Karolinska KI/K COVID‐19 Study Group, Eriksson LI, Aleman S, Malmberg K, Strålin K, Björkström NK. The Karolinska KI/K COVID-19 Immune Atlas: An open resource for immunological research and educational purposes. Scand J Immunol 2022; 96:e13195. [PMID: 35652743 PMCID: PMC9287045 DOI: 10.1111/sji.13195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/30/2022]
Abstract
The Karolinska KI/K COVID-19 Immune Atlas project was conceptualized in March 2020 as a part of the academic research response to the developing SARS-CoV-2 pandemic. The aim was to rapidly provide a curated dataset covering the acute immune response towards SARS-CoV-2 infection in humans, as it occurred during the first wave. The Immune Atlas was built as an open resource for broad research and educational purposes. It contains a presentation of the response evoked by different immune and inflammatory cells in defined naïve patient-groups as they presented with moderate and severe COVID-19 disease. The present Resource Article describes how the Karolinska KI/K COVID-19 Immune Atlas allow scientists, students, and other interested parties to freely explore the nature of the immune response towards human SARS-CoV-2 infection in an online setting.
Collapse
Affiliation(s)
- Hans‐Gustaf Ljunggren
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Eivind Heggernes Ask
- Department of Cancer Immunology, Institute for Cancer ResearchOslo University HospitalOsloNorway
- Institute of Clinical MedicineUniversity of OsloOsloNorway
| | - Martin Cornillet
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Benedikt Strunz
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Puran Chen
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Jagadeeswara Rao Muvva
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Mira Akber
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Benedict J. Chambers
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Angelica Cuapio Gomez
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Majda Dzidic
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Iva Filipovic
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Malin Flodström‐Tullberg
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Marina Garcia
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Jean‐Baptiste Gorin
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Sara Gredmark‐Russ
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Laura Hertwig
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Jonas Klingström
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Efthymia Kokkinou
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Egle Kvedaraite
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Magda Lourda
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Jenny Mjösberg
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Christopher Maucourant
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Anna Norrby‐Teglund
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Laura M. Palma Medina
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Tiphaine Parrot
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - André Perez‐Potti
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Andrea Ponzetta
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Emma Ringqvist
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Olga Rivera‐Ballesteros
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Olav Rooyackers
- Department of Emergency MedicineKarolinska University HospitalStockholmSweden
| | - Johan K. Sandberg
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - John Tyler Sandberg
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Takuya Sekine
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Mattias Svensson
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - Renata Varnaite
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | - David Wullimann
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| | | | - Lars I. Eriksson
- Department of Emergency MedicineKarolinska University HospitalStockholmSweden
| | - Soo Aleman
- Department of Infectious DiseasesKarolinska University HospitalStockholmSweden
| | - Karl‐Johan Malmberg
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
- Department of Cancer Immunology, Institute for Cancer ResearchOslo University HospitalOsloNorway
| | - Kristoffer Strålin
- Department of Infectious DiseasesKarolinska University HospitalStockholmSweden
| | - Niklas K. Björkström
- Department of Medicine Huddinge, Center for Infectious MedicineKarolinska InstitutetStockholmSweden
| |
Collapse
|
36
|
Boulouis C, Kammann T, Cuapio A, Parrot T, Gao Y, Mouchtaridi E, Wullimann D, Lange J, Chen P, Akber M, Rivera Ballesteros O, Muvva JR, Smith CIE, Vesterbacka J, Kieri O, Nowak P, Bergman P, Buggert M, Ljunggren HG, Aleman S, Sandberg JK. MAIT cell compartment characteristics are associated with the immune response magnitude to the BNT162b2 mRNA anti-SARS-CoV-2 vaccine. Mol Med 2022; 28:54. [PMID: 35562666 PMCID: PMC9100314 DOI: 10.1186/s10020-022-00484-7] [Citation(s) in RCA: 18] [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: 02/18/2022] [Accepted: 04/25/2022] [Indexed: 12/21/2022] Open
Abstract
Mucosa-associated invariant T (MAIT) cells are unconventional T cells with innate-like capacity to rapidly respond to microbial infection via MR1-restricted antigen recognition. Emerging evidence indicate that they can also act as rapid sensors of viral infection via innate cytokine activation. However, their possible role in the immune response to mRNA vaccination is unknown. Here, we evaluated the involvement of MAIT cells in individuals vaccinated with the BNT162b2 mRNA SARS-CoV-2 vaccine. MAIT cell levels, phenotype and function in circulation were preserved and unperturbed through day 35 post-vaccination in healthy donor (HD) vaccinees, as well as people living with HIV (PLWH) or with primary immunodeficiency (PID). Unexpectedly, pre-vaccination and post-vaccination levels of MAIT cells correlated positively with the magnitude of the SARS-CoV-2 spike protein-specific CD4 T cell and antibody responses in the HD vaccinees. This pattern was largely preserved in the PID group, but less so in the PLWH group. Furthermore, in the HD vaccinees levels of MAIT cell activation and cytolytic potential correlated negatively to the adaptive antigen-specific immune responses. These findings indicate an unexpected association between MAIT cell compartment characteristics and the immune response magnitude to the BNT162b2 mRNA vaccine.
Collapse
Affiliation(s)
- Caroline Boulouis
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Tobias Kammann
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Angelica Cuapio
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Tiphaine Parrot
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Yu Gao
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Elli Mouchtaridi
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - David Wullimann
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Joshua Lange
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Puran Chen
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Mira Akber
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Olga Rivera Ballesteros
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Jagadeeswara Rao Muvva
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - C I Edvard Smith
- Department of Laboratory Medicine, Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
- Department of Laboratory Medicine, Translational Research Center Karolinska (TRACK), Karolinska Institutet, Stockholm, Sweden
| | - Jan Vesterbacka
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Oscar Kieri
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Piotr Nowak
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Peter Bergman
- Department of Laboratory Medicine, Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden
| | - Soo Aleman
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Johan K Sandberg
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 14152, Stockholm, Sweden.
| |
Collapse
|
37
|
Role of triggering receptor expressed on myeloid cells-1 (TREM-1) in COVID-19 and other viral pneumonias: a systematic review and meta-analysis of clinical studies. Inflammopharmacology 2022; 30:1037-1045. [PMID: 35347523 PMCID: PMC8959072 DOI: 10.1007/s10787-022-00972-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/05/2022] [Indexed: 12/29/2022]
Abstract
Background Triggering receptor expressed on myeloid cells-1 (TREM-1) has emerged as an important inflammatory marker of immune response associated with severity and mortality outcomes in infection diseases, including viral pneumonias. Aim (1) To evaluate the expression of TREM-1 in patients with COVID-19 and other viral pneumonias compared to healthy individuals; and (2) to analyze the levels of these biomarkers according to disease severity. Materials and methods This review was conducted according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guideline. Searches were performed in PubMed, Scopus, Embase, and Google Scholar. Studies were considered eligible if they were observational studies that provided data on the levels of TREM-1 in humans with viral pneumonia compared to healthy controls. The results of the meta-analysis were expressed as standardized mean difference (SMD) and an effect size of 0.8 was considered a large effect. A subgroup analysis was performed according to the disease severity. Results Seven studies were included in this systematic review. Four studies included patients with COVID-19 and three analyzed patients with different viruses. The meta-analysis was performed only with patients with COVID-19, which showed increased levels of soluble form of TREM-1 (sTREM-1) among patients with COVID-19 compared to healthy controls (SMD 1.53; 95% CI 0.53–2.52; p < 0.01). No differences were found between patients with mild-to-moderate COVID-19 and healthy controls, but higher levels of sTREM-1 were shown among patients with severe COVID-19 (SMD 1.83; 95% CI 0.77–2.88; p < 0.01). All three studies including patients with other viral pneumonias showed that TREM-1 levels were significantly elevated in infected patients compared with controls. Conclusion These findings may provide evidence on the pro-inflammatory role of TREM-1 in these infections, contributing to the inflammatory profile and disease progression. Supplementary Information The online version contains supplementary material available at 10.1007/s10787-022-00972-6.
Collapse
|
38
|
Jing X, Xu M, Song D, Yue T, Wang Y, Zhang P, Zhong Y, Zhang M, Lam TTY, Faria NR, De Clercq E, Li G. Association between inflammatory cytokines and anti-SARS-CoV-2 antibodies in hospitalized patients with COVID-19. Immun Ageing 2022; 19:12. [PMID: 35248063 PMCID: PMC8897556 DOI: 10.1186/s12979-022-00271-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 02/11/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND COVID-19 patients may experience "cytokine storm" when human immune system produces excessive cytokines/chemokines. However, it remains unclear whether early responses of inflammatory cytokines would lead to high or low titers of anti-SARS-CoV-2 antibodies. METHODS This retrospective study enrolled a cohort of 272 hospitalized patients with laboratory-confirmed SARS-CoV-2. Laboratory assessments of serum cytokines (IL-2R, IL-6, IL-8, IL-10, TNF-α), anti-SARS-CoV-2 IgG/IgM antibodies, and peripheral blood biomarkers were conducted during hospitalization. RESULTS At hospital admission, 36.4% patients were severely ill, 51.5% patients were ≥ 65 years, and 60.3% patients had comorbidities. Higher levels of IL-2R and IL-6 were observed in older patients (≥65 years). Significant differences of IL-2R (week 2 to week ≥5 from symptom onset), IL-6 (week 1 to week ≥5), IL-8 (week 2 to week ≥5), and IL-10 (week 1 to week 3) were observed between moderately-ill and severely ill patients. Anti-SARS-CoV-2 IgG titers were significantly higher in severely ill patients than in moderately ill patients, but such difference was not observed for IgM. High titers of early-stage IL-6, IL-8, and TNF-α (≤2 weeks after symptom onset) were positively correlated with high titers of late-stage IgG (≥5 weeks after symptom onset). Deaths were mostly observed in severely ill older patients (45.9%). Survival analyses revealed risk factors of patient age, baseline COVID-19 severity, and baseline IL-6 that affected survival time, especially in severely ill older patients. CONCLUSION Early responses of elevated cytokines such as IL-6 reflect the active immune responses, leading to high titers of IgG antibodies against COVID-19.
Collapse
Affiliation(s)
- Xixi Jing
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Min Xu
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Deye Song
- Department of Orthopedics, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Tingting Yue
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Yali Wang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Pan Zhang
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, 410078, China
| | - Yanjun Zhong
- Department of Critical Care Medicine, The Second Xiangya Hospital, Central South University, Changsha, 410011, China
| | - Min Zhang
- Institute of Hepatology and Department of Infectious Diseases, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Tommy Tsan-Yuk Lam
- State Key Laboratory of Emerging Infectious Diseases, School of Public Health, The University of Hong Kong, Hong Kong, SAR, China
| | - Nuno Rodrigues Faria
- Department of Infectious Disease Epidemiology, School of Public Health, Imperial College London, London, UK
- Department of Zoology, University of Oxford, Oxford, UK
| | - Erik De Clercq
- Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, 3000, Leuven, Belgium
| | - Guangdi Li
- Hunan Provincial Key Laboratory of Clinical Epidemiology, Xiangya School of Public Health, Central South University, Changsha, 410078, China.
| |
Collapse
|
39
|
Ahern DJ, Ai Z, Ainsworth M, Allan C, Allcock A, Angus B, Ansari MA, Arancibia-Cárcamo CV, Aschenbrenner D, Attar M, Baillie JK, Barnes E, Bashford-Rogers R, Bashyal A, Beer S, Berridge G, Beveridge A, Bibi S, Bicanic T, Blackwell L, Bowness P, Brent A, Brown A, Broxholme J, Buck D, Burnham KL, Byrne H, Camara S, Candido Ferreira I, Charles P, Chen W, Chen YL, Chong A, Clutterbuck EA, Coles M, Conlon CP, Cornall R, Cribbs AP, Curion F, Davenport EE, Davidson N, Davis S, Dendrou CA, Dequaire J, Dib L, Docker J, Dold C, Dong T, Downes D, Drakesmith H, Dunachie SJ, Duncan DA, Eijsbouts C, Esnouf R, Espinosa A, Etherington R, Fairfax B, Fairhead R, Fang H, Fassih S, Felle S, Fernandez Mendoza M, Ferreira R, Fischer R, Foord T, Forrow A, Frater J, Fries A, Gallardo Sanchez V, Garner LC, Geeves C, Georgiou D, Godfrey L, Golubchik T, Gomez Vazquez M, Green A, Harper H, Harrington HA, Heilig R, Hester S, Hill J, Hinds C, Hird C, Ho LP, Hoekzema R, Hollis B, Hughes J, Hutton P, Jackson-Wood MA, Jainarayanan A, James-Bott A, Jansen K, Jeffery K, Jones E, Jostins L, Kerr G, Kim D, Klenerman P, Knight JC, Kumar V, et alAhern DJ, Ai Z, Ainsworth M, Allan C, Allcock A, Angus B, Ansari MA, Arancibia-Cárcamo CV, Aschenbrenner D, Attar M, Baillie JK, Barnes E, Bashford-Rogers R, Bashyal A, Beer S, Berridge G, Beveridge A, Bibi S, Bicanic T, Blackwell L, Bowness P, Brent A, Brown A, Broxholme J, Buck D, Burnham KL, Byrne H, Camara S, Candido Ferreira I, Charles P, Chen W, Chen YL, Chong A, Clutterbuck EA, Coles M, Conlon CP, Cornall R, Cribbs AP, Curion F, Davenport EE, Davidson N, Davis S, Dendrou CA, Dequaire J, Dib L, Docker J, Dold C, Dong T, Downes D, Drakesmith H, Dunachie SJ, Duncan DA, Eijsbouts C, Esnouf R, Espinosa A, Etherington R, Fairfax B, Fairhead R, Fang H, Fassih S, Felle S, Fernandez Mendoza M, Ferreira R, Fischer R, Foord T, Forrow A, Frater J, Fries A, Gallardo Sanchez V, Garner LC, Geeves C, Georgiou D, Godfrey L, Golubchik T, Gomez Vazquez M, Green A, Harper H, Harrington HA, Heilig R, Hester S, Hill J, Hinds C, Hird C, Ho LP, Hoekzema R, Hollis B, Hughes J, Hutton P, Jackson-Wood MA, Jainarayanan A, James-Bott A, Jansen K, Jeffery K, Jones E, Jostins L, Kerr G, Kim D, Klenerman P, Knight JC, Kumar V, Kumar Sharma P, Kurupati P, Kwok A, Lee A, Linder A, Lockett T, Lonie L, Lopopolo M, Lukoseviciute M, Luo J, Marinou S, Marsden B, Martinez J, Matthews PC, Mazurczyk M, McGowan S, McKechnie S, Mead A, Mentzer AJ, Mi Y, Monaco C, Montadon R, Napolitani G, Nassiri I, Novak A, O'Brien DP, O'Connor D, O'Donnell D, Ogg G, Overend L, Park I, Pavord I, Peng Y, Penkava F, Pereira Pinho M, Perez E, Pollard AJ, Powrie F, Psaila B, Quan TP, Repapi E, Revale S, Silva-Reyes L, Richard JB, Rich-Griffin C, Ritter T, Rollier CS, Rowland M, Ruehle F, Salio M, Sansom SN, Sanches Peres R, Santos Delgado A, Sauka-Spengler T, Schwessinger R, Scozzafava G, Screaton G, Seigal A, Semple MG, Sergeant M, Simoglou Karali C, Sims D, Skelly D, Slawinski H, Sobrinodiaz A, Sousos N, Stafford L, Stockdale L, Strickland M, Sumray O, Sun B, Taylor C, Taylor S, Taylor A, Thongjuea S, Thraves H, Todd JA, Tomic A, Tong O, Trebes A, Trzupek D, Tucci FA, Turtle L, Udalova I, Uhlig H, van Grinsven E, Vendrell I, Verheul M, Voda A, Wang G, Wang L, Wang D, Watkinson P, Watson R, Weinberger M, Whalley J, Witty L, Wray K, Xue L, Yeung HY, Yin Z, Young RK, Youngs J, Zhang P, Zurke YX. A blood atlas of COVID-19 defines hallmarks of disease severity and specificity. Cell 2022; 185:916-938.e58. [PMID: 35216673 PMCID: PMC8776501 DOI: 10.1016/j.cell.2022.01.012] [Show More Authors] [Citation(s) in RCA: 179] [Impact Index Per Article: 59.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 08/16/2021] [Accepted: 01/17/2022] [Indexed: 02/06/2023]
Abstract
Treatment of severe COVID-19 is currently limited by clinical heterogeneity and incomplete description of specific immune biomarkers. We present here a comprehensive multi-omic blood atlas for patients with varying COVID-19 severity in an integrated comparison with influenza and sepsis patients versus healthy volunteers. We identify immune signatures and correlates of host response. Hallmarks of disease severity involved cells, their inflammatory mediators and networks, including progenitor cells and specific myeloid and lymphocyte subsets, features of the immune repertoire, acute phase response, metabolism, and coagulation. Persisting immune activation involving AP-1/p38MAPK was a specific feature of COVID-19. The plasma proteome enabled sub-phenotyping into patient clusters, predictive of severity and outcome. Systems-based integrative analyses including tensor and matrix decomposition of all modalities revealed feature groupings linked with severity and specificity compared to influenza and sepsis. Our approach and blood atlas will support future drug development, clinical trial design, and personalized medicine approaches for COVID-19.
Collapse
|
40
|
McElvaney OF, Asakura T, Meinig SL, Torres-Castillo JL, Hagan RS, Gabillard-Lefort C, Murphy MP, Thorne LB, Borczuk A, Reeves EP, Zumwalt RE, Mikami Y, Carroll TP, Okuda K, Hogan G, McElvaney OJ, Clarke J, McEvoy NL, Mallon PW, McCarthy C, Curley G, Wolfgang MC, Boucher RC, McElvaney NG. Protease-anti-protease compartmentalization in SARS-CoV-2 ARDS: Therapeutic implications. EBioMedicine 2022; 77:103894. [PMID: 35217407 PMCID: PMC8861575 DOI: 10.1016/j.ebiom.2022.103894] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 02/08/2022] [Accepted: 02/08/2022] [Indexed: 12/14/2022] Open
Abstract
Background Interleukin-6 (IL-6) is elevated in SARS-CoV-2 infection. IL-6 regulates acute-phase proteins, such as alpha-1 antitrypsin (AAT), a key lung anti-protease. We investigated the protease-anti-protease balance in the circulation and pulmonary compartments in SARS-CoV-2 acute respiratory distress syndrome (ARDS) compared to non-SARS-CoV-2 ARDS (nsARDS) and the effects of tocilizumab (IL-6 receptor antagonist) on anti-protease defence in SARS-CoV-2 infection. Methods Levels and activity of AAT and neutrophil elastase (NE) were measured in plasma, airway tissue and tracheal secretions (TA) of people with SARS-CoV-2 ARDS or nsARDS. AAT and IL-6 levels were evaluated in people with moderate SARS-CoV-2 infection who received standard of care +/- tocilizumab. Findings AAT plasma levels doubled in SARS-CoV-2 ARDS. In lung parenchyma AAT levels were increased, as was the percentage of neutrophils involved in NET formation. A protease-anti-protease imbalance was detected in TA with active NE and no active AAT. The airway anti-protease, secretory leukoprotease inhibitor was decreased in SARS-CoV-2-infected lungs and cleaved in TA. In nsARDS, plasma AAT levels were elevated but TA samples had less AAT cleavage, with no detectable active NE in most samples Induction of AAT in ARDS occurred mainly through IL-6. Tocilizumab down-regulated AAT during SARS-CoV-2 infection. Interpretation There is a protease-anti-protease imbalance in the airways of SARS-CoV-2-ARDS patients. This imbalance is a target for anti-protease therapy.
Collapse
Affiliation(s)
- Oisin F McElvaney
- Irish Centre for Genetic Lung Disease, RCSI Education and Research Centre, Beaumont Hospital, Dublin 9, Dublin, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Takanori Asakura
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Suzanne L Meinig
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jose L Torres-Castillo
- Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Robert S Hagan
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Division of Pulmonary Diseases and Critical Care Medicine, Department of Medicine, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Claudie Gabillard-Lefort
- Irish Centre for Genetic Lung Disease, RCSI Education and Research Centre, Beaumont Hospital, Dublin 9, Dublin, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Mark P Murphy
- Irish Centre for Genetic Lung Disease, RCSI Education and Research Centre, Beaumont Hospital, Dublin 9, Dublin, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland.
| | - Leigh B Thorne
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Alain Borczuk
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Emer P Reeves
- Irish Centre for Genetic Lung Disease, RCSI Education and Research Centre, Beaumont Hospital, Dublin 9, Dublin, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ross E Zumwalt
- Department of Pathology, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Yu Mikami
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Tomas P Carroll
- Irish Centre for Genetic Lung Disease, RCSI Education and Research Centre, Beaumont Hospital, Dublin 9, Dublin, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland; Alpha-1 Foundation, Ireland
| | - Kenichi Okuda
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Grace Hogan
- Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Oliver J McElvaney
- Irish Centre for Genetic Lung Disease, RCSI Education and Research Centre, Beaumont Hospital, Dublin 9, Dublin, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Jennifer Clarke
- Department of Anaesthesia and Critical Care, Beaumont Hospital, Dublin, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Natalie L McEvoy
- Department of Anaesthesia and Critical Care, Beaumont Hospital, Dublin, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Patrick W Mallon
- Department of Infectious Diseases, St Vincent's University Hospital, Dublin, Ireland; Centre for Experimental Pathogen Host Research (CEPHR), University College Dublin, Dublin, Ireland
| | - Cormac McCarthy
- Department of Respiratory Medicine, St Vincent's University Hospital, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland
| | - Ger Curley
- Department of Anaesthesia and Critical Care, Beaumont Hospital, Dublin, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Matthew C Wolfgang
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Microbiology and Immunology, University of North Carolina School of Medicine, Chapel Hill, NC 27599, USA
| | - Richard C Boucher
- Marsico Lung Institute/Cystic Fibrosis Research Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Noel G McElvaney
- Irish Centre for Genetic Lung Disease, RCSI Education and Research Centre, Beaumont Hospital, Dublin 9, Dublin, Ireland; Royal College of Surgeons in Ireland, Dublin, Ireland
| |
Collapse
|
41
|
Georg P, Astaburuaga-García R, Bonaguro L, Brumhard S, Michalick L, Lippert LJ, Kostevc T, Gäbel C, Schneider M, Streitz M, Demichev V, Gemünd I, Barone M, Tober-Lau P, Helbig ET, Hillus D, Petrov L, Stein J, Dey HP, Paclik D, Iwert C, Mülleder M, Aulakh SK, Djudjaj S, Bülow RD, Mei HE, Schulz AR, Thiel A, Hippenstiel S, Saliba AE, Eils R, Lehmann I, Mall MA, Stricker S, Röhmel J, Corman VM, Beule D, Wyler E, Landthaler M, Obermayer B, von Stillfried S, Boor P, Demir M, Wesselmann H, Suttorp N, Uhrig A, Müller-Redetzky H, Nattermann J, Kuebler WM, Meisel C, Ralser M, Schultze JL, Aschenbrenner AC, Thibeault C, Kurth F, Sander LE, Blüthgen N, Sawitzki B. Complement activation induces excessive T cell cytotoxicity in severe COVID-19. Cell 2022; 185:493-512.e25. [PMID: 35032429 PMCID: PMC8712270 DOI: 10.1016/j.cell.2021.12.040] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 10/22/2021] [Accepted: 12/22/2021] [Indexed: 12/20/2022]
Abstract
Severe COVID-19 is linked to both dysfunctional immune response and unrestrained immunopathology, and it remains unclear whether T cells contribute to disease pathology. Here, we combined single-cell transcriptomics and single-cell proteomics with mechanistic studies to assess pathogenic T cell functions and inducing signals. We identified highly activated CD16+ T cells with increased cytotoxic functions in severe COVID-19. CD16 expression enabled immune-complex-mediated, T cell receptor-independent degranulation and cytotoxicity not found in other diseases. CD16+ T cells from COVID-19 patients promoted microvascular endothelial cell injury and release of neutrophil and monocyte chemoattractants. CD16+ T cell clones persisted beyond acute disease maintaining their cytotoxic phenotype. Increased generation of C3a in severe COVID-19 induced activated CD16+ cytotoxic T cells. Proportions of activated CD16+ T cells and plasma levels of complement proteins upstream of C3a were associated with fatal outcome of COVID-19, supporting a pathological role of exacerbated cytotoxicity and complement activation in COVID-19.
Collapse
Affiliation(s)
- Philipp Georg
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rosario Astaburuaga-García
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany; IRI Life Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lorenzo Bonaguro
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; Systems Medicine, Deutsches Zentrum für Neurodegenerativen Erkrankungen (DZNE), Bonn, Germany
| | - Sophia Brumhard
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Laura Michalick
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lena J Lippert
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tomislav Kostevc
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christiane Gäbel
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Maria Schneider
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Mathias Streitz
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Vadim Demichev
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK; Department of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany; Department of Biochemistry, Cambridge Centre for Proteomics, University of Cambridge, Cambridge, UK
| | - Ioanna Gemünd
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; PRECISE Platform for Genomics and Epigenomics at DZNE, University of Bonn, Bonn, Germany; Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Matthias Barone
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Pinkus Tober-Lau
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Elisa T Helbig
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - David Hillus
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Lev Petrov
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Julia Stein
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Hannah-Philine Dey
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Daniela Paclik
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christina Iwert
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Michael Mülleder
- Core Facility, High Throughput Mass Spectrometry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Simran Kaur Aulakh
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Sonja Djudjaj
- Institute of Pathology, University Clinic Aachen, RWTH Aachen, Aachen, Germany
| | - Roman D Bülow
- Institute of Pathology, University Clinic Aachen, RWTH Aachen, Aachen, Germany
| | - Henrik E Mei
- Mass Cytometry Laboratory, DRFZ Berlin, A Leibniz Institute, Berlin, Germany
| | - Axel R Schulz
- Mass Cytometry Laboratory, DRFZ Berlin, A Leibniz Institute, Berlin, Germany
| | - Andreas Thiel
- Si-M/"Der Simulierte Mensch" a Science Framework of Technische Universität Berlin and Charité - Universitätsmedizin Berlin, Berlin, Germany; Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt - Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Stefan Hippenstiel
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Antoine-Emmanuel Saliba
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), Würzburg, Germany
| | - Roland Eils
- Center for Digital Health, Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL), Berlin, Germany
| | - Irina Lehmann
- Center for Digital Health, Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL), Berlin, Germany
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany; German Center for Lung Research (DZL), Associated Partner, Berlin, Germany; Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sebastian Stricker
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Jobst Röhmel
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Victor M Corman
- Institute of Virology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Emanuel Wyler
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Markus Landthaler
- IRI Life Sciences, Humboldt-Universität zu Berlin, Berlin, Germany; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin Institute for Medical Systems Biology, Berlin, Germany
| | - Benedikt Obermayer
- Core Unit Bioinformatics, Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Peter Boor
- Institute of Pathology, University Clinic Aachen, RWTH Aachen, Aachen, Germany; Department of Nephrology, University Clinic Aachen, RWTH Aachen, Aachen, Germany; Electron Microscopy Facility, University Clinic Aachen, RWTH Aachen, Aachen, Germany
| | - Münevver Demir
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Hans Wesselmann
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Norbert Suttorp
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; German Center for Lung Research (DZL), Gießen, Germany
| | - Alexander Uhrig
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Holger Müller-Redetzky
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Jacob Nattermann
- Department of Internal Medicine I, University Hospital Bonn, Bonn, Germany
| | - Wolfgang M Kuebler
- Institute of Physiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christian Meisel
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Department of Immunology, Labor Berlin, Charité Vivantes, Berlin, Germany
| | - Markus Ralser
- Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK; Department of Biochemistry, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Joachim L Schultze
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; Systems Medicine, Deutsches Zentrum für Neurodegenerativen Erkrankungen (DZNE), Bonn, Germany; PRECISE Platform for Genomics and Epigenomics at DZNE, University of Bonn, Bonn, Germany
| | - Anna C Aschenbrenner
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany; Systems Medicine, Deutsches Zentrum für Neurodegenerativen Erkrankungen (DZNE), Bonn, Germany; PRECISE Platform for Genomics and Epigenomics at DZNE, University of Bonn, Bonn, Germany; Department of Internal Medicine, Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Charlotte Thibeault
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Florian Kurth
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany; Department of Tropical Medicine, Bernhard Nocht Institute for Tropical Medicine, Department of Medicine I, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Leif E Sander
- Department of Infectious Diseases and Respiratory Medicine, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nils Blüthgen
- Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany; IRI Life Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Birgit Sawitzki
- Institute of Medical Immunology, Charité - Universitätsmedizin Berlin, Berlin, Germany; Berlin Institute of Health (BIH), Charité - Universitätsmedizin Berlin, Berlin, Germany.
| |
Collapse
|
42
|
Frost JN, Hamilton F, Arnold D, Elvers KT, Shah A, Armitage AE, Milne A, McKernon J, Attwood M, Chen YL, Xue L, Youngs J, Provine NM, Bicanic T, Klenerman P, Drakesmith H, Ghazal P. Evaluation of perturbed iron-homeostasis in a prospective cohort of patients with COVID-19. Wellcome Open Res 2022; 7:173. [PMID: 35935705 PMCID: PMC9307999 DOI: 10.12688/wellcomeopenres.17904.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/13/2022] [Indexed: 11/20/2022] Open
Abstract
Background: Marked reductions in serum iron concentrations are commonly induced during the acute phase of infection. This phenomenon, termed hypoferremia of inflammation, leads to inflammatory anemia, but could also have broader pathophysiological implications. In patients with coronavirus disease 2019 (COVID-19), hypoferremia is associated with disease severity and poorer outcomes, although there are few reported cohorts. Methods: In this study, we leverage a well characterised prospective cohort of hospitalised COVID-19 patients and perform a set of analyses focussing on iron and related biomarkers and both acute severity of COVID-19 and longer-term symptomatology. Results: We observed no associations between acute serum iron and long-term outcomes (including fatigue, breathlessness or quality of life); however, lower haemoglobin was associated with poorer quality of life. We also quantified iron homeostasis associated parameters, demonstrating that among 50 circulating mediators of inflammation IL-6 concentrations were strongly associated with serum iron, consistent with its central role in inflammatory control of iron homeostasis. Surprisingly, we observed no association between serum hepcidin and serum iron concentrations. We also observed elevated erythroferrone concentrations in COVID-19 patients with anaemia of inflammation. Conclusions: These results enhance our understanding of the regulation and pathophysiological consequences of disturbed iron homeostasis during SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Joe N. Frost
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Fergus Hamilton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, BS10 5NB, UK
- North Bristol NHS Trust, Bristol, BS10 5NB, UK
| | | | - Karen T. Elvers
- Medicines Discovery Institute, Cardiff University, Cardiff, UK
| | - Akshay Shah
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Andrew E. Armitage
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Alice Milne
- North Bristol NHS Trust, Bristol, BS10 5NB, UK
| | | | | | - Yi-Ling Chen
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Luzheng Xue
- Respiratory Medicine Unit and Oxford NIHR Biomedical Research Centre, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Jonathan Youngs
- Institute for Infection and Immunity, St George's, University of London, London, UK
- Clinical Academic Group in Infection and Immunity, St George's Hospital, London, London, UK
| | - Nicholas M. Provine
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Tihana Bicanic
- Institute for Infection and Immunity, St George's, University of London, London, UK
- Clinical Academic Group in Infection and Immunity, St George's Hospital, London, London, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, UK
- Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Hal Drakesmith
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX1 2JD, UK
| | - Peter Ghazal
- Medicines Discovery Institute, Cardiff University, Cardiff, UK
- Project Sepsis, Systems Immunity Research Institute, Division of Infection and Immunity, Cardiff University, Cardiff, UK
| |
Collapse
|
43
|
Abstract
Mucosal associated invariant T (MAIT) cells were first identified as specific for bacterial, mycobacterial, and fungal organisms, which detect microbially-derived biosynthetic ligands presented by MHC-related protein 1 (MR1). More recently two unexpected, additional roles have been identified for these ancient and abundant cells: a TCR-depen-dent role in tissue repair and a TCR-independent role in antiviral host defence. Data from several classes of viral disease shows their capability for activation by the cytokines interleukin (IL)-12, IL-15, IL-18, and type I interferon. MAIT cells are abundant at mucosal surfaces, particularly in the lung, and it seems likely a primary reason for their striking evolutionary conservation is an important role in early innate defence against respiratory infections, including both bacteria and viruses. Here we review evidence for their TCR-independent activation, observational human data for their activation in influenza A virus, and in vivo murine evidence of their protection against severe influenza A infection, mediated at least partially via IFN-gamma. We then survey evidence emerging from other respiratory viral infections including recent evidence for an important adjuvant role in adenovirus infection, specifically chimpanzee adenoviruses used in recent coronavirus vaccines, and data for strong associations between MAIT cell responses and adverse outcomes from coronavirus-19 (COVID-19) disease. We speculate on potential translational implications of these findings, either using corticosteroids or inhibitory ligands to suppress deleterious MAIT cell responses, or the potential utility of stimulatory MR1 ligands to boost MAIT cell frequencies to enhance innate viral defences.
Collapse
Affiliation(s)
- Yuqing Long
- Respiratory Medicine Unit and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Nuffield Department of Medicine Experimental Medicine, University of Oxford, OX3 9DU, Oxfordshire, UK
- Chinese Academy of Medical Sciences Oxford Institute (COI), University of Oxford, Oxford, UK
| | - Timothy SC Hinks
- Respiratory Medicine Unit and National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC), Nuffield Department of Medicine Experimental Medicine, University of Oxford, OX3 9DU, Oxfordshire, UK
| |
Collapse
|