1
|
Stern L, Emanuel Z, Traves R, Willis K, Purohit SK, Samer C, Mak JYW, Fairlie DP, Tscharke DC, Corbett AJ, Abendroth A, Slobedman B. Herpes simplex virus type 1 impairs mucosal-associated invariant T cells. mBio 2025; 16:e0388724. [PMID: 40135871 PMCID: PMC12077205 DOI: 10.1128/mbio.03887-24] [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: 01/13/2025] [Accepted: 02/04/2025] [Indexed: 03/27/2025] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a highly successful pathogen that infects mucosal sites and adopts an arsenal of strategies to manipulate host immunity. Mucosal-associated invariant T (MAIT) cells are abundant innate-like T lymphocytes that recognize bacterial and fungal-derived vitamin B-related metabolites presented by major histocompatibility complex class I-related protein 1 (MR1). MAIT cells can also be activated in an MR1-independent manner via cytokine stimulation, predominantly by IL-12 and IL-18. MAIT cell alterations have been identified as being associated with a number of viral infections, but direct interactions between viruses and MAIT cells are poorly understood. It is unknown whether HSV-1 can infect MAIT cells and modulate their functions. Here, we show that HSV-1 can infect primary human MAIT cells, including CD4±/CD8± and CD56± MAIT cell subpopulations. Furthermore, HSV-1 infection profoundly inhibits the functional capacity of MAIT cells to respond to T cell receptor (TCR)-dependent stimulation by the MAIT cell activating ligand 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil (5-OP-RU) and to cytokine stimulation by IL-12/IL-18. HSV-1-infected MAIT cells display reduced cytotoxic potential, diminished synthesis of effector cytokines, and decreased expression of key cytokine receptors including IL-18R. In addition, MAIT cells exposed to HSV-1-infected fibroblasts but which remained uninfected (viral GFP-negative) also exhibit a suppressed effector response to TCR-dependent stimulation. The functional suppression of HSV-1-exposed MAIT cells was not mediated by a soluble factor within the supernatant, suggesting direct contact of MAIT cells with HSV-1-infected fibroblasts is required. Overall, this study reveals that HSV-1 can infect MAIT cells and substantially impair MAIT cell effector functions. IMPORTANCE Mucosal-associated invariant T cells (MAIT cells) are "unconventional" immune cells that are becoming increasingly appreciated to play important roles in a variety of viral infections. Herpes simplex virus (HSV) causes significant human disease and is a master manipulator of multiple immune functions, but how this virus may control MAIT cells is poorly understood. We discovered that HSV can infect human MAIT cells and impair their functional capacity and also show that MAIT cells exposed to HSV, but which do not show evidence of infection, are similarly impaired. This study therefore identifies an additional immunomodulatory function of HSV.
Collapse
Affiliation(s)
- Lauren Stern
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Zoe Emanuel
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Renee Traves
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Katherine Willis
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Shivam K. Purohit
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Carolyn Samer
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Jeffrey Y. W. Mak
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - David P. Fairlie
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - David C. Tscharke
- John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Alexandra J. Corbett
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Allison Abendroth
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Barry Slobedman
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| |
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
|
Yang Z, Luo B, Li M, He Z, Ren C, Chen X, Kang X, Chen H, Xu E, Guan W, Xia X. The effector function of mucosal associated invariant T cells alters with aging and is regulated by RORγt. Front Immunol 2024; 15:1504806. [PMID: 39669566 PMCID: PMC11634854 DOI: 10.3389/fimmu.2024.1504806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/01/2024] [Indexed: 12/14/2024] Open
Abstract
Introduction Mucosal-associated invariant T (MAIT) cells are a predominant subset of innate-like T cells in humans, characterized by diverse gene expression profiles and functional capabilities. However, the factors influencing the transcriptomes and effector functions of MAIT cells, particularly at mucosal barriers, remain largely unclear. Methods In this study, we employed single-cell RNA sequencing (scRNA-seq) and functional assays to investigate the transcriptomic and functional characteristics of intestinal MAIT cells in mouse models during aging. We also extended scRNA-seq analysis to human intestinal MAIT cells to compare their gene expression patterns with those observed in aged mice. Results Our findings demonstrated that the transcriptomes and functional capabilities of intestinal MAIT cells shifted from MAIT17 to MAIT1 profiles with aging in mouse models, with notable changes in the production of cytotoxic molecules. Further scRNA-seq analysis of human intestinal MAIT cells revealed a segregation into MAIT1 and MAIT17 subsets, displaying gene expression patterns that mirrored those seen in aged mouse models. The transcription factor RORγt was expressed in both MAIT1 and MAIT17 cells, acting to repress IFNγ production while promoting IL17 expression. Moreover, reduced expression of RORC and Il17A was correlated with poorer survival outcomes in colorectal cancer patients. Discussion These results suggest that aging induces a functional shift between MAIT1 and MAIT17 cells, which may be influenced by transcriptional regulators like RORγt. The observed alterations in MAIT cell activity could potentially impact disease prognosis, particularly in colorectal cancer. This study provides new insights into the dynamics of MAIT cell responses at mucosal barriers, highlighting possible therapeutic targets for modulating MAIT cell functions in aging and disease.
Collapse
Affiliation(s)
- Zhi Yang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Banxin Luo
- Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Minhuan Li
- Department of Andrology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Ziyun He
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chuanfu Ren
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Xin Chen
- Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xing Kang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hong Chen
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - En Xu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wenxian Guan
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
- Department of General Surgery, Taikang Xianlin DrumTower Hospital, The Affiliated Hospital of Wuhan University Medical School, Nanjing, China
| | - Xuefeng Xia
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
- Department of General Surgery, Taikang Xianlin DrumTower Hospital, The Affiliated Hospital of Wuhan University Medical School, Nanjing, China
| |
Collapse
|
6
|
Loh L, Saunders PM, Faoro C, Font-Porterias N, Nemat-Gorgani N, Harrison GF, Sadeeq S, Hensen L, Wong SC, Widjaja J, Clemens EB, Zhu S, Kichula KM, Tao S, Zhu F, Montero-Martin G, Fernandez-Vina M, Guethlein LA, Vivian JP, Davies J, Mentzer AJ, Oppenheimer SJ, Pomat W, Ioannidis AG, Barberena-Jonas C, Moreno-Estrada A, Miller A, Parham P, Rossjohn J, Tong SYC, Kedzierska K, Brooks AG, Norman PJ. An archaic HLA class I receptor allele diversifies natural killer cell-driven immunity in First Nations peoples of Oceania. Cell 2024; 187:7008-7024.e19. [PMID: 39476840 PMCID: PMC11606752 DOI: 10.1016/j.cell.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 05/24/2024] [Accepted: 10/03/2024] [Indexed: 11/02/2024]
Abstract
Genetic variation in host immunity impacts the disproportionate burden of infectious diseases that can be experienced by First Nations peoples. Polymorphic human leukocyte antigen (HLA) class I and killer cell immunoglobulin-like receptors (KIRs) are key regulators of natural killer (NK) cells, which mediate early infection control. How this variation impacts their responses across populations is unclear. We show that HLA-A∗24:02 became the dominant ligand for inhibitory KIR3DL1 in First Nations peoples across Oceania, through positive natural selection. We identify KIR3DL1∗114, widespread across and unique to Oceania, as an allele lineage derived from archaic humans. KIR3DL1∗114+NK cells from First Nations Australian donors are inhibited through binding HLA-A∗24:02. The KIR3DL1∗114 lineage is defined by phenylalanine at residue 166. Structural and binding studies show phenylalanine 166 forms multiple unique contacts with HLA-peptide complexes, increasing both affinity and specificity. Accordingly, assessing immunogenetic variation and the functional implications for immunity are fundamental toward understanding population-based disease associations.
Collapse
Affiliation(s)
- Liyen Loh
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia; Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Philippa M Saunders
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Camilla Faoro
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Neus Font-Porterias
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Neda Nemat-Gorgani
- Department of Structural Biology and Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Genelle F Harrison
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Suraju Sadeeq
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Luca Hensen
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Shu Cheng Wong
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jacqueline Widjaja
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - E Bridie Clemens
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Shiying Zhu
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Katherine M Kichula
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Sudan Tao
- Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA; Blood Center of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Faming Zhu
- Blood Center of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Gonzalo Montero-Martin
- Stanford Blood Centre, Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Marcelo Fernandez-Vina
- Stanford Blood Centre, Department of Pathology, Stanford University, Stanford, CA 94305, USA
| | - Lisbeth A Guethlein
- Department of Structural Biology and Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Julian P Vivian
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia
| | - Jane Davies
- Menzies School of Health Research, Charles Darwin University, Darwin, NT 0810, Australia; Department of Infectious Diseases, Royal Darwin Hospital, Casuarina, NT 0810, Australia
| | - Alexander J Mentzer
- Wellcome Centre for Human Genetics, University of Oxford, Oxford OX3 7BN, UK; Big Data Institute, Li Ka Shing Centre for Health Information and Discovery, University of Oxford, Oxford OX3 7LF, UK
| | - Stephen J Oppenheimer
- Institute of Social and Cultural Anthropology, School of Anthropology and Museum Ethnography, University of Oxford, Oxford OX3 7LF, UK
| | - William Pomat
- Papua New Guinea Institute of Medical Research, Post Office Box 60, Goroka, Papua New Guinea
| | | | - Carmina Barberena-Jonas
- Advanced Genomics Unit, Center for Research and Advanced Studies (CINVESTAV), Irapuato 36821, Mexico
| | - Andrés Moreno-Estrada
- Advanced Genomics Unit, Center for Research and Advanced Studies (CINVESTAV), Irapuato 36821, Mexico
| | - Adrian Miller
- Jawun Research Centre, Central Queensland University, Cairns, QLD 4870, Australia
| | - Peter Parham
- Department of Structural Biology and Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC 3800, Australia; Institute of Infection and Immunity, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, UK.
| | - Steven Y C Tong
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3000, Australia; Department of Infectious Diseases, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, VIC 3000, Australia.
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Andrew G Brooks
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia.
| | - Paul J Norman
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Biomedical Informatics, University of Colorado School of Medicine, Aurora, CO 80045, USA; Department of Structural Biology and Department of Microbiology and Immunology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
7
|
Hsieh LL, Looney M, Figueroa A, Massaccesi G, Stavrakis G, Anaya EU, D'Alessio FR, Ordonez AA, Pekosz AS, DeFilippis VR, Karakousis PC, Karaba AH, Cox AL. Bystander monocytic cells drive infection-independent NLRP3 inflammasome response to SARS-CoV-2. mBio 2024; 15:e0081024. [PMID: 39240187 PMCID: PMC11481483 DOI: 10.1128/mbio.00810-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/26/2024] [Indexed: 09/07/2024] Open
Abstract
The pathogenesis of COVID-19 is associated with a hyperinflammatory immune response. Monocytes and macrophages play a central role in this hyperinflammatory response to SARS-CoV-2. NLRP3 inflammasome activation has been observed in monocytes of patients with COVID-19, but the mechanism and consequences of inflammasome activation require further investigation. In this study, we inoculated a macrophage-like THP-1 cell line, primary differentiated human nasal epithelial cell (hNEC) cultures, and primary monocytes with SARS-CoV-2. We found that the activation of the NLRP3 inflammasome in macrophages does not rely on viral replication, receptor-mediated entry, or actin-dependent entry. SARS-CoV-2 productively infected hNEC cultures without triggering the production of inflammasome cytokines IL-18 and IL-1β. Importantly, these cytokines did not inhibit viral replication in hNEC cultures. SARS-CoV-2 inoculation of primary monocytes led to inflammasome activation and induced a macrophage phenotype in these cells. Monocytic cells from bronchoalveolar lavage (BAL) fluid, but not from peripheral blood, of patients with COVID-19, showed evidence of inflammasome activation, expressed the proinflammatory marker CD11b, and displayed oxidative burst. These findings highlight the central role of activated macrophages, as a result of direct viral sensing, in COVID-19 and support the inhibition of IL-1β and IL-18 as potential therapeutic strategies to reduce immunopathology without increasing viral replication. IMPORTANCE Inflammasome activation is associated with severe COVID-19. The impact of inflammasome activation on viral replication and mechanistic details of this activation are not clarified. This study advances our understanding of the role of inflammasome activation in macrophages by identifying TLR2, NLRP3, ASC, and caspase-1 as dependent factors in this activation. Further, it highlights that SARS-CoV-2 inflammasome activation is not a feature of nasal epithelial cells but rather activation of bystander macrophages in the airway. Finally, we demonstrate that two pro inflammatory cytokines produced by inflammasome activation, IL-18 and IL-1β, do not restrict viral replication and are potential targets to ameliorate pathological inflammation in severe COVID-19.
Collapse
Affiliation(s)
- Leon L. Hsieh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Monika Looney
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alexis Figueroa
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Guido Massaccesi
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Georgia Stavrakis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Eduardo U. Anaya
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Franco R. D'Alessio
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Alvaro A. Ordonez
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrew S. Pekosz
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Victor R. DeFilippis
- Vaccine and Gene Therapy Institute, Oregon Health and Science University, Portland, Oregon, USA
| | - Petros C. Karakousis
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
| | - Andrew H. Karaba
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Andrea L. Cox
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland, USA
| |
Collapse
|
8
|
López-Rodríguez JC, Barral P. Mucosal associated invariant T cells: Powerhouses of the lung. Immunol Lett 2024; 269:106910. [PMID: 39128630 PMCID: PMC11835791 DOI: 10.1016/j.imlet.2024.106910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The lungs face constant environmental challenges from harmless molecules, airborne pathogens and harmful agents that can damage the tissue. The lungs' immune system includes numerous tissue-resident lymphocytes that contribute to maintain tissue homeostasis and to the early initiation of immune responses. Amongst tissue-resident lymphocytes, Mucosal Associated Invariant T (MAIT) cells are present in human and murine lungs and emerging evidence supports their contribution to immune responses during infections, chronic inflammatory disorders and cancer. This review explores the mechanisms underpinning MAIT cell functions in the airways, their impact on lung immunity and the potential for targeting pulmonary MAIT cells in a therapeutic context.
Collapse
Affiliation(s)
- J C López-Rodríguez
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK.
| | - P Barral
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK.
| |
Collapse
|
9
|
Saka VP, G V NK, Sanapalli BKR, Goswami A, Roy A, Agrawal A, Gupta P, Verma D, Kaushik S. Unveiling the Immunostimulatory Potential of Rhus Toxicodendron in Immunocompromised Balb/C Mice Induced with Cyclophosphamide. Diseases 2024; 12:178. [PMID: 39195177 DOI: 10.3390/diseases12080178] [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: 06/20/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
This study investigated how Rhus toxicodendron (RT) (6C, 30C, and 200C) can boost the immune system of BALB/c mice that were given cyclophosphamide (CPM), which is an anticancer drug that weakens the immune system. RT, known for its historical use in traditional homeopathic remedies, has demonstrated immunomodulatory and anti-inflammatory effects in various experimental models. To test the immune-boosting effects of RT, CPM (80 mg/kg) was given intraperitoneally to mice on days 4, 8, and 12 of the study but not to the normal control group. CPM-induced immunosuppression led to significant decreases in red blood cell (RBC), white blood cell (WBC), and hemoglobin (Hb) levels, and reduced spleen and thymus indices. Phagocytic activity, cytokine concentrations, and spleen architecture were also adversely affected. RT treatment, particularly at 200C, significantly ameliorated these effects, improving RBC, WBC, and Hb levels. Furthermore, RT partially prevented CPM-induced atrophy of immune organs. Treatment positively influenced cytokine production at both the protein and mRNA levels, restoring immune balance. Histopathological results confirmed that RT stimulated the immune system. The cells were more stable, and the white pulp in the spleen was arranged in a regular pattern. These findings suggest that RT may serve as an adjunctive immunostimulant therapy for conditions characterized by immunosuppression. However, further investigations in other immunocompromised states must validate these results before considering human clinical trials.
Collapse
Affiliation(s)
- Vara Prasad Saka
- Department of Pharmacology, Drug Standardization, Dr. Anjali Chatterji Regional Research Institute for Homeopathy, Under Central Council for Research in Homeopathy, Ministry of AYUSH, Kolkata 700035, West Bengal, India
| | - Narasimha Kumar G V
- Department of Pharmacology, Drug Standardization, Dr. Anjali Chatterji Regional Research Institute for Homeopathy, Under Central Council for Research in Homeopathy, Ministry of AYUSH, Kolkata 700035, West Bengal, India
| | - Bharat Kumar Reddy Sanapalli
- Department of Pharmacology, School of Pharmacy and Technology Management, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be-University, Jadcherla 509301, Hyderabad, India
| | - Abanti Goswami
- Department of Pharmacology, Drug Standardization, Dr. Anjali Chatterji Regional Research Institute for Homeopathy, Under Central Council for Research in Homeopathy, Ministry of AYUSH, Kolkata 700035, West Bengal, India
| | - Anirban Roy
- Virology Laboratory, Dr. Anjali Chatterji Regional Research Institute for Homeopathy, Rajendra Chatterjee Road, Kolkata 700035, West Bengal, India
| | - Anurag Agrawal
- Department of Pharmacology, Ram-Eesh Institute of Vocational and Technical Education, Greater Noida 201310, Uttar Pradesh, India
| | - Pankaj Gupta
- Department of Pharmacology, Drug Standardization, Dr. D P Rastogi Central Research Institute for Homeopathy, Under Central Council for Research in Homeopathy, Ministry of AYUSH, Noida 201301, Uttar Pradesh, India
| | - Digvijay Verma
- Drug Standardization, Central Council for Research in Homeopathy, Ministry of AYUSH, Janakpuri 110058, New Delhi, India
| | - Subhash Kaushik
- Director General, Central Council for Research in Homeopathy, Ministry of AYUSH, Governmentof India, Janakpuri 110058, New Delhi, India
| |
Collapse
|
10
|
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
|
11
|
Li X, Fu S, Cheng H, Ma M, Song Z, Li J, Wu S, Zhang C, Wang X, Tang M, Pu X, Ji Q, Liang J, Zhao Z, Körner H, Li B, Shao M, Wang H. Differentiation of Type 17 Mucosal-Associated Invariant T Cells in Circulation Contributes to the Severity of Sepsis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1248-1261. [PMID: 38599461 DOI: 10.1016/j.ajpath.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/05/2024] [Accepted: 03/15/2024] [Indexed: 04/12/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells are essential in defending against infection. Sepsis is a systemic inflammatory response to infection and a leading cause of death. The relationship between the overall competency of the host immune response and disease severity is not fully elucidated. This study identified a higher proportion of circulating MAIT17 with expression of IL-17A and retinoic acid receptor-related orphan receptor γt in patients with sepsis. The proportion of MAIT17 was correlated with the severity of sepsis. Single-cell RNA-sequencing analysis revealed an enhanced expression of lactate dehydrogenase A (LDHA) in MAIT17 in patients with sepsis. Cell-culture experiments demonstrated that phosphoinositide 3-kinase-LDHA signaling was required for retinoic acid receptor-related orphan receptor γt expression in MAIT17. Finally, the elevated levels of plasma IL-18 promoted the differentiation of circulating MAIT17 cells in sepsis. In summary, this study reveals a new role of circulating MAIT17 in promoting sepsis severity and suggests the phosphoinositide 3-kinase-LDHA signaling as a driving force in MAIT17 responses.
Collapse
Affiliation(s)
- Xinying Li
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; School of Life Sciences, Anhui Medical University, Hefei, China
| | - Sicheng Fu
- School of Basic Medical Sciences, University of Science and Technology of China, Hefei, China
| | - Hao Cheng
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
| | - Min Ma
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zijian Song
- School of Pharmacy, Anhui Medical University, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Jun Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Shuang Wu
- School of Pharmacy, Anhui Medical University, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China
| | - Chong Zhang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xiaoxia Wang
- School of Public Health, Anhui Medical University, Hefei, China
| | - Maoyu Tang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China; Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, China
| | - Xuexue Pu
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Qiang Ji
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jinquan Liang
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Zhibin Zhao
- Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Heinrich Körner
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Departments of Respiratory and Critical Care Medicine and Thoracic Surgery, Ruijin Hospital, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Shao
- Department of Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China.
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, China; Inflammation and Immune Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, China.
| |
Collapse
|
12
|
Smits HH, Jochems SP. Diverging patterns in innate immunity against respiratory viruses during a lifetime: lessons from the young and the old. Eur Respir Rev 2024; 33:230266. [PMID: 39009407 PMCID: PMC11262623 DOI: 10.1183/16000617.0266-2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 04/16/2024] [Indexed: 07/17/2024] Open
Abstract
Respiratory viral infections frequently lead to severe respiratory disease, particularly in vulnerable populations such as young children, individuals with chronic lung conditions and older adults, resulting in hospitalisation and, in some cases, fatalities. The innate immune system plays a crucial role in monitoring for, and initiating responses to, viruses, maintaining a state of preparedness through the constant expression of antimicrobial defence molecules. Throughout the course of infection, innate immunity remains actively involved, contributing to viral clearance and damage control, with pivotal contributions from airway epithelial cells and resident and newly recruited immune cells. In instances where viral infections persist or are not effectively eliminated, innate immune components prominently contribute to the resulting pathophysiological consequences. Even though both young children and older adults are susceptible to severe respiratory disease caused by various respiratory viruses, the underlying mechanisms may differ significantly. Children face the challenge of developing and maturing their immunity, while older adults contend with issues such as immune senescence and inflammaging. This review aims to compare the innate immune responses in respiratory viral infections across both age groups, identifying common central hubs that could serve as promising targets for innovative therapeutic and preventive strategies, despite the apparent differences in underlying mechanisms.
Collapse
Affiliation(s)
- Hermelijn H Smits
- Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center, Leiden, The Netherlands
| | - Simon P Jochems
- Leiden University Center of Infectious Disease (LU-CID), Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
13
|
Hou J, Yang X, Xie S, Zhu B, Zha H. Circulating T cells: a promising biomarker of anti-PD-(L)1 therapy. Front Immunol 2024; 15:1371559. [PMID: 38576625 PMCID: PMC10991692 DOI: 10.3389/fimmu.2024.1371559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
Anti-PD-(L)1 therapy has shown great efficacy in some patients with cancer. However, a significant proportion of patients with cancer do not respond to it. Another unmet clinical need for anti-PD-(L)1 therapy is the dynamic monitoring of treatment effects. Therefore, identifying biomarkers that can stratify potential responders before PD-(L)1 treatment and timely monitoring of the efficacy of PD-(L)1 treatment are crucial in the clinical setting. The identification of biomarkers by liquid biopsy has attracted considerable attention. Among the identified biomarkers, circulating T cells are one of the most promising because of their indispensable contribution to anti-PD-(L)1 therapy. The present review aimed to thoroughly explore the potential of circulating T cells as biomarkers of anti-PD-(L)1 therapy and its advantages and limitations.
Collapse
Affiliation(s)
- Junlei Hou
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Xuezhi Yang
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Shuanglong Xie
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Bo Zhu
- Institute of Cancer, Xinqiao Hospital, Third Military Medical University, Chongqing, China
- Chongqing Key Laboratory of Immunotherapy, Xinqiao Hospital, Third Military Medical University, Chongqing, China
| | - Haoran Zha
- Department of Oncology, PLA Rocket Force Characteristic Medical Center, Beijing, China
| |
Collapse
|
14
|
Yigit M, Basoglu OF, Unutmaz D. Mucosal-associated invariant T cells in cancer: dual roles, complex interactions and therapeutic potential. Front Immunol 2024; 15:1369236. [PMID: 38545100 PMCID: PMC10965779 DOI: 10.3389/fimmu.2024.1369236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 02/26/2024] [Indexed: 04/17/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells play diverse roles in cancer, infectious diseases, and immunotherapy. This review explores their intricate involvement in cancer, from early detection to their dual functions in promoting inflammation and mediating anti-tumor responses. Within the solid tumor microenvironment (TME), MAIT cells can acquire an 'exhausted' state and secrete tumor-promoting cytokines. On the other hand, MAIT cells are highly cytotoxic, and there is evidence that they may have an anti-tumor immune response. The frequency of MAIT cells and their subsets has also been shown to have prognostic value in several cancer types. Recent innovative approaches, such as programming MAIT cells with chimeric antigen receptors (CARs), provide a novel and exciting approach to utilizing these cells in cell-based cancer immunotherapy. Because MAIT cells have a restricted T cell receptor (TCR) and recognize a common antigen, this also mitigates potential graft-versus-host disease (GVHD) and opens the possibility of using allogeneic MAIT cells as off-the-shelf cell therapies in cancer. Additionally, we outline the interactions of MAIT cells with the microbiome and their critical role in infectious diseases and how this may impact the tumor responses of these cells. Understanding these complex roles can lead to novel therapeutic strategies harnessing the targeting capabilities of MAIT cells.
Collapse
Affiliation(s)
- Mesut Yigit
- Human Immunology Laboratory, Acibadem University School of Medicine, Istanbul, Türkiye
| | - Omer Faruk Basoglu
- Human Immunology Laboratory, Acibadem University School of Medicine, Istanbul, Türkiye
| | - Derya Unutmaz
- Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| |
Collapse
|
15
|
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
|
16
|
Lin X, Wang Y, He Y. Mucosal-associated invariant T cells in infectious diseases of respiratory system: recent advancements and applications. J Inflamm (Lond) 2024; 21:6. [PMID: 38419084 PMCID: PMC10902946 DOI: 10.1186/s12950-024-00376-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 02/01/2024] [Indexed: 03/02/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are an atypical subset of T lymphocytes, which have a highly conserved semi-constant αβ chain of T-cell receptor (TCR) and recognize microbe-derived vitamin B metabolites via major histocompatibility complex class I related-1 molecule (MR1). MAIT cells get activated mainly through unique TCR-dependent and TCR-independent pathways, and express multiple functional and phenotypic traits, including innate-like functionality, T helper (Th) 1 cell immunity, Th 17 cell immunity, and tissue homing. Given the functions, MAIT cells are extensively reported to play a key role in mucosal homeostasis and infectious diseases. In the current work, we review the basic characteristics of MAIT cells and their roles in mucosal homeostasis and development of respiratory infectious diseases as well as their potential therapeutic targets.
Collapse
Affiliation(s)
- Xue Lin
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Ye Wang
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yanqi He
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
17
|
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
|
18
|
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
|
19
|
Kwon DI, Park S, Jeong YL, Kim YM, Min J, Lee C, Choi JA, Choi YH, Kong HJ, Choi Y, Baek S, Lee KJ, Kang YW, Jeong C, You G, Oh Y, Im SK, Song M, Kim JK, Chang J, Choi D, Lee SW. Fc-fused IL-7 provides broad antiviral effects against respiratory virus infections through IL-17A-producing pulmonary innate-like T cells. Cell Rep Med 2024; 5:101362. [PMID: 38232693 PMCID: PMC10829794 DOI: 10.1016/j.xcrm.2023.101362] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 10/15/2023] [Accepted: 12/12/2023] [Indexed: 01/19/2024]
Abstract
Repeated pandemics caused by the influenza virus and severe acute respiratory syndrome coronavirus (SARS-CoV) have resulted in serious problems in global public health, emphasizing the need for broad-spectrum antiviral therapeutics against respiratory virus infections. Here, we show the protective effects of long-acting recombinant human interleukin-7 fused with hybrid Fc (rhIL-7-hyFc) against major respiratory viruses, including influenza virus, SARS-CoV-2, and respiratory syncytial virus. Administration of rhIL-7-hyFc in a therapeutic or prophylactic regimen induces substantial antiviral effects. During an influenza A virus (IAV) infection, rhIL-7-hyFc treatment increases pulmonary T cells composed of blood-derived interferon γ (IFNγ)+ conventional T cells and locally expanded IL-17A+ innate-like T cells. Single-cell RNA transcriptomics reveals that rhIL-7-hyFc upregulates antiviral genes in pulmonary T cells and induces clonal expansion of type 17 innate-like T cells. rhIL-7-hyFc-mediated disease prevention is dependent on IL-17A in both IAV- and SARS-CoV-2-infected mice. Collectively, we suggest that rhIL-7-hyFc can be used as a broadly active therapeutic for future respiratory virus pandemic.
Collapse
Affiliation(s)
- Dong-Il Kwon
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Subin Park
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Yujin L Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Young-Min Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Jeongyong Min
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Changhyung Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Jung-Ah Choi
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Yoon Ha Choi
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Hyun-Jung Kong
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Youngwon Choi
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Seungtae Baek
- Research Institute of NeoImmuneTech Co., Ltd., Pohang 37666, Republic of Korea
| | - Kun-Joo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Yeon-Woo Kang
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Chaerim Jeong
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Gihoon You
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Youngsik Oh
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Sun-Kyoung Im
- Research Institute of NeoImmuneTech Co., Ltd., Pohang 37666, Republic of Korea
| | - Manki Song
- Science Unit, International Vaccine Institute, Seoul 08826, Republic of Korea
| | - Jong Kyoung Kim
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea
| | - Jun Chang
- Graduate School of Pharmaceutical Sciences, Ewha Womans University, Seoul 03760, Republic of Korea
| | - Donghoon Choi
- Research Institute of NeoImmuneTech Co., Ltd., Pohang 37666, Republic of Korea.
| | - Seung-Woo Lee
- Department of Life Sciences, Pohang University of Science and Technology, Pohang 37666, Republic of Korea.
| |
Collapse
|
20
|
Wei L, Chen Z, Lv Q. Mucosal-associated invariant T cells display both pathogenic and protective roles in patients with inflammatory bowel diseases. Amino Acids 2023; 55:1819-1827. [PMID: 37819474 DOI: 10.1007/s00726-023-03344-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 09/26/2023] [Indexed: 10/13/2023]
Abstract
An important subtype of the innate-like T lymphocytes is mucosal-associated invariant T (MAIT) cells expressing a semi-invariant T cell receptor α (TCR-α) chain. MAIT cells could be activated mainly by TCR engagement or cytokines. They have been found to have essential roles in various immune mediated. There have been growing preclinical and clinical findings that show an association between MAIT cells and the physiopathology of inflammatory bowel diseases (IBD). Of note, published reports demonstrate contradictory findings regarding the role of MAIT cells in IBD patients. A number of reports suggests a protective effect, whereas others show a pathogenic impact. The present review article aimed to explore and discuss the findings of experimental and clinical investigations evaluating the effects of MAIT cells in IBD subjects and animal models. Findings indicate that MAIT cells could exert opposite effects in the course of IBD, including an anti-inflammatory protective effect of blood circulating MAIT cells and an effector pathogenic effect of colonic MAIT cells. Another important finding is that blood levels of MAIT cells can be considered as a potential biomarker in IBD patients.
Collapse
Affiliation(s)
- Lei Wei
- Department of General Surgery, Pudong New District Gongli Hospital of Shanghai, Shanghai, 200120, China
| | - Zhigang Chen
- Department of General Surgery, Pudong New District Gongli Hospital of Shanghai, Shanghai, 200120, China
| | - Qiang Lv
- Department of General Surgery, Pudong New District Gongli Hospital of Shanghai, Shanghai, 200120, China.
| |
Collapse
|
21
|
Westmeier J, Brochtrup A, Paniskaki K, Karakoese Z, Werner T, Sutter K, Dolff S, Limmer A, Mittermüller D, Liu J, Zheng X, Koval T, Kaidashev I, Berger MM, Herbstreit F, Brenner T, Witzke O, Trilling M, Lu M, Yang D, Babel N, Westhoff T, Dittmer U, Zelinskyy G. Macrophage migration inhibitory factor receptor CD74 expression is associated with expansion and differentiation of effector T cells in COVID-19 patients. Front Immunol 2023; 14:1236374. [PMID: 37946732 PMCID: PMC10631787 DOI: 10.3389/fimmu.2023.1236374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 10/02/2023] [Indexed: 11/12/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused millions of COVID-19 cases and deaths worldwide. Severity of pulmonary pathologies and poor prognosis were reported to be associated with the activation non-virus-specific bystander T cells. In addition, high concentrations of the macrophage migration inhibitory factor (MIF) were found in serum of COVID-19 patients. We hypothesized that these two pathogenic factors might be related and analyzed the expression of receptors for MIF on T cells in COVID-19. T cells from PBMCs of hospitalized patients with mild and severe COVID-19 were characterized. A significantly higher proportion of CD4+ and CD8+ T cells from COVID-19 patients expressed CD74 on the cell surface compared to healthy controls. To induce intracellular signaling upon MIF binding, CD74 forms complexes with CD44, CXCR2, or CXCR4. The vast majority of CD74+ T cells expressed CD44, whereas expression of CXCR2 and CXCR4 was low in controls but increased upon SARS-CoV-2 infection. Hence, T cells in COVID-19 patients express receptors that render them responsive to MIF. A detailed analysis of CD74+ T cell populations revealed that most of them had a central memory phenotype early in infection, while cells with an effector and effector memory phenotype arose later during infection. Furthermore, CD74+ T cells produced more cytotoxic molecules and proliferation markers. Our data provide new insights into the MIF receptor and co-receptor repertoire of bystander T cells in COVID-19 and uncovers a novel and potentially druggable aspect of the immunological footprint of SARS-CoV-2.
Collapse
Affiliation(s)
- Jaana Westmeier
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Annika Brochtrup
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Krystallenia Paniskaki
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Center for Translational Medicine, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
| | - Zehra Karakoese
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tanja Werner
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Kathrin Sutter
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Sebastian Dolff
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Andreas Limmer
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- Department of Pediatric Heart Surgery, Friedrich-Alexander- Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Daniela Mittermüller
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Jia Liu
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
- Department of Infectious Diseases, Union Hospital of Tonji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Xin Zheng
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
- Department of Infectious Diseases, Union Hospital of Tonji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Tetiana Koval
- Department of Infectious Diseases with Epidemiology, Poltava State Medical University, Poltava, Ukraine
| | - Igor Kaidashev
- Department of Internal Medicine №3 with Phthisiology, Poltava State Medical University, Poltava, Ukraine
| | - Marc Moritz Berger
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Frank Herbstreit
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Thorsten Brenner
- Department of Anesthesiology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Oliver Witzke
- Department of Infectious Diseases, West German Centre of Infectious Diseases, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Mengji Lu
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Dongliang Yang
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
- Department of Infectious Diseases, Union Hospital of Tonji Medical College, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Nina Babel
- Center for Translational Medicine, Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, Herne, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin-Brandenburg Center for Regenerative Therapies, Berlin, Germany
| | - Timm Westhoff
- Medical Department I, Marien Hospital Herne, University Hospital of the Ruhr University of Bochum, Herne, Germany
| | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| | - Gennadiy Zelinskyy
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
- Joint International Laboratory of Infection and Immunity, Huazhong University of Science and Technology (HUST), Wuhan, China
| |
Collapse
|
22
|
Wang X, Liang M, Song P, Guan W, Shen X. Mucosal-associated invariant T cells in digestive tract: Local guardians or destroyers? Immunology 2023; 170:167-179. [PMID: 37132045 DOI: 10.1111/imm.13653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 04/17/2023] [Indexed: 05/04/2023] Open
Abstract
Mucosa-associated invariant T cells (MAIT) are a class of innate-like T lymphocytes mainly presenting CD8+ phenotype with a semi-invariant αβ T-cell receptor, which specifically recognises MR1-presented biosynthetic derivatives of riboflavin synthesis produced by various types of microbiomes. As innate-like T lymphocytes, MAIT can be activated by a variety of cytokines, leading to immediate immune responses to infection and tumour cues. As an organ that communicates with the external environment, the digestive tract, especially the gastrointestinal tract, contains abundant microbial populations. Communication between MAIT and local microbiomes is important for the homeostasis of mucosal immunity. In addition, accumulating evidence suggests changes in the abundance and structure of the microbial community during inflammation and tumorigenesis plays a critical role in disease progress partly through their impact on MAIT development and function. Therefore, it is essential for the understanding of MAIT response and their interaction with microbiomes in the digestive tract. Here, we summarised MAIT characteristics in the digestive tract and its alteration facing inflammation and tumour, raising that targeting MAIT can be a candidate for treatment of gastrointestinal diseases.
Collapse
Affiliation(s)
- Xingzhou Wang
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Mengjie Liang
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Peng Song
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Wenxian Guan
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Xiaofei Shen
- Department of General Surgery, Affiliated Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
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
|
26
|
Finn CM, Dhume K, Prokop E, Strutt TM, McKinstry KK. STAT1 Controls the Functionality of Influenza-Primed CD4 T Cells but Therapeutic STAT4 Engagement Maximizes Their Antiviral Impact. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1292-1304. [PMID: 36961447 PMCID: PMC10121883 DOI: 10.4049/jimmunol.2200407] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 02/22/2023] [Indexed: 03/25/2023]
Abstract
It is generally accepted that influenza A virus (IAV) infection promotes a Th1-like CD4 T cell response and that this effector program underlies its protective impact. Canonical Th1 polarization requires cytokine-mediated activation of the transcription factors STAT1 and STAT4 that synergize to maximize the induction of the "master regulator" Th1 transcription factor, T-bet. Here, we determine the individual requirements for these transcription factors in directing the Th1 imprint primed by influenza infection in mice by tracking virus-specific wild-type or T-bet-deficient CD4 T cells in which STAT1 or STAT4 is knocked out. We find that STAT1 is required to protect influenza-primed CD4 T cells from NK cell-mediated deletion and for their expression of hallmark Th1 attributes. STAT1 is also required to prevent type I IFN signals from inhibiting the induction of the Th17 master regulator, Rorγt, in Th17-prone T-bet-/- cells responding to IAV. In contrast, STAT4 expression does not appreciably impact the phenotypic or functional attributes of wild-type or T-bet-/- CD4 T cell responses. However, cytokine-mediated STAT4 activation in virus-specific CD4 T cells enhances their Th1 identity in a T-bet-dependent manner, indicating that influenza infection does not promote maximal Th1 induction. Finally, we show that the T-bet-dependent protective capacity of CD4 T cell effectors against IAV is optimized by engaging both STAT1 and STAT4 during Th1 priming, with important implications for vaccine strategies aiming to generate T cell immunity.
Collapse
Affiliation(s)
- Caroline M. Finn
- Burnett School of Biomedical Sciences, Division of Immunity and Pathogenesis, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Kunal Dhume
- Burnett School of Biomedical Sciences, Division of Immunity and Pathogenesis, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Emily Prokop
- Burnett School of Biomedical Sciences, Division of Immunity and Pathogenesis, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - Tara M. Strutt
- Burnett School of Biomedical Sciences, Division of Immunity and Pathogenesis, College of Medicine, University of Central Florida, Orlando, FL, USA
| | - K. Kai McKinstry
- Burnett School of Biomedical Sciences, Division of Immunity and Pathogenesis, College of Medicine, University of Central Florida, Orlando, FL, USA
| |
Collapse
|
27
|
Joyce S, Okoye GD, Driver JP. Die Kämpfe únd schláchten-the struggles and battles of innate-like effector T lymphocytes with microbes. Front Immunol 2023; 14:1117825. [PMID: 37168859 PMCID: PMC10165076 DOI: 10.3389/fimmu.2023.1117825] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/22/2023] [Indexed: 05/13/2023] Open
Abstract
The large majority of lymphocytes belong to the adaptive immune system, which are made up of B2 B cells and the αβ T cells; these are the effectors in an adaptive immune response. A multitudinous group of lymphoid lineage cells does not fit the conventional lymphocyte paradigm; it is the unconventional lymphocytes. Unconventional lymphocytes-here called innate/innate-like lymphocytes, include those that express rearranged antigen receptor genes and those that do not. Even though the innate/innate-like lymphocytes express rearranged, adaptive antigen-specific receptors, they behave like innate immune cells, which allows them to integrate sensory signals from the innate immune system and relay that umwelt to downstream innate and adaptive effector responses. Here, we review natural killer T cells and mucosal-associated invariant T cells-two prototypic innate-like T lymphocytes, which sense their local environment and relay that umwelt to downstream innate and adaptive effector cells to actuate an appropriate host response that confers immunity to infectious agents.
Collapse
Affiliation(s)
- Sebastian Joyce
- Department of Veterans Affairs, Tennessee Valley Healthcare Service, Nashville, TN, United States
- Department of Pathology, Microbiology and Immunology, The Vanderbilt Institute for Infection, Immunology and Inflammation and Vanderbilt Center for Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Gosife Donald Okoye
- Department of Pathology, Microbiology and Immunology, The Vanderbilt Institute for Infection, Immunology and Inflammation and Vanderbilt Center for Immunology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - John P. Driver
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States
| |
Collapse
|
28
|
Habel JR, Chua BY, Kedzierski L, Selva KJ, Damelang T, Haycroft ER, Nguyen TH, Koay HF, Nicholson S, McQuilten HA, Jia X, Allen LF, Hensen L, Zhang W, van de Sandt CE, Neil JA, Pragastis K, Lau JS, Jumarang J, Allen EK, Amanant F, Krammer F, Wragg KM, Juno JA, Wheatley AK, Tan HX, Pell G, Walker S, Audsley J, Reynaldi A, Thevarajan I, Denholm JT, Subbarao K, Davenport MP, Hogarth PM, Godfrey DI, Cheng AC, Tong SY, Bond K, Williamson DA, McMahon JH, Thomas PG, Pannaraj PS, James F, Holmes NE, Smibert OC, Trubiano JA, Gordon CL, Chung AW, Whitehead CL, Kent SJ, Lappas M, Rowntree LC, Kedzierska K. Immune profiling of SARS-CoV-2 infection during pregnancy reveals NK cell and γδ T cell perturbations. JCI Insight 2023; 8:e167157. [PMID: 37036008 PMCID: PMC10132165 DOI: 10.1172/jci.insight.167157] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/15/2023] [Indexed: 04/11/2023] Open
Abstract
Pregnancy poses a greater risk for severe COVID-19; however, underlying immunological changes associated with SARS-CoV-2 during pregnancy are poorly understood. We defined immune responses to SARS-CoV-2 in unvaccinated pregnant and nonpregnant women with acute and convalescent COVID-19, quantifying 217 immunological parameters. Humoral responses to SARS-CoV-2 were similar in pregnant and nonpregnant women, although our systems serology approach revealed distinct antibody and FcγR profiles between pregnant and nonpregnant women. Cellular analyses demonstrated marked differences in NK cell and unconventional T cell activation dynamics in pregnant women. Healthy pregnant women displayed preactivated NK cells and γδ T cells when compared with healthy nonpregnant women, which remained unchanged during acute and convalescent COVID-19. Conversely, nonpregnant women had prototypical activation of NK and γδ T cells. Activation of CD4+ and CD8+ T cells and T follicular helper cells was similar in SARS-CoV-2-infected pregnant and nonpregnant women, while antibody-secreting B cells were increased in pregnant women during acute COVID-19. Elevated levels of IL-8, IL-10, and IL-18 were found in pregnant women in their healthy state, and these cytokine levels remained elevated during acute and convalescent COVID-19. Collectively, we demonstrate perturbations in NK cell and γδ T cell activation in unvaccinated pregnant women with COVID-19, which may impact disease progression and severity during pregnancy.
Collapse
Affiliation(s)
- Jennifer R. Habel
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Brendon Y. Chua
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| | - Lukasz Kedzierski
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Melbourne, Victoria, Australia
| | - Kevin J. Selva
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Timon Damelang
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Ebene R. Haycroft
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Thi H.O. Nguyen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hui-Fern Koay
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Suellen Nicholson
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hayley A. McQuilten
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Xiaoxiao Jia
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Lilith F. Allen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Luca Hensen
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Wuji Zhang
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Carolien E. van de Sandt
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jessica A. Neil
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Katherine Pragastis
- Department of Infectious Diseases, Alfred Health, Monash University, Melbourne, Victoria, Australia
| | - Jillian S.Y. Lau
- Department of Infectious Diseases, Alfred Health, Monash University, Melbourne, Victoria, Australia
- Department of Infectious Diseases, Eastern Health, Box Hill, Victoria, Australia
| | - Jaycee Jumarang
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, California, USA
| | - E. Kaitlynn Allen
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Fatima Amanant
- Department of Microbiology, and
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | | | - Kathleen M. Wragg
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jennifer A. Juno
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Adam K. Wheatley
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, Victoria, Australia
| | - Hyon-Xhi Tan
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Gabrielle Pell
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Susan Walker
- Mercy Perinatal Research Centre, Mercy Hospital for Women, Heidelberg, Victoria, Australia
| | - Jennifer Audsley
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Arnold Reynaldi
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - Irani Thevarajan
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Justin T. Denholm
- Department of Infectious Diseases, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Kanta Subbarao
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- WHO Collaborating Centre for Reference and Research on Influenza, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Miles P. Davenport
- Kirby Institute, University of New South Wales, Sydney, New South Wales, Australia
| | - P. Mark Hogarth
- Immune Therapies Laboratory, Burnet Institute, Melbourne, Victoria, Australia
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Clinical Pathology, University of Melbourne, Parkville, Victoria, Australia
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Allen C. Cheng
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Infection Prevention and Healthcare Epidemiology Unit, Alfred Health, and Monash Infectious Diseases, Monash Health, Melbourne, Victoria, Australia
| | - Steven Y.C. Tong
- Victorian Infectious Diseases Service, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Katherine Bond
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Microbiology, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Deborah A. Williamson
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Microbiology, Royal Melbourne Hospital, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - James H. McMahon
- Department of Infectious Diseases, Alfred Health, Monash University, Melbourne, Victoria, Australia
| | - Paul G. Thomas
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, USA
| | - Pia S. Pannaraj
- Division of Infectious Diseases, Children’s Hospital Los Angeles, Los Angeles, California, USA
- Departments of Pediatrics, Molecular Microbiology and Immunology, Keck School of Medicine, UCLA, Los Angeles, California, USA
| | - Fiona James
- Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
| | - Natasha E. Holmes
- Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
- Department of Critical Care, University of Melbourne, Parkville, Victoria, Australia
- Data Analytics Research and Evaluation Centre, Austin Health, University of Melbourne, Heidelberg, Victoria, Australia
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
| | - Olivia C. Smibert
- Departments of Pediatrics, Molecular Microbiology and Immunology, Keck School of Medicine, UCLA, Los Angeles, California, USA
- Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
- Department of Infectious Diseases, and
- National Centre for Infections in Cancer, Peter McCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Jason A. Trubiano
- Centre for Antibiotic Allergy and Research, Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
- Department of Infectious Diseases, and
- National Centre for Infections in Cancer, Peter McCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Medicine (Austin Health), University of Melbourne, Heidelberg, Victoria, Australia
| | - Claire L. Gordon
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Department of Infectious Diseases, Austin Health, Heidelberg, Victoria, Australia
| | - Amy W. Chung
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Clare L. Whitehead
- Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
- Pregnancy Research Centre, Royal Women’s Hospital, Parkville, Victoria, Australia
| | - Stephen J. Kent
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, University of Melbourne, Melbourne, Victoria, Australia
- Melbourne Sexual Health Centre, Infectious Diseases Department, Alfred Health, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Martha Lappas
- Obstetrics, Nutrition and Endocrinology Group, Department of Obstetrics and Gynaecology, University of Melbourne, Victoria, Australia
| | - Louise C. Rowntree
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Katherine Kedzierska
- Department of Microbiology and Immunology, University of Melbourne, Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Global Institution for Collaborative Research and Education (GI-CoRE), Hokkaido University, Sapporo, Japan
| |
Collapse
|
29
|
Si Y, Zhang Y, Zuloaga K, Yang Q. The role of innate lymphocytes in regulating brain and cognitive function. Neurobiol Dis 2023; 179:106061. [PMID: 36870457 PMCID: PMC11194859 DOI: 10.1016/j.nbd.2023.106061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 02/26/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023] Open
Abstract
Mounting evidence indicates complex interaction between the immune system and the nervous system, challenging the traditional view about the immune privilege of the brain. Innate lymphoid cells (ILCs) and innate-like T cells are unique families of immune cells that functionally mirror traditional T cells but may function via antigen- and T cell antigen receptor (TCR)-independent mechanisms. Recent work indicates that various ILCs and innate-like T cell subsets are present in the brain barrier tissue, where they play important roles in regulating brain barrier integrity, brain homeostasis and cognitive function. In this review, we discuss recent advances in understanding the intricate roles for innate and innate-like lymphocytes in regulating brain and cognitive function.
Collapse
Affiliation(s)
- Youwen Si
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Yuanyue Zhang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA
| | - Kristen Zuloaga
- Department of Neuroscience and Experimental Therapeutics, Albany Medical College, Albany, NY, 12208, USA
| | - Qi Yang
- Child Health Institute of New Jersey, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; Rutgers Institute for Translational Medicine and Science, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA; Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ 08901, USA.
| |
Collapse
|
30
|
Zhang Y, Wang M, Zhang X, Tang K, Zhang C, Jia X, Hu H, Liu H, Li N, Zhuang R, Jin B, Ma Y, Zhang Y. HTNV infection induces activation and deficiency of CD8+MAIT cells in HFRS patients. Clin Exp Immunol 2023; 211:1-14. [PMID: 36480318 PMCID: PMC9993462 DOI: 10.1093/cei/uxac111] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 10/19/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022] Open
Abstract
Hantaan virus (HTNV) infection causes an epidemic of hemorrhagic fever with renal syndrome (HFRS) mainly in Asia. Mucosal-associated invariant T (MAIT) cells are innate-like T lymphocytes known to play an important role in innate host defense during virus infection. However, their roles and phenotypes during HTNV infection have not yet been explored. We characterized CD8+MAIT cells from HFRS patients based on scRNA-seq data combined with flow cytometry data. We showed that HTNV infection caused the loss and activation of CD8+MAIT cells in the peripheral blood, which were correlated with disease severity. The production of granzyme B and IFN-γ from CD8+MAIT cells and the limitation of HTNV replication in endothelia cells indicated the anti-viral property of CD8+MAIT cells. In addition, in vitro infection of MAIT cells by HTNV or HTNV-exposed monocytes showed that the activation of MAIT cells was IL-18 mediated. In conclusion, this study identified, for the first time, gene expression profiles of MAIT cells, provided underlying molecular mechanisms for activation of MAIT cells during HTNV infection, and suggested a potential anti-viral role of MAIT cells in HFRS.
Collapse
Affiliation(s)
- Yusi Zhang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, China
| | - Meng Wang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, China
- Department of Immunology, School of Basic Medical Sciences, Yan’an university, Yan’an 716000, China
| | - Xiyue Zhang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, China
- Department of Pathogenic Biology, School of Basic Medical Sciences, Yan’an university, Yan’an 716000, China
| | - Kang Tang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, China
| | - Chunmei Zhang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, China
| | | | - Haifeng Hu
- Center for Infectious Diseases, Second Affiliated Hospital of Air Force Medical University (Fourth Military Medical University), Xi’an 710038, China
| | - He Liu
- Department of Microbiology, School of Basic Medicine, Fourth Military Medical University, Xi΄an 710032, China
| | - Na Li
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi’an 710032, Shaanxi, China
| | - Ran Zhuang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, China
| | - Boquan Jin
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, China
| | - Ying Ma
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, China
| | - Yun Zhang
- Department of Immunology, School of Basic Medicine, Fourth Military Medical University, Xi’an 710032, China
| |
Collapse
|
31
|
Korkmaz FT, Traber KE. Innate immune responses in pneumonia. Pneumonia (Nathan) 2023; 15:4. [PMID: 36829255 PMCID: PMC9957695 DOI: 10.1186/s41479-023-00106-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 01/05/2023] [Indexed: 02/26/2023] Open
Abstract
The lungs are an immunologically unique environment; they are exposed to innumerable pathogens and particulate matter daily. Appropriate clearance of pathogens and response to pollutants is required to prevent overwhelming infection, while preventing tissue damage and maintaining efficient gas exchange. Broadly, the innate immune system is the collection of immediate, intrinsic immune responses to pathogen or tissue injury. In this review, we will examine the innate immune responses of the lung, with a particular focus on their role in pneumonia. We will discuss the anatomic barriers and antimicrobial proteins of the lung, pathogen and injury recognition, and the role of leukocytes (macrophages, neutrophils, and innate lymphocytes) and lung stromal cells in innate immunity. Throughout the review, we will focus on new findings in innate immunity as well as features that are unique to the lung.
Collapse
Affiliation(s)
- Filiz T Korkmaz
- Department of Medicine, Division of Immunology & Infectious Disease, University of Massachusetts, Worcester, MA, USA
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA
| | - Katrina E Traber
- Pulmonary Center, Boston University School of Medicine, Boston, MA, USA.
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
32
|
Courtney AN, Tian G, Metelitsa LS. Natural killer T cells and other innate-like T lymphocytes as emerging platforms for allogeneic cancer cell therapy. Blood 2023; 141:869-876. [PMID: 36347021 PMCID: PMC10023720 DOI: 10.1182/blood.2022016201] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/19/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022] Open
Abstract
T cells expressing chimeric antigen receptors (CARs) have achieved major clinical success in patients with hematologic malignancies. However, these treatments remain largely ineffective for solid cancers and require significant time and resources to be manufactured in an autologous setting. Developing alternative immune effector cells as cancer immunotherapy agents that can be employed in allogeneic settings is crucial for the advancement of cell therapy. Unlike T cells, Vα24-invariant natural killer T cells (NKTs) are not alloreactive and can therefore be generated from allogeneic donors for rapid infusion into numerous patients without the risk of graft-versus-host disease. Additionally, NKT cells demonstrate inherent advantages over T-cell products, including the ability to traffic to tumor tissues, target tumor-associated macrophages, transactivate NK cells, and cross-prime tumor-specific CD8 T cells. Both unmodified NKTs, which specifically recognize CD1d-bound glycolipid antigens expressed by certain types of tumors, and CAR-redirected NKTs are being developed as the next generation of allogeneic cell therapy products. In this review, we describe studies on the biology of NKTs and other types of innate-like T cells and summarize the clinical experiences of unmodified and CAR-redirected NKTs, including recent interim reports on allogeneic NKTs.
Collapse
Affiliation(s)
- Amy N. Courtney
- Department of Pediatrics, Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX
| | - Gengwen Tian
- Department of Pediatrics, Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX
| | - Leonid S. Metelitsa
- Department of Pediatrics, Center for Advanced Innate Cell Therapy, Baylor College of Medicine, Houston, TX
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX
| |
Collapse
|
33
|
Ashley CL, McSharry BP, McWilliam HEG, Stanton RJ, Fielding CA, Mathias RA, Fairlie DP, McCluskey J, Villadangos JA, Rossjohn J, Abendroth A, Slobedman B. Suppression of MR1 by human cytomegalovirus inhibits MAIT cell activation. Front Immunol 2023; 14:1107497. [PMID: 36845106 PMCID: PMC9950634 DOI: 10.3389/fimmu.2023.1107497] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 01/25/2023] [Indexed: 02/12/2023] Open
Abstract
Introduction The antigen presentation molecule MHC class I related protein-1 (MR1) is best characterized by its ability to present bacterially derived metabolites of vitamin B2 biosynthesis to mucosal-associated invariant T-cells (MAIT cells). Methods Through in vitro human cytomegalovirus (HCMV) infection in the presence of MR1 ligand we investigate the modulation of MR1 expression. Using coimmunoprecipitation, mass spectrometry, expression by recombinant adenovirus and HCMV deletion mutants we investigate HCMV gpUS9 and its family members as potential regulators of MR1 expression. The functional consequences of MR1 modulation by HCMV infection are explored in coculture activation assays with either Jurkat cells engineered to express the MAIT cell TCR or primary MAIT cells. MR1 dependence in these activation assays is established by addition of MR1 neutralizing antibody and CRISPR/Cas-9 mediated MR1 knockout. Results Here we demonstrate that HCMV infection efficiently suppresses MR1 surface expression and reduces total MR1 protein levels. Expression of the viral glycoprotein gpUS9 in isolation could reduce both cell surface and total MR1 levels, with analysis of a specific US9 HCMV deletion mutant suggesting that the virus can target MR1 using multiple mechanisms. Functional assays with primary MAIT cells demonstrated the ability of HCMV infection to inhibit bacterially driven, MR1-dependent activation using both neutralizing antibodies and engineered MR1 knockout cells. Discussion This study identifies a strategy encoded by HCMV to disrupt the MR1:MAIT cell axis. This immune axis is less well characterized in the context of viral infection. HCMV encodes hundreds of proteins, some of which regulate the expression of antigen presentation molecules. However the ability of this virus to regulate the MR1:MAIT TCR axis has not been studied in detail.
Collapse
Affiliation(s)
- Caroline L. Ashley
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, and the Charles Perkins Centre, The University of Sydney, Sydney, NSW, 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, Sydney, NSW, Australia
- School of Dentistry and Medical Sciences, Faculty of Science and Health, Charles Sturt University, Wagga Wagga, NSW, Australia
| | - Hamish E. G. McWilliam
- Department of Microbiology and Immunology, The Peter Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Pharmacology, Institute of Molecular Science and Biotechnology (Bio21), The University of Melbourne, Melbourne, VIC, Australia
| | - Richard J. Stanton
- Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Ceri A. Fielding
- Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
| | - Rommel A. Mathias
- Infection and Immunity Program, Department of Microbiology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC, Australia
| | - David P. Fairlie
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, The Peter Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
| | - Jose A. Villadangos
- Department of Microbiology and Immunology, The Peter Doherty Institute of Infection and Immunity, The University of Melbourne, Melbourne, VIC, Australia
- Department of Biochemistry and Pharmacology, Institute of Molecular Science and Biotechnology (Bio21), The University of Melbourne, Melbourne, VIC, Australia
| | - Jamie Rossjohn
- Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, United Kingdom
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, 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, Sydney, 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, Sydney, NSW, Australia
| |
Collapse
|
34
|
Kedia-Mehta N, Hogan AE. MAITabolism 2 - the emerging understanding of MAIT cell metabolism and their role in metabolic disease. Front Immunol 2023; 13:1108071. [PMID: 36741413 PMCID: PMC9892190 DOI: 10.3389/fimmu.2022.1108071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 12/19/2022] [Indexed: 01/20/2023] Open
Abstract
Mucosal associated invariant T (MAIT) cells are a population of unconventional innate T cells due to their non-MHC restriction and rapid effector responses. MAIT cells can recognise bacterial derived antigens presented on the MHC-like protein via their semi-restricted T cell receptor (TCR). Upon TCR triggering MAIT cells rapidly produce a range of effector molecules including cytokines, lytic granules and chemokines. This rapid and robust effector response makes MAIT cells critical in host responses against many bacterial pathogens. MAIT cells can also respond independent of their TCR via innate cytokines such as interleukin (IL)-18, triggering cytokine production, and are important in anti-viral responses. In addition to their protective role, MAIT cells have been implicated in numerous inflammatory diseases, including metabolic diseases often contributing to the pathogenesis via their robust cytokine production. Effector cells such as MAIT cells require significant amounts of energy to support their potent responses, and the type of nutrients available can dictate the functionality of the cell. Although data on MAIT cell metabolism is just emerging, several recent studies are starting to define the intrinsic metabolic requirements and regulators of MAIT cells. In this review we will outline our current understanding of MAIT cell metabolism, and outline their role in metabolic disease, and how disease-related changes in extrinsic metabolism can alter MAIT cell responses.
Collapse
Affiliation(s)
- Nidhi Kedia-Mehta
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co Kildare, Ireland
- Obesity Immunology Group, Education and Research Centre, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
| | - Andrew E Hogan
- Kathleen Lonsdale Institute for Human Health Research, Maynooth University, Maynooth, Co Kildare, Ireland
- Obesity Immunology Group, Education and Research Centre, St Vincent's University Hospital, University College Dublin, Dublin, Ireland
- National Children's Research Centre, Dublin, Ireland
| |
Collapse
|
35
|
Jiang X, Zhao Q, Huang Z, Ma F, Chen K, Li Z. Relevant mechanisms of MAIT cells involved in the pathogenesis of periodontitis. Front Cell Infect Microbiol 2023; 13:1104932. [PMID: 36896188 PMCID: PMC9988952 DOI: 10.3389/fcimb.2023.1104932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Accepted: 01/31/2023] [Indexed: 02/23/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are a group of unconventional T cells that are abundant in the human body, recognize microbial-derived vitamin B metabolites presented by MHC class I-related protein 1 (MR1), and rapidly produce proinflammatory cytokines, which are widely involved in the immune response to various infectious diseases. In the oral mucosa, MAIT cells tend to accumulate near the mucosal basal lamina and are more inclined to secrete IL-17 when activated. Periodontitis is a group of diseases that manifests mainly as inflammation of the gums and resorption of the alveolar bone due to periodontal tissue invasion by plaque bacteria on the dental surface. The course of periodontitis is often accompanied by a T-cell-mediated immune response. This paper discussed the pathogenesis of periodontitis and the potential contribution of MAIT cells to periodontitis.
Collapse
Affiliation(s)
- Xinrong Jiang
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangzhou, Guangdong, China
- College of Stomatology, Jinan University, Guangzhou, Guangdong, China
| | - Qingtong Zhao
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangzhou, Guangdong, China
- Department of Stomatology, The Sixth Affiliated Hospital of Jinan University, Dongguan, Guangdong, China
| | - Zhanyu Huang
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangzhou, Guangdong, China
- College of Stomatology, Jinan University, Guangzhou, Guangdong, China
| | - Fengyu Ma
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangzhou, Guangdong, China
- College of Stomatology, Jinan University, Guangzhou, Guangdong, China
| | - Kexiao Chen
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangzhou, Guangdong, China
- College of Stomatology, Jinan University, Guangzhou, Guangdong, China
| | - Zejian Li
- Hospital of Stomatology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangzhou, Guangdong, China
- Chaoshan Hospital, The First Affiliated Hospital of Jinan University, Chaozhou, Guangdong, China
- *Correspondence: Zejian Li,
| |
Collapse
|
36
|
Jakob J, Kröger A, Klawonn F, Bruder D, Jänsch L. Translatome analyses by bio-orthogonal non-canonical amino acid labeling reveal that MR1-activated MAIT cells induce an M1 phenotype and antiviral programming in antigen-presenting monocytes. Front Immunol 2023; 14:1091837. [PMID: 36875139 PMCID: PMC9977998 DOI: 10.3389/fimmu.2023.1091837] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
MAIT cells are multifunctional innate-like effector cells recognizing bacterial-derived vitamin B metabolites presented by the non-polymorphic MHC class I related protein 1 (MR1). However, our understanding of MR1-mediated responses of MAIT cells upon their interaction with other immune cells is still incomplete. Here, we performed the first translatome study of primary human MAIT cells interacting with THP-1 monocytes in a bicellular system. We analyzed the interaction between MAIT and THP-1 cells in the presence of the activating 5-OP-RU or the inhibitory Ac-6-FP MR1-ligand. Using bio-orthogonal non-canonical amino acid tagging (BONCAT) we were able to enrich selectively those proteins that were newly translated during MR1-dependent cellular interaction. Subsequently, newly translated proteins were measured cell-type-specifically by ultrasensitive proteomics to decipher the coinciding immune responses in both cell types. This strategy identified over 2,000 MAIT and 3,000 THP-1 active protein translations following MR1 ligand stimulations. Translation in both cell types was found to be increased by 5-OP-RU, which correlated with their conjugation frequency and CD3 polarization at MAIT cell immunological synapses in the presence of 5-OP-RU. In contrast, Ac-6-FP only regulated a few protein translations, including GSK3B, indicating an anergic phenotype. In addition to known effector responses, 5-OP-RU-induced protein translations uncovered type I and type II Interferon-driven protein expression profiles in both MAIT and THP-1 cells. Interestingly, the translatome of THP-1 cells suggested that activated MAIT cells can impact M1/M2 polarization in these cells. Indeed, gene and surface expression of CXCL10, IL-1β, CD80, and CD206 confirmed an M1-like phenotype of macrophages being induced in the presence of 5-OP-RU-activated MAIT cells. Furthermore, we validated that the Interferon-driven translatome was accompanied by the induction of an antiviral phenotype in THP-1 cells, which were found able to suppress viral replication following conjugation with MR1-activated MAIT cells. In conclusion, BONCAT translatomics extended our knowledge of MAIT cell immune responses at the protein level and discovered that MR1-activated MAIT cells are sufficient to induce M1 polarization and an anti-viral program of macrophages.
Collapse
Affiliation(s)
- Josefine Jakob
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute of Medical Microbiology and Hospital Hygiene, Infection Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Andrea Kröger
- Innate Immunity and Infection, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.,Institute of Medical Microbiology and Hospital Hygiene, Molecular Microbiology, Health Campus Immunology, Infectiology and Inflammation, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Frank Klawonn
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Dunja Bruder
- Institute of Medical Microbiology and Hospital Hygiene, Infection Immunology, Health Campus Immunology, Infectiology and Inflammation, Otto-von-Guericke University Magdeburg, Magdeburg, Germany.,Immune Regulation, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Lothar Jänsch
- Cellular Proteomics, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
37
|
Lett MJ, Mehta H, Keogh A, Jaeger T, Jacquet M, Powell K, Meier MA, Fofana I, Melhem H, Vosbeck J, Cathomas G, Heigl A, Heim MH, Burri E, Mertz KD, Niess JH, Kollmar O, Zech CJ, Ivanek R, Duthaler U, Klenerman P, Stroka D, Filipowicz Sinnreich M. Stimulatory MAIT cell antigens reach the circulation and are efficiently metabolised and presented by human liver cells. Gut 2022; 71:2526-2538. [PMID: 35058274 PMCID: PMC9664123 DOI: 10.1136/gutjnl-2021-324478] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 01/08/2022] [Indexed: 01/01/2023]
Abstract
OBJECTIVE Mucosal-associated invariant T (MAIT) cells are the most abundant T cells in human liver. They respond to bacterial metabolites presented by major histocompatibility complex-like molecule MR1. MAIT cells exert regulatory and antimicrobial functions and are implicated in liver fibrogenesis. It is not well understood which liver cells function as antigen (Ag)-presenting cells for MAIT cells, and under which conditions stimulatory Ags reach the circulation. DESIGN We used different types of primary human liver cells in Ag-presentation assays to blood-derived and liver-derived MAIT cells. We assessed MAIT cell stimulatory potential of serum from healthy subjects and patients with portal hypertension undergoing transjugular intrahepatic portosystemic shunt stent, and patients with inflammatory bowel disease (IBD). RESULTS MAIT cells were dispersed throughout healthy human liver and all tested liver cell types stimulated MAIT cells, hepatocytes being most efficient. MAIT cell activation by liver cells occurred in response to bacterial lysate and pure Ag, and was prevented by non-activating MR1 ligands. Serum derived from peripheral and portal blood, and from patients with IBD stimulated MAIT cells in MR1-dependent manner. CONCLUSION Our findings reveal previously unrecognised roles of liver cells in Ag metabolism and activation of MAIT cells, repression of which creates an opportunity to design antifibrotic therapies. The presence of MAIT cell stimulatory Ags in serum rationalises the observed activated MAIT cell phenotype in liver. Increased serum levels of gut-derived MAIT cell stimulatory ligands in patients with impaired intestinal barrier function indicate that intrahepatic Ag-presentation may represent an important step in the development of liver disease.
Collapse
Affiliation(s)
- Martin J Lett
- Department of Biomedicine, Liver Immunology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Hema Mehta
- Peter Medawar Building for Pathogen Research and Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Adrian Keogh
- Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Tina Jaeger
- Department of Biomedicine, Liver Immunology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Maxime Jacquet
- Department of Biomedicine, Liver Immunology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Kate Powell
- Peter Medawar Building for Pathogen Research and Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Marie-Anne Meier
- Department of Biomedicine, Hepatology, University Hospital Basel and University of Basel, Basel, Switzerland,Division of Gastroenterology and Hepatology, Clarunis University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Isabel Fofana
- Department of Biomedicine, Hepatology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Hassan Melhem
- Department of Biomedicine, Gastroenterology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Jürg Vosbeck
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Gieri Cathomas
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Andres Heigl
- Department of Surgery, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Markus H Heim
- Department of Biomedicine, Hepatology, University Hospital Basel and University of Basel, Basel, Switzerland,Division of Gastroenterology and Hepatology, Clarunis University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Emanuel Burri
- Gastroenterology and Hepatology, University Department of Medicine, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Kirsten D Mertz
- Institute of Pathology, Cantonal Hospital Baselland, Liestal, Switzerland
| | - Jan Hendrik Niess
- Division of Gastroenterology and Hepatology, Clarunis University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland,Department of Biomedicine, Gastroenterology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Otto Kollmar
- Division of Visceral Surgery, Clarunis University Center for Gastrointestinal and Liver Diseases, Basel, Switzerland
| | - Christoph J Zech
- Radiology and Nuclear Medicine, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Robert Ivanek
- Department of Biomedicine, Bioinformatics Core Facility, University Hospital Basel and University of Basel, Basel, Switzerland,DBM Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Urs Duthaler
- Department of Biomedicine, Clinical Pharmacology and Toxicology, University Hospital Basel and University of Basel, Basel, Switzerland
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research and Translational Gastroenterology Unit, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Deborah Stroka
- Department of Visceral Surgery and Medicine, University of Bern, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Magdalena Filipowicz Sinnreich
- Department of Biomedicine, Liver Immunology, University Hospital Basel and University of Basel, Basel, Switzerland .,Gastroenterology and Hepatology, University Department of Medicine, Cantonal Hospital Baselland, Liestal, Switzerland
| |
Collapse
|
38
|
Hinrichs AC, Kruize AA, Leavis HL, van Roon JAG. In patients with primary Sjögren's syndrome innate-like MAIT cells display upregulated IL-7R, IFN-γ, and IL-21 expression and have increased proportions of CCR9 and CXCR5-expressing cells. Front Immunol 2022; 13:1017157. [PMID: 36505431 PMCID: PMC9729251 DOI: 10.3389/fimmu.2022.1017157] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 11/04/2022] [Indexed: 11/25/2022] Open
Abstract
Introduction Mucosal-associated invariant T (MAIT) cells might play a role in B cell hyperactivity and local inflammation in primary Sjögren's syndrome (pSS), just like previously studied mucosa-associated CCR9+ and CXCR5+ T helper cells. Here, we investigated expression of CCR9, CXCR5, IL-18R and IL-7R on MAIT cells in pSS, and assessed the capacity of DMARDs to inhibit the activity of MAIT cells. Methods Circulating CD161+ and IL-18Rα+ TCRVα7.2+ MAIT cells from pSS patients and healthy controls (HC) were assessed using flow cytometry, and expression of CCR9, CXCR5, and IL-7R on MAIT cells was studied. Production of IFN-γ and IL-21 by MAIT cells was measured upon IL-7 stimulation in the presence of leflunomide (LEF) and hydroxychloroquine (HCQ). Results The numbers of CD161+ and IL-18Rα+ MAIT cells were decreased in pSS patients compared to HC. Relative increased percentages of CD4 MAIT cells in pSS patients caused significantly higher CD4/CD8 ratios in MAIT cells. The numbers of CCR9 and CXCR5-expressing MAIT cells were significantly higher in pSS patients. IL-7R expression was higher in CD8 MAIT cells as compared to all CD8 T cells, and changes in IL-7R expression correlated to several clinical parameters. The elevated production of IL-21 by MAIT cells was significantly inhibited by LEF/HCQ treatment. Conclusion Circulating CD161+ and IL-18Rα+ MAIT cell numbers are decreased in pSS patients. Given their enriched CCR9/CXCR5 expression this may facilitate migration to inflamed salivary glands known to overexpress CCL25/CXCL13. Given the pivotal role of IL-7 and IL-21 in inflammation in pSS this indicates a potential role for MAIT cells in driving pSS immunopathology.
Collapse
Affiliation(s)
- Anneline C. Hinrichs
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Aike A. Kruize
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Helen L. Leavis
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Joel A. G. van Roon
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
39
|
Shibata K, Motozono C, Nagae M, Shimizu T, Ishikawa E, Motooka D, Okuzaki D, Izumi Y, Takahashi M, Fujimori N, Wing JB, Hayano T, Asai Y, Bamba T, Ogawa Y, Furutani-Seiki M, Shirai M, Yamasaki S. Symbiotic bacteria-dependent expansion of MR1-reactive T cells causes autoimmunity in the absence of Bcl11b. Nat Commun 2022; 13:6948. [PMID: 36376329 PMCID: PMC9663695 DOI: 10.1038/s41467-022-34802-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 11/08/2022] [Indexed: 11/16/2022] Open
Abstract
MHC class I-related protein 1 (MR1) is a metabolite-presenting molecule that restricts MR1-reactive T cells including mucosal-associated invariant T (MAIT) cells. In contrast to MAIT cells, the function of other MR1-restricted T cell subsets is largely unknown. Here, we report that mice in which a T cell-specific transcription factor, B-cell lymphoma/leukemia 11B (Bcl11b), was ablated in immature thymocytes (Bcl11b∆iThy mice) develop chronic inflammation. Bcl11b∆iThy mice lack conventional T cells and MAIT cells, whereas CD4+IL-18R+ αβ T cells expressing skewed Traj33 (Jα33)+ T cell receptors (TCR) accumulate in the periphery, which are necessary and sufficient for the pathogenesis. The disorders observed in Bcl11b∆iThy mice are ameliorated by MR1-deficiency, transfer of conventional T cells, or germ-free conditions. We further show the crystal structure of the TCR expressed by Traj33+ T cells expanded in Bcl11b∆iThy mice. Overall, we establish that MR1-reactive T cells have pathogenic potential.
Collapse
Affiliation(s)
- Kensuke Shibata
- grid.268397.10000 0001 0660 7960Department of Microbiology and Immunology, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan ,grid.177174.30000 0001 2242 4849Department of Ophthalmology, Department of Ocular Pathology and Imaging Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan ,grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Chihiro Motozono
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.274841.c0000 0001 0660 6749Division of Infection and Immunity, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto, 860-0871 Japan
| | - Masamichi Nagae
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan
| | - Takashi Shimizu
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Eri Ishikawa
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan
| | - Daisuke Motooka
- grid.136593.b0000 0004 0373 3971Department of Infection Metagenomics, Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Daisuke Okuzaki
- grid.136593.b0000 0004 0373 3971Single Cell Genomics, Human Immunology, World Premier International Research Center Initiative Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Genome Information Research Center, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan
| | - Yoshihiro Izumi
- grid.177174.30000 0001 2242 4849Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| | - Masatomo Takahashi
- grid.177174.30000 0001 2242 4849Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| | - Nao Fujimori
- grid.177174.30000 0001 2242 4849Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan
| | - James B. Wing
- grid.136593.b0000 0004 0373 3971Laboratory of Human Immunology (Single Cell Immunology), World Premier International Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Experimental Immunology, World Premier International Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan
| | - Takahide Hayano
- grid.268397.10000 0001 0660 7960Department of Systems Bioinformatics, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Yoshiyuki Asai
- grid.268397.10000 0001 0660 7960Department of Systems Bioinformatics, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Takeshi Bamba
- grid.177174.30000 0001 2242 4849Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan
| | - Yoshihiro Ogawa
- grid.177174.30000 0001 2242 4849Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, 812-8582 Japan ,grid.419082.60000 0004 1754 9200Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology, Tokyo, 100-0004 Japan ,grid.27476.300000 0001 0943 978XDepartment of Molecular Medicine and Metabolism, Research Institute of Environmental Medicine, Nagoya University, Nagoya, 464-8601 Japan
| | - Makoto Furutani-Seiki
- grid.268397.10000 0001 0660 7960Systems Biochemistry in Pathology and Regeneration, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Mutsunori Shirai
- grid.268397.10000 0001 0660 7960Department of Microbiology and Immunology, Graduate School of Medicine, Yamaguchi University, Ube, 755-8505 Japan
| | - Sho Yamasaki
- grid.136593.b0000 0004 0373 3971Department of Molecular Immunology, Research Institute for Microbial Diseases, Osaka University, Suita, 565-0871 Japan ,grid.136593.b0000 0004 0373 3971Laboratory of Molecular Immunology, Immunology Frontier Research Center, Osaka University, Suita, 565-0871 Japan ,grid.177174.30000 0001 2242 4849Division of Molecular Design, Medical Institute of Bioregulation, Kyushu University, Fukuoka, 812-8582 Japan ,grid.136304.30000 0004 0370 1101Division of Molecular Immunology, Medical Mycology Research Center, Chiba University, Chiba, 260-8673 Japan
| |
Collapse
|
40
|
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
|
41
|
Han F, Gulam MY, Zheng Y, Zulhaimi NS, Sia WR, He D, Ho A, Hadadi L, Liu Z, Qin P, Lobie PE, Kamarulzaman A, Wang LF, Sandberg JK, Lewin SR, Rajasuriar R, Leeansyah E. IL7RA single nucleotide polymorphisms are associated with the size and function of the MAIT cell population in treated HIV-1 infection. Front Immunol 2022; 13:985385. [PMID: 36341446 PMCID: PMC9632172 DOI: 10.3389/fimmu.2022.985385] [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: 07/03/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022] Open
Abstract
MAIT cells are persistently depleted and functionally exhausted in HIV-1-infected patients despite long-term combination antiretroviral therapy (cART). IL-7 treatment supports MAIT cell reconstitution in vivo HIV-1-infected individuals and rescues their functionality in vitro. Single-nucleotide polymorphisms (SNPs) of the IL-7RA gene modulate the levels of soluble(s)IL-7Rα (sCD127) levels and influence bioavailability of circulating IL-7. Here we evaluate the potential influence of IL-7RA polymorphisms on MAIT cell numbers and function in healthy control (HC) subjects and HIV-1-infected individuals on long-term cART. Our findings indicate that IL-7RA haplotype 2 (H2*T), defined as T-allele carriers at the tagging SNP rs6897932, affects the size of the peripheral blood MAIT cell pool, as well as their production of cytokines and cytolytic effector proteins in response to bacterial stimulation. H2*T carriers had lower sIL-7Rα levels and higher MAIT cell frequency with enhanced functionality linked to higher expression of MAIT cell-associated transcription factors. Despite an average of 7 years on suppressive cART, MAIT cell levels and function in HIV-1-infected individuals were still significantly lower than those of HC. Notably, we observed a significant correlation between MAIT cell levels and cART duration only in HIV-1-infected individuals carrying IL-7RA haplotype 2. Interestingly, treatment with sIL-7Rα in vitro suppressed IL-7-dependent MAIT cell proliferation and function following cognate stimulations. These observations suggest that sIL-7Rα levels may influence MAIT cell numbers and function in vivo by limiting IL-7 bioavailability to MAIT cells. Collectively, these observations suggest that IL-7RA polymorphisms may play a significant role in MAIT cell biology and influence MAIT cells recovery in HIV-1 infection. The potential links between IL7RA polymorphisms, MAIT cell immunobiology, and HIV-1 infection warrant further studies going forward.
Collapse
Affiliation(s)
- Fei Han
- 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
| | - Muhammad Yaaseen Gulam
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Yichao Zheng
- 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
| | - Nurul Syuhada Zulhaimi
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- Department of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Wan Rong Sia
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Dan He
- 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
| | - Amanda Ho
- 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
| | - Leila Hadadi
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Zhenyu Liu
- 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
| | - Peiwu Qin
- 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
| | - Peter E. Lobie
- 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
| | - Adeeba Kamarulzaman
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- Department of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Johan K. Sandberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sharon R. Lewin
- Peter Doherty Institute for Infection and Immunity, Melbourne University, Victoria, Australia
| | - Reena Rajasuriar
- Centre of Excellence for Research in AIDS (CERiA), University of Malaya, Kuala Lumpur, Malaysia
- Department of Medicine, University of Malaya, Kuala Lumpur, Malaysia
- Peter Doherty Institute for Infection and Immunity, Melbourne University, Victoria, Australia
| | - 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
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- *Correspondence: Edwin Leeansyah,
| |
Collapse
|
42
|
Moreira MDL, Borges-Fernandes LO, Pascoal-Xavier MA, Ribeiro ÁL, Pereira VHS, Pediongco T, Araújo MSDS, Teixeira-Carvalho A, de Carvalho AL, Mourão MVA, Campos FA, Borges M, Carneiro M, Chen Z, Saunders E, McConville M, Tsuji M, McCluskey J, Martins-Filho OA, Eckle SBG, Coelho-dos-Reis JGA, Peruhype-Magalhães V. The role of mucosal-associated invariant T cells in visceral leishmaniasis. Front Immunol 2022; 13:926446. [PMID: 36189274 PMCID: PMC9521739 DOI: 10.3389/fimmu.2022.926446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are restricted by MR1 and are known to protect against bacterial and viral infections. Our understanding of the role of MAIT cells in parasitic infections, such as visceral leishmaniasis (VL) caused by protozoan parasites of Leishmania donovani, is limited. This study showed that in response to L. infantum, human peripheral blood MAIT cells from children with leishmaniasis produced TNF and IFN-γ in an MR1-dependent manner. The overall frequency of MAIT cells was inversely correlated with alanine aminotransferase levels, a specific marker of liver damage strongly associated with severe hepatic involvement in VL. In addition, there was a positive correlation between total protein levels and the frequency of IL-17A+ CD8+ MAIT cells, whereby reduced total protein levels are a marker of liver and kidney damage. Furthermore, the frequencies of IFN-γ+ and IL-10+ MAIT cells were inversely correlated with hemoglobin levels, a marker of severe anemia. In asymptomatic individuals and VL patients after treatment, MAIT cells also produced IL-17A, a cytokine signature associated with resistance to visceral leishmaniasis, suggesting that MAIT cells play important role in protecting against VL. In summary, these results broaden our understanding of MAIT-cell immunity to include protection against parasitic infections, with implications for MAIT-cell-based therapeutics and vaccines. At last, this study paves the way for the investigation of putative MAIT cell antigens that could exist in the context of Leishmania infection.
Collapse
Affiliation(s)
- Marcela de Lima Moreira
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | | | - Marcelo Antônio Pascoal-Xavier
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
- School of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ágata Lopes Ribeiro
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
| | | | - Troi Pediongco
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | | | - Andréa Teixeira-Carvalho
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
| | - Andrea Lucchesi de Carvalho
- João Paulo II Children’s Hospital, Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Flávia Alves Campos
- João Paulo II Children’s Hospital, Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Marineide Borges
- João Paulo II Children’s Hospital, Fundação Hospitalar do Estado de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Mariângela Carneiro
- Parasitology Department, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Zhenjun Chen
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Eleanor Saunders
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Malcolm McConville
- Department of Biochemistry and Molecular Biology, The University of Melbourne, Melbourne, VIC, Australia
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Department of Medicine, Columbia University Irving Medical Center, New York, NY, United States
| | - James McCluskey
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | | | - Sidonia Barbara Guiomar Eckle
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jordana Grazziela Alves Coelho-dos-Reis
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
- Department of Microbiology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
- *Correspondence: Vanessa Peruhype-Magalhães, ; ; Jordana Grazziela Alves Coelho-dos-Reis, ;
| | - Vanessa Peruhype-Magalhães
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
- *Correspondence: Vanessa Peruhype-Magalhães, ; ; Jordana Grazziela Alves Coelho-dos-Reis, ;
| |
Collapse
|
43
|
New Insights into the Pathogenesis of Giant Cell Arteritis: Mechanisms Involved in Maintaining Vascular Inflammation. J Clin Med 2022; 11:jcm11102905. [PMID: 35629030 PMCID: PMC9143803 DOI: 10.3390/jcm11102905] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/16/2022] [Accepted: 05/19/2022] [Indexed: 02/04/2023] Open
Abstract
The giant cell arteritis (GCA) pathophysiology is complex and multifactorial, involving a predisposing genetic background, the role of immune aging and the activation of vascular dendritic cells by an unknown trigger. Once activated, dendritic cells recruit CD4 T cells and induce their activation, proliferation and polarization into Th1 and Th17, which produce interferon-gamma (IFN-γ) and interleukin-17 (IL-17), respectively. IFN-γ triggers the production of chemokines by vascular smooth muscle cells, which leads to the recruitment of additional CD4 and CD8 T cells and also monocytes that differentiate into macrophages. Recent data have shown that IL-17, IFN-γ and GM-CSF induce the differentiation of macrophage subpopulations, which play a role in the destruction of the arterial wall, in neoangiogenesis or intimal hyperplasia. Under the influence of different mediators, mainly endothelin-1 and PDGF, vascular smooth muscle cells migrate to the intima, proliferate and change their phenotype to become myofibroblasts that further proliferate and produce extracellular matrix proteins, increasing the vascular stenosis. In addition, several defects in the immune regulatory mechanisms probably contribute to chronic vascular inflammation in GCA: a defect in the PD-1/PD-L1 pathway, a quantitative and qualitative Treg deficiency, the implication of resident cells, the role of GM-CSF and IL-6, the implication of the NOTCH pathway and the role of mucosal‑associated invariant T cells and tissue‑resident memory T cells.
Collapse
|
44
|
Immunoprofiling reveals cell subsets associated with the trajectory of cytomegalovirus reactivation post stem cell transplantation. Nat Commun 2022; 13:2603. [PMID: 35546552 PMCID: PMC9095831 DOI: 10.1038/s41467-022-29943-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Accepted: 03/15/2022] [Indexed: 11/11/2022] Open
Abstract
Human cytomegalovirus reactivation is a major opportunistic infection after allogeneic haematopoietic stem cell transplantation and has a complex relationship with post-transplant immune reconstitution. Here, we use mass cytometry to define patterns of innate and adaptive immune cell reconstitution at key phases of human cytomegalovirus reactivation in the first 100 days post haematopoietic stem cell transplantation. Human cytomegalovirus reactivation is associated with the development of activated, memory T-cell profiles, with faster effector-memory CD4+ T-cell recovery in patients with low-level versus high-level human cytomegalovirus DNAemia. Mucosal-associated invariant T cell levels at the initial detection of human cytomegalovirus DNAemia are significantly lower in patients who subsequently develop high-level versus low-level human cytomegalovirus reactivation. Our data describe distinct immune signatures that emerged with human cytomegalovirus reactivation after haematopoietic stem cell transplantation, and highlight Mucosal-associated invariant T cell levels at the first detection of reactivation as a marker that may be useful to anticipate the magnitude of human cytomegalovirus DNAemia. Human cytomegalovirus is a major cause of morbidity and mortality in transplant patients and multiple immune cells types are critical during infection and reactivation. Here the authors assess the immune cell compartments of haematopoietic stem cell recipients in the early period post transplantation and identify key features of effector memory CD4+ T cells and mucosal associated invariant T cells in this context.
Collapse
|
45
|
Abstract
COVID-19 is a respiratory disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). It was first documented in late 2019, but within months, a worldwide pandemic was declared due to the easily transmissible nature of the virus. Research to date on the immune response to SARS-CoV-2 has focused largely on conventional B and T lymphocytes. This review examines the emerging role of unconventional T cell subsets, including γδ T cells, invariant natural killer T (iNKT) cells and mucosal associated invariant T (MAIT) cells in human SARS-CoV-2 infection.Some of these T cell subsets have been shown to play protective roles in anti-viral immunity by suppressing viral replication and opsonising virions of SARS-CoV. Here, we explore whether unconventional T cells play a protective role in SARS-CoV-2 infection as well. Unconventional T cells are already under investigation as cell-based immunotherapies for cancer. We discuss the potential use of these cells as therapeutic agents in the COVID-19 setting. Due to the rapidly evolving situation presented by COVID-19, there is an urgent need to understand the pathogenesis of this disease and the mechanisms underlying its immune response. Through this, we may be able to better help those with severe cases and lower the mortality rate by devising more effective vaccines and novel treatment strategies.
Collapse
Affiliation(s)
- Kristen Orumaa
- Department of Clinical Microbiology and Department of Immunology, Trinity Translational Medicine Institute, St James's Hospital, Dublin 8, Ireland
| | - Margaret R Dunne
- Department of Clinical Microbiology and Department of Immunology, Trinity Translational Medicine Institute, St James's Hospital, Dublin 8, Ireland.
| |
Collapse
|
46
|
Palmieri F, Koutsokera A, Bernasconi E, Junier P, von Garnier C, Ubags N. Recent Advances in Fungal Infections: From Lung Ecology to Therapeutic Strategies With a Focus on Aspergillus spp. Front Med (Lausanne) 2022; 9:832510. [PMID: 35386908 PMCID: PMC8977413 DOI: 10.3389/fmed.2022.832510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/22/2022] [Indexed: 12/15/2022] Open
Abstract
Fungal infections are estimated to be the main cause of death for more than 1.5 million people worldwide annually. However, fungal pathogenicity has been largely neglected. This is notably the case for pulmonary fungal infections, which are difficult to diagnose and to treat. We are currently facing a global emergence of antifungal resistance, which decreases the chances of survival for affected patients. New therapeutic approaches are therefore needed to face these life-threatening fungal infections. In this review, we will provide a general overview on respiratory fungal infections, with a focus on fungi of the genus Aspergillus. Next, the immunological and microbiological mechanisms of fungal pathogenesis will be discussed. The role of the respiratory mycobiota and its interactions with the bacterial microbiota on lung fungal infections will be presented from an ecological perspective. Finally, we will focus on existing and future innovative approaches for the treatment of respiratory fungal infections.
Collapse
Affiliation(s)
- Fabio Palmieri
- Laboratory of Microbiology, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
- *Correspondence: Fabio Palmieri,
| | - Angela Koutsokera
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Eric Bernasconi
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Pilar Junier
- Laboratory of Microbiology, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | - Christophe von Garnier
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
| | - Niki Ubags
- Faculty of Biology and Medicine, University of Lausanne, Service de Pneumologie, Centre Hospitalier Universitaire Vaudois (CHUV), Lausanne, Switzerland
- Niki Ubags,
| |
Collapse
|
47
|
Su B, Kong D, Yang X, Zhang T, Kuang YQ. Mucosal-associated invariant T cells: a cryptic coordinator in HIV-infected immune reconstitution. J Med Virol 2022; 94:3043-3053. [PMID: 35243649 DOI: 10.1002/jmv.27696] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/20/2022] [Accepted: 03/01/2022] [Indexed: 11/11/2022]
Abstract
Human immunodeficiency virus type 1 (HIV-1) infection causes considerable morbidity and mortality worldwide. Although antiretroviral therapy (ART) has largely transformed HIV infection from a fatal disease to a chronic condition, approximately 10%~40% of HIV-infected individuals who receive effective ART and sustain long-term viral suppression still cannot achieve optimal immune reconstitution. These patients are called immunological non-responders, a state associated with poor clinical prognosis. Mucosal-associated invariant T (MAIT) cells are an evolutionarily conserved unconventional T cell subset defined by expression of semi-invariant αβ T cell receptor (TCR), which recognizes metabolites derived from the riboflavin biosynthetic pathway presented on major histocompatibility complex (MHC)-related protein-1 (MR1). MAIT cells, which are considered to act as a bridge between innate and adaptive immunity, produce a wide range of cytokines and cytotoxic molecules upon activation through TCR-dependent and TCR-independent mechanisms, which is of major importance in defense against a variety of pathogens. In addition, MAIT cells are involved in autoimmune and immune-mediated diseases. The number of MAIT cells is dramatically and irreversibly decreased in the early stage of HIV infection and is not fully restored even after long-term suppressive ART. In light of the important role of MAIT cells in mucosal immunity and because microbial translocation is inversely associated with CD4+ T cell counts, we propose that MAIT cells participate in the maintenance of intestinal barrier integrity and microbial homeostasis, thus further affecting immune reconstitution in HIV-infected individuals. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.,Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Deshenyue Kong
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, China.,Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| | - Xiaodong Yang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.,Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China.,Sino-French Joint Laboratory for Research on Humoral Immune Response to HIV Infection, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, 100069, China
| | - Yi-Qun Kuang
- NHC Key Laboratory of Drug Addiction Medicine, First Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, 650032, China.,Scientific Research Laboratory Center, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| |
Collapse
|
48
|
Ishikawa Y, Yamada M, Wada N, Takahashi E, Imadome KI. Mucosal-associated invariant T cells are activated in an interleukin-18-dependent manner in Epstein-Barr virus-associated T/natural killer cell lymphoproliferative diseases. Clin Exp Immunol 2022; 207:141-148. [PMID: 35380609 PMCID: PMC8982962 DOI: 10.1093/cei/uxab004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 09/30/2021] [Accepted: 10/15/2021] [Indexed: 02/03/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are a type of innate immune cells that protect against some infections. However, the involvement of MAIT cells in Epstein-Barr virus-associated T/natural killer cell lymphoproliferative diseases (EBV-T/NK-LPD) is unclear. In this study, we found that MAIT cells were highly activated in the blood of patients with EBV-T/NK-LPD. MAIT cell activation levels correlated with disease severity and plasma IL-18 levels. Stimulation of healthy peripheral blood mononuclear cells with EBV resulted in activation of MAIT cells, and this activation level was enhanced by exogenous IL-18. MAIT cells stimulated by IL-18 might thus be involved in the immunopathogenesis of EBV-T/NK-LPD.
Collapse
Affiliation(s)
- Yuriko Ishikawa
- Department of Advanced Medicine for Viral Infections, National Center for Child Health and Development (NCCHD), Tokyo, Japan
- Correspondence: Yuriko Ishikawa, Department of Advanced Medicine for Infections, National Center for Child Health and Development (NCCHD), Tokyo, 157–8535, Japan.
| | - Masaki Yamada
- Department of Advanced Medicine for Viral Infections, National Center for Child Health and Development (NCCHD), Tokyo, Japan
| | - Naomi Wada
- Department of Advanced Medicine for Viral Infections, National Center for Child Health and Development (NCCHD), Tokyo, Japan
| | - Etsuko Takahashi
- Department of Advanced Medicine for Viral Infections, National Center for Child Health and Development (NCCHD), Tokyo, Japan
| | - Ken-Ichi Imadome
- Department of Advanced Medicine for Viral Infections, National Center for Child Health and Development (NCCHD), Tokyo, Japan
| |
Collapse
|
49
|
Exploring the Role of Innate Lymphocytes in the Immune System of Bats and Virus-Host Interactions. Viruses 2022; 14:v14010150. [PMID: 35062356 PMCID: PMC8781337 DOI: 10.3390/v14010150] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/10/2022] [Accepted: 01/12/2022] [Indexed: 01/27/2023] Open
Abstract
Bats are reservoirs of a large number of viruses of global public health significance, including the ancestral virus for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and the causative agent of coronavirus disease 2019 (COVID-19). Although bats are natural carriers of multiple pathogenic viruses, they rarely display signs of disease. Recent insights suggest that bats have a more balanced host defense and tolerance system to viral infections that may be linked to the evolutionary adaptation to powered flight. Therefore, a deeper understanding of bat immune system may provide intervention strategies to prevent zoonotic disease transmission and to identify new therapeutic targets. Similar to other eutherian mammals, bats have both innate and adaptive immune systems that have evolved to detect and respond to invading pathogens. Bridging these two systems are innate lymphocytes, which are highly abundant within circulation and barrier tissues. These cells share the characteristics of both innate and adaptive immune cells and are poised to mount rapid effector responses. They are ideally suited as the first line of defense against early stages of viral infections. Here, we will focus on the current knowledge of innate lymphocytes in bats, their function, and their potential role in host–pathogen interactions. Moreover, given that studies into bat immune systems are often hindered by a lack of bat-specific research tools, we will discuss strategies that may aid future research in bat immunity, including the potential use of organoid models to delineate the interplay between innate lymphocytes, bat viruses, and host tolerance.
Collapse
|
50
|
Significance of bystander T cell activation in microbial infection. Nat Immunol 2022; 23:13-22. [PMID: 34354279 DOI: 10.1038/s41590-021-00985-3] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023]
Abstract
During microbial infection, pre-existing memory CD8+ T cells that are not specific for the infecting pathogens can be activated by cytokines without cognate antigens, termed bystander activation. Studies in mouse models and human patients demonstrate bystander activation of memory CD8+ T cells, which exerts either protective or detrimental effects on the host, depending on the infection model or disease. Research has elucidated mechanisms underlying the bystander activation of CD8+ T cells in terms of the responsible cytokines and the effector mechanisms of bystander-activated CD8+ T cells. In this Review, we describe the history of research on bystander CD8+ T cell activation as well as evidence of bystander activation. We also discuss the mechanisms and immunopathological roles of bystander activation in various microbial infections.
Collapse
|