1
|
Germain L, Veloso P, Lantz O, Legoux F. MAIT cells: Conserved watchers on the wall. J Exp Med 2025; 222:e20232298. [PMID: 39446132 PMCID: PMC11514058 DOI: 10.1084/jem.20232298] [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: 07/26/2024] [Revised: 09/17/2024] [Accepted: 10/03/2024] [Indexed: 10/25/2024] Open
Abstract
MAIT cells are innate-like T cells residing in barrier tissues such as the lung, skin, and intestine. Both the semi-invariant T cell receptor of MAIT cells and the restricting element MR1 are deeply conserved across mammals, indicating non-redundant functions linked to antigenic specificity. MAIT cells across species concomitantly express cytotoxicity and tissue-repair genes, suggesting versatile functions. Accordingly, MAIT cells contribute to antibacterial responses as well as to the repair of damaged barrier tissues. MAIT cells recognize riboflavin biosynthetic pathway-derived metabolites, which rapidly cross epithelial barriers to be presented by antigen-presenting cells. Changes in gut ecology during intestinal inflammation drive the expansion of strong riboflavin and MAIT ligand producers. Thus, MAIT cells may enable real-time surveillance of microbiota dysbiosis across intact epithelia and provide rapid and context-dependent responses. Here, we discuss recent findings regarding the origin and regulation of MAIT ligands and the role of MAIT cells in barrier tissues. We speculate on the potential reasons for MAIT cell conservation during evolution.
Collapse
Affiliation(s)
- Lilou Germain
- INSERM ERL1305, CNRS UMR6290, Institut de Génétique and Développement de Rennes, Université de Rennes, Rennes, France
| | - Pablo Veloso
- INSERM ERL1305, CNRS UMR6290, Institut de Génétique and Développement de Rennes, Université de Rennes, Rennes, France
| | - Olivier Lantz
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
- Laboratoire d’immunologie Clinique, Institut Curie, Paris, France
- Centre d’investigation Clinique en Biothérapie Gustave-Roussy Institut Curie (CIC-BT1428), Paris, France
| | - François Legoux
- INSERM ERL1305, CNRS UMR6290, Institut de Génétique and Développement de Rennes, Université de Rennes, Rennes, France
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| |
Collapse
|
2
|
Wang H, Souter MNT, de Lima Moreira M, Li S, Zhou Y, Nelson AG, Yu J, Meehan LJ, Meehan BS, Eckle SBG, Lee HJ, Schröder J, Haque A, Mak JYW, Fairlie DP, McCluskey J, Wang Z, Chen Z, Corbett AJ. MAIT cell plasticity enables functional adaptation that drives antibacterial immune protection. Sci Immunol 2024; 9:eadp9841. [PMID: 39642244 DOI: 10.1126/sciimmunol.adp9841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 11/08/2024] [Indexed: 12/08/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells are known for their rapid effector functions and antibacterial immune protection. Here, we define the plasticity of interferon-γ (IFN-γ)-producing MAIT1 and interleukin-17A (IL-17A)-producing MAIT17 cell subsets in vivo. Whereas T-bet+ MAIT1 cells remained stable in all experimental settings, after adoptive transfer or acute Legionella or Francisella infection, RORγt+ MAIT17 cells could undergo phenotypic and functional conversion into both RORγt+T-bet+ MAIT1/17 and RORγt-T-bet+ MAIT1 cells. This plasticity ensured that MAIT17 cells played a dominant role in generating antibacterial MAIT1 responses in mucosal tissues. Single-cell transcriptomics revealed that MAIT17-derived MAIT1 cells were distinct from canonical MAIT1 cells yet could migrate out of mucosal tissues to contribute to the global MAIT1 pool in subsequent systemic infections. Human IL-17A-secreting MAIT cells also showed similar functional plasticity. Our findings have broad implications for understanding the role of MAIT cells in combatting infections and their potential utility in MAIT cell-targeted vaccines.
Collapse
Affiliation(s)
- Huimeng Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Michael N T Souter
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Marcela de Lima Moreira
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Shihan Li
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Computational Sciences Initiative, Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Yuchen Zhou
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Tsinghua Medicine, School of Medicine, Tsinghua University, Beijing, China
| | - Adam G Nelson
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jinhan Yu
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Department of Clinical Laboratory, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lucy J Meehan
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Bronwyn S Meehan
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Sidonia B G Eckle
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Hyun Jae Lee
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jan Schröder
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
- Computational Sciences Initiative, Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Ashraful Haque
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Jeffrey Y W Mak
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - David P Fairlie
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - James McCluskey
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Zhongfang Wang
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Zhenjun Chen
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Alexandra J Corbett
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| |
Collapse
|
3
|
Gleeson PJ, Camara NOS, Launay P, Lehuen A, Monteiro RC. Immunoglobulin A Antibodies: From Protection to Harmful Roles. Immunol Rev 2024; 328:171-191. [PMID: 39578936 DOI: 10.1111/imr.13424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/15/2024] [Accepted: 11/07/2024] [Indexed: 11/24/2024]
Abstract
Immunoglobulin A (IgA) is the most abundantly produced antibody in humans. IgA is a unique class of immunoglobulin due to its multiple molecular forms, and a defining difference between the two subclasses: IgA1 has a long hinge-region that is heavily O-glycosylated, whereas the IgA2 hinge-region is shorter but resistant to bacterial proteases prevalent at mucosal sites. IgA is essential for immune homeostasis and education. Mucosal IgA plays a crucial role in maintaining the integrity of the mucosal barrier by immune exclusion of pathobionts while facilitating colonization with certain commensals; a large part of the gut microbiota is coated with IgA. In the circulation, monomeric IgA that has not been engaged by antigen plays a discrete role in dampening inflammatory responses. Protective and harmful roles of IgA have been studied over several decades, but a new understanding of the complex role of this immunoglobulin in health and disease has been provided by recent studies. Here, we discuss the physiological and pathological roles of IgA with a special focus on the gut, kidneys, and autoimmunity. We also discuss new IgA-based therapeutic approaches.
Collapse
Affiliation(s)
- Patrick J Gleeson
- Center for Research on Inflammation, Paris Cité University, Paris, France
- INSERM, Paris, France
- CNRS, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
- Nephrology Department, Bichat Hospital, Paris, France
| | - Niels O S Camara
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Pierre Launay
- Center for Research on Inflammation, Paris Cité University, Paris, France
- INSERM, Paris, France
- CNRS, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| | - Agnès Lehuen
- Inflamex Laboratory of Excellence, Paris, France
- Cochin Institute, INSERM, CNRS, Paris Cité University, Paris, France
| | - Renato C Monteiro
- Center for Research on Inflammation, Paris Cité University, Paris, France
- INSERM, Paris, France
- CNRS, Paris, France
- Inflamex Laboratory of Excellence, Paris, France
| |
Collapse
|
4
|
Walkenhorst M, Sonner JK, Meurs N, Engler JB, Bauer S, Winschel I, Woo MS, Raich L, Winkler I, Vieira V, Unger L, Salinas G, Lantz O, Friese MA, Willing A. Protective effect of TCR-mediated MAIT cell activation during experimental autoimmune encephalomyelitis. Nat Commun 2024; 15:9287. [PMID: 39468055 PMCID: PMC11519641 DOI: 10.1038/s41467-024-53657-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 10/18/2024] [Indexed: 10/30/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells express semi-invariant T cell receptors (TCR) for recognizing bacterial and yeast antigens derived from riboflavin metabolites presented on the non-polymorphic MHC class I-related protein 1 (MR1). Neuroinflammation in multiple sclerosis (MS) is likely initiated by autoreactive T cells and perpetuated by infiltration of additional immune cells, but the precise role of MAIT cells in MS pathogenesis remains unknown. Here, we use experimental autoimmune encephalomyelitis (EAE), a mouse model of MS, and find an accumulation of MAIT cells in the inflamed central nervous system (CNS) enriched for MAIT17 (RORγt+) and MAIT1/17 (T-bet+RORγt+) subsets with inflammatory and protective features. Results from transcriptome profiling and Nur77GFP reporter mice show that these CNS MAIT cells are activated via cytokines and TCR. Blocking TCR activation with an anti-MR1 antibody exacerbates EAE, whereas enhancing TCR activation with the cognate antigen, 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil, ameliorates EAE severity, potentially via the induction of amphiregulin (AREG). In summary, our findings suggest that TCR-mediated MAIT cell activation is protective in CNS inflammation, likely involving an induction of AREG.
Collapse
Affiliation(s)
- Mark Walkenhorst
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jana K Sonner
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nina Meurs
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Broder Engler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Simone Bauer
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ingo Winschel
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Marcel S Woo
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lukas Raich
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Iris Winkler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Vanessa Vieira
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lisa Unger
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gabriela Salinas
- NGS-Integrative Genomics Core Unit, Institute of Pathology, University Medical Center Göttingen, Göttingen, Germany
| | - Olivier Lantz
- Institut National de la Santé et de la Recherche Médicale U932, PSL University, Institut Curie, Paris, France
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| | - Anne Willing
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
5
|
Samer C, McWilliam HEG, McSharry BP, Burchfield JG, Stanton RJ, Rossjohn J, Villadangos JA, Abendroth A, Slobedman B. Impaired endocytosis and accumulation in early endosomal compartments defines herpes simplex virus-mediated disruption of the nonclassical MHC class I-related molecule MR1. J Biol Chem 2024; 300:107748. [PMID: 39260697 PMCID: PMC11736056 DOI: 10.1016/j.jbc.2024.107748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 08/13/2024] [Accepted: 08/21/2024] [Indexed: 09/13/2024] Open
Abstract
Presentation of metabolites by the major histocompatibility complex class I-related protein 1 (MR1) molecule to mucosal-associated invariant T cells is impaired during herpes simplex virus type 1 (HSV-1) and type 2 (HSV-2) infections. This is surprising given these viruses do not directly synthesise MR1 ligands. We have previously identified several HSV proteins responsible for rapidly downregulating the intracellular pool of immature MR1, effectively inhibiting new surface antigen presentation, while preexisting ligand-bound mature MR1 is unexpectedly upregulated by HSV-1. Using flow cytometry, immunoblotting, and high-throughput fluorescence microscopy, we demonstrate that the endocytosis of surface MR1 is impaired during HSV infection and that internalized molecules accumulate in EEA1-labeled early endosomes, avoiding degradation. We establish that the short MR1 cytoplasmic tail is not required for HSV-1-mediated downregulation of immature molecules; however it may play a role in the retention of mature molecules on the surface and in early endosomes. We also determine that the HSV-1 US3 protein, the shorter US3.5 kinase and the full-length HSV-2 homolog, all predominantly target mature surface rather than total MR1 levels. We propose that the downregulation of intracellular and cell surface MR1 molecules by US3 and other HSV proteins is an immune-evasive countermeasure to minimize the effect of impaired MR1 endocytosis, which might otherwise render infected cells susceptible to MR1-mediated killing by mucosal-associated invariant T cells.
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, New South Wales, Australia
| | - Hamish E G McWilliam
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, 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, New South Wales, Australia; School of Dentistry and Medical Sciences, Faculty of Science and Health, and Gulbali Institute, Charles Sturt University, Wagga Wagga, New South Wales, Australia
| | - James G Burchfield
- Charles Perkins Centre, The University of Sydney, Camperdown, New South Wales, Australia; School of Life and Environmental Sciences, The University of Sydney, Camperdown, New South Wales, Australia
| | - Richard J Stanton
- Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Jamie Rossjohn
- Division of Infection & Immunity, School of Medicine, Cardiff University, Cardiff, Wales, UK; Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jose A Villadangos
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia; Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 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, New South Wales, 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, New South Wales, Australia.
| |
Collapse
|
6
|
López-Rodríguez JC, Barral P. Mucosal associated invariant T cells: Powerhouses of the lung. Immunol Lett 2024; 269:106910. [PMID: 39128630 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
|
7
|
Zhao C, Zhao R, Wu X, Tang K, Xu P, Chen X, Zhu P, He Y. Function of unconventional T cells in oral lichen planus revealed by single-cell RNA sequencing. Inflamm Res 2024; 73:1477-1492. [PMID: 39073597 DOI: 10.1007/s00011-024-01912-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/30/2024] Open
Abstract
OBJECTIVE We intended to map the single-cell profile of OLP, explore the molecular characteristics of unconventional T cells in OLP tissues. METHODS Buccal mucosa samples from OLP patients and healthy individuals were used to prepare single-cell suspension. Single-cell RNA sequencing was used to analyze the proportion of all the cells, and the molecular characteristics of unconventional T cells. Immunohistochemical staining was used to detect the expression of unconventional T cells marker genes. RESULTS The cell clusters from buccal mucosa were categorized into immune cells, fibroblasts, endothelial cells, and epithelial cells. Unconventional T cells with phenotype of CD247+TRDC+NCAM1+ were identified. Immunohistochemical staining revealed higher expression of unconventional T cell marker genes in OLP tissue, predominantly in the lamina propria. In OLP, unconventional T cells are in a unique stress response state, exhibited enhanced NF-κB signaling and apoptosis inhibition, enhanced heat shock protein genes expression, weakened cytotoxic function. A large number of ligand-receptor pairs were found between unconventional T cells and other cells, particularly with fibroblasts and endothelial cells. CONCLUSIONS This study mapped the single-cell profile of OLP, delineated the molecular characteristics of unconventional T cells in OLP, and uncovered that these unconventional T cells are in a stress response state.
Collapse
Affiliation(s)
- Chen Zhao
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Oral Medicine, School of Stomatology, Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Ruowen Zhao
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Oral Medicine, School of Stomatology, Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Xinwen Wu
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Kailin Tang
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Pan Xu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Oral Medicine, School of Stomatology, Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Xin Chen
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Oral Medicine, School of Stomatology, Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Pingyi Zhu
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Oral Medicine, School of Stomatology, Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China
| | - Yuan He
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration & Tongji Research Institute of Stomatology & Department of Oral Medicine, School of Stomatology, Stomatological Hospital and Dental School, Tongji University, Shanghai, 200072, China.
| |
Collapse
|
8
|
Purohit SK, Stern L, Corbett AJ, Mak JYW, Fairlie DP, Slobedman B, Abendroth A. Varicella Zoster Virus disrupts MAIT cell polyfunctional effector responses. PLoS Pathog 2024; 20:e1012372. [PMID: 39110717 PMCID: PMC11305569 DOI: 10.1371/journal.ppat.1012372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 06/25/2024] [Indexed: 08/10/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are unconventional T cells that respond to riboflavin biosynthesis and cytokines through TCR-dependent and -independent pathways, respectively. MAIT cell activation plays an immunoprotective role against several pathogens, however the functional capacity of MAIT cells following direct infection or exposure to infectious agents remains poorly defined. We investigated the impact of Varicella Zoster Virus (VZV) on blood-derived MAIT cells and report virus-mediated impairment of activation, cytokine production, and altered transcription factor expression by VZV infected (antigen+) and VZV exposed (antigen-) MAIT cells in response to TCR-dependent and -independent stimulation. Furthermore, we reveal that suppression of VZV exposed (antigen-) MAIT cells is not mediated by a soluble factor from neighbouring VZV infected (antigen+) MAIT cells. Finally, we demonstrate that VZV impairs the cytolytic potential of MAIT cells in response to riboflavin synthesising bacteria. In summary, we report a virus-mediated immune-evasion strategy that disarms MAIT cell responses.
Collapse
Affiliation(s)
- Shivam. K. Purohit
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Lauren Stern
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Alexandra J. Corbett
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 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
| | - Barry Slobedman
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| | - Allison Abendroth
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
9
|
Edmans MD, Connelley TK, Morgan S, Pediongco TJ, Jayaraman S, Juno JA, Meehan BS, Dewar PM, Maze EA, Roos EO, Paudyal B, Mak JYW, Liu L, Fairlie DP, Wang H, Corbett AJ, McCluskey J, Benedictus L, Tchilian E, Klenerman P, Eckle SBG. MAIT cell-MR1 reactivity is highly conserved across multiple divergent species. J Biol Chem 2024; 300:107338. [PMID: 38705391 PMCID: PMC11190491 DOI: 10.1016/j.jbc.2024.107338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 04/03/2024] [Accepted: 04/24/2024] [Indexed: 05/07/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are a subset of unconventional T cells that recognize small molecule metabolites presented by major histocompatibility complex class I related protein 1 (MR1), via an αβ T cell receptor (TCR). MAIT TCRs feature an essentially invariant TCR α-chain, which is highly conserved between mammals. Similarly, MR1 is the most highly conserved major histocompatibility complex-I-like molecule. This extreme conservation, including the mode of interaction between the MAIT TCR and MR1, has been shown to allow for species-mismatched reactivities unique in T cell biology, thereby allowing the use of selected species-mismatched MR1-antigen (MR1-Ag) tetramers in comparative immunology studies. However, the pattern of cross-reactivity of species-mismatched MR1-Ag tetramers in identifying MAIT cells in diverse species has not been formally assessed. We developed novel cattle and pig MR1-Ag tetramers and utilized these alongside previously developed human, mouse, and pig-tailed macaque MR1-Ag tetramers to characterize cross-species tetramer reactivities. MR1-Ag tetramers from each species identified T cell populations in distantly related species with specificity that was comparable to species-matched MR1-Ag tetramers. However, there were subtle differences in staining characteristics with practical implications for the accurate identification of MAIT cells. Pig MR1 is sufficiently conserved across species that pig MR1-Ag tetramers identified MAIT cells from the other species. However, MAIT cells in pigs were at the limits of phenotypic detection. In the absence of sheep MR1-Ag tetramers, a MAIT cell population in sheep blood was identified phenotypically, utilizing species-mismatched MR1-Ag tetramers. Collectively, our results validate the use and define the limitations of species-mismatched MR1-Ag tetramers in comparative immunology studies.
Collapse
Affiliation(s)
- Matthew D Edmans
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom; Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom.
| | - Timothy K Connelley
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Roslin, United Kingdom
| | - Sophie Morgan
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Troi J Pediongco
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Siddharth Jayaraman
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Roslin, United Kingdom
| | - Jennifer A Juno
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Bronwyn S Meehan
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Phoebe M Dewar
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Emmanuel A Maze
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Eduard O Roos
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Basudev Paudyal
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Jeffrey Y W Mak
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Ligong Liu
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - David P Fairlie
- Centre for Chemistry and Drug Discovery, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia; Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Huimeng Wang
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia; State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Alexandra J Corbett
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Lindert Benedictus
- Division of Infection and Immunity, The Roslin Institute, The University of Edinburgh, Roslin, United Kingdom; Faculty of Veterinary Medicine, Department of Population Health Sciences, Utrecht University, Utrecht, The Netherlands
| | - Elma Tchilian
- Department of Enhanced Host Responses, The Pirbright Institute, Pirbright, United Kingdom
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Sidonia B G Eckle
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
10
|
Burton RJ, Raffray L, Moet LM, Cuff SM, White DA, Baker SE, Moser B, O’Donnell VB, Ghazal P, Morgan MP, Artemiou A, Eberl M. Conventional and unconventional T-cell responses contribute to the prediction of clinical outcome and causative bacterial pathogen in sepsis patients. Clin Exp Immunol 2024; 216:293-306. [PMID: 38430552 PMCID: PMC11097916 DOI: 10.1093/cei/uxae019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 02/12/2024] [Accepted: 02/28/2024] [Indexed: 03/04/2024] Open
Abstract
Sepsis is characterized by a dysfunctional host response to infection culminating in life-threatening organ failure that requires complex patient management and rapid intervention. Timely diagnosis of the underlying cause of sepsis is crucial, and identifying those at risk of complications and death is imperative for triaging treatment and resource allocation. Here, we explored the potential of explainable machine learning models to predict mortality and causative pathogen in sepsis patients. By using a modelling pipeline employing multiple feature selection algorithms, we demonstrate the feasibility of identifying integrative patterns from clinical parameters, plasma biomarkers, and extensive phenotyping of blood immune cells. While no single variable had sufficient predictive power, models that combined five and more features showed a macro area under the curve (AUC) of 0.85 to predict 90-day mortality after sepsis diagnosis, and a macro AUC of 0.86 to discriminate between Gram-positive and Gram-negative bacterial infections. Parameters associated with the cellular immune response contributed the most to models predictive of 90-day mortality, most notably, the proportion of T cells among PBMCs, together with expression of CXCR3 by CD4+ T cells and CD25 by mucosal-associated invariant T (MAIT) cells. Frequencies of Vδ2+ γδ T cells had the most profound impact on the prediction of Gram-negative infections, alongside other T-cell-related variables and total neutrophil count. Overall, our findings highlight the added value of measuring the proportion and activation patterns of conventional and unconventional T cells in the blood of sepsis patients in combination with other immunological, biochemical, and clinical parameters.
Collapse
Affiliation(s)
- Ross J Burton
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Adult Critical Care, University Hospital of Wales, Cardiff and Vale University Health Board, Cardiff, UK
| | - Loïc Raffray
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Department of Internal Medicine, Félix Guyon University Hospital of La Réunion, Saint Denis, Réunion Island, France
| | - Linda M Moet
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Simone M Cuff
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Daniel A White
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Sarah E Baker
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
| | - Bernhard Moser
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Valerie B O’Donnell
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Peter Ghazal
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| | - Matt P Morgan
- Adult Critical Care, University Hospital of Wales, Cardiff and Vale University Health Board, Cardiff, UK
| | - Andreas Artemiou
- School of Mathematics, Cardiff University, Cardiff, UK
- Department of Information Technologies, University of Limassol, 3025 Limassol, Cyprus
| | - Matthias Eberl
- Division of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK
- Systems Immunity Research Institute, Cardiff University, Cardiff, UK
| |
Collapse
|
11
|
Ciacchi L, Mak JYW, Le JP, Fairlie DP, McCluskey J, Corbett AJ, Rossjohn J, Awad W. Mouse mucosal-associated invariant T cell receptor recognition of MR1 presenting the vitamin B metabolite, 5-(2-oxopropylideneamino)-6-d-ribitylaminouracil. J Biol Chem 2024; 300:107229. [PMID: 38537698 PMCID: PMC11066510 DOI: 10.1016/j.jbc.2024.107229] [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: 02/14/2024] [Revised: 03/18/2024] [Accepted: 03/20/2024] [Indexed: 04/19/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells can elicit immune responses against riboflavin-based antigens presented by the evolutionary conserved MHC class I related protein, MR1. While we have an understanding of the structural basis of human MAIT cell receptor (TCR) recognition of human MR1 presenting a variety of ligands, how the semi-invariant mouse MAIT TCR binds mouse MR1-ligand remains unknown. Here, we determine the crystal structures of 2 mouse TRAV1-TRBV13-2+ MAIT TCR-MR1-5-OP-RU ternary complexes, whose TCRs differ only in the composition of their CDR3β loops. These mouse MAIT TCRs mediate high affinity interactions with mouse MR1-5-OP-RU and cross-recognize human MR1-5-OP-RU. Similarly, a human MAIT TCR could bind mouse MR1-5-OP-RU with high affinity. This cross-species recognition indicates the evolutionary conserved nature of this MAIT TCR-MR1 axis. Comparing crystal structures of the mouse versus human MAIT TCR-MR1-5-OP-RU complexes provides structural insight into the conserved nature of this MAIT TCR-MR1 interaction and conserved specificity for the microbial antigens, whereby key germline-encoded interactions required for MAIT activation are maintained. This is an important consideration for the development of MAIT cell-based therapeutics that will rely on preclinical mouse models of disease.
Collapse
Affiliation(s)
- Lisa Ciacchi
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jeffrey Y W Mak
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - Jeremy P Le
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - David P Fairlie
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Alexandra J Corbett
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, Victoria, Australia
| | - Jamie Rossjohn
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Institute of Infection and Immunity, School of Medicine, Cardiff University, Cardiff, UK.
| | - Wael Awad
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
12
|
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
|
13
|
McWilliam HEG, Villadangos JA. MR1 antigen presentation to MAIT cells and other MR1-restricted T cells. Nat Rev Immunol 2024; 24:178-192. [PMID: 37773272 PMCID: PMC11108705 DOI: 10.1038/s41577-023-00934-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2023] [Indexed: 10/01/2023]
Abstract
MHC antigen presentation plays a fundamental role in adaptive and semi-invariant T cell immunity. Distinct MHC molecules bind antigens that differ in chemical structure, origin and location and present them to specialized T cells. MHC class I-related protein 1 (MR1) presents a range of small molecule antigens to MR1-restricted T (MR1T) lymphocytes. The best studied MR1 ligands are derived from microbial metabolism and are recognized by a major class of MR1T cells known as mucosal-associated invariant T (MAIT) cells. Here, we describe the MR1 antigen presentation pathway: the known types of antigens presented by MR1, the location where MR1-antigen complexes form, the route followed by the complexes to the cell surface, the mechanisms involved in termination of MR1 antigen presentation and the accessory cellular proteins that comprise the MR1 antigen presentation machinery. The current road map of the MR1 antigen presentation pathway reveals potential strategies for therapeutic manipulation of MR1T cell function and provides a foundation for further studies that will lead to a deeper understanding of MR1-mediated immunity.
Collapse
Affiliation(s)
- Hamish E G McWilliam
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia.
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia.
| | - Jose A Villadangos
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia.
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
14
|
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
|
15
|
Bugaut H, El Morr Y, Mestdagh M, Darbois A, Paiva RA, Salou M, Perrin L, Fürstenheim M, du Halgouet A, Bilonda-Mutala L, Le Gac AL, Arnaud M, El Marjou A, Guerin C, Chaiyasitdhi A, Piquet J, Smadja DM, Cieslak A, Ryffel B, Maciulyte V, Turner JM, Bernardeau K, Montagutelli X, Lantz O, Legoux F. A conserved transcriptional program for MAIT cells across mammalian evolution. J Exp Med 2024; 221:e20231487. [PMID: 38117256 PMCID: PMC10733631 DOI: 10.1084/jem.20231487] [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: 08/22/2023] [Revised: 10/20/2023] [Accepted: 11/22/2023] [Indexed: 12/21/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells harbor evolutionarily conserved TCRs, suggesting important functions. As human and mouse MAIT functional programs appear distinct, the evolutionarily conserved MAIT functional features remain unidentified. Using species-specific tetramers coupled to single-cell RNA sequencing, we characterized MAIT cell development in six species spanning 110 million years of evolution. Cross-species analyses revealed conserved transcriptional events underlying MAIT cell maturation, marked by ZBTB16 induction in all species. MAIT cells in human, sheep, cattle, and opossum acquired a shared type-1/17 transcriptional program, reflecting ancestral features. This program was also acquired by human iNKT cells, indicating common differentiation for innate-like T cells. Distinct type-1 and type-17 MAIT subsets developed in rodents, including pet mice and genetically diverse mouse strains. However, MAIT cells further matured in mouse intestines to acquire a remarkably conserved program characterized by concomitant expression of type-1, type-17, cytotoxicity, and tissue-repair genes. Altogether, the study provides a unifying view of the transcriptional features of innate-like T cells across evolution.
Collapse
Affiliation(s)
- Hélène Bugaut
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
| | - Yara El Morr
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
| | - Martin Mestdagh
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
| | - Aurélie Darbois
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
| | - Rafael A. Paiva
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
| | - Marion Salou
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
| | - Laetitia Perrin
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
| | - Mariela Fürstenheim
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
- Université Paris Cité, Paris, France
| | - Anastasia du Halgouet
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
| | - Linda Bilonda-Mutala
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
| | - Anne-Laure Le Gac
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
| | - Manon Arnaud
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
| | | | - Coralie Guerin
- Cytometry Platform, CurieCoreTech, Institut Curie, Paris, France
- Innovative Therapies in Haemostasis, Institut National de La Santé et de La Recherche Médicale, Université de Paris, Paris, France
| | - Atitheb Chaiyasitdhi
- Laboratoire Physico-Chimie Curie, Institut Curie, Paris Sciences et Lettres Research University, Centre national de la recherche scientifique UMR168, Paris, France
- Sorbonne Université, Paris, France
| | - Julie Piquet
- Biosurgical Research Laboratory, Carpentier Foundation, Paris, France
| | - David M. Smadja
- Innovative Therapies in Haemostasis, Institut National de La Santé et de La Recherche Médicale, Université de Paris, Paris, France
- Hematology Department and Biosurgical Research Lab (Carpentier Foundation), Assistance Publique Hôpitaux de Paris-Centre-Université de Paris, Paris, France
| | - Agata Cieslak
- Université de Paris (Descartes), Institut Necker-Enfants Malades, Institut National de La Santé et de La Recherche Médicale U1151, and Laboratory of Onco-Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Paris, France
| | - Bernhard Ryffel
- Université D’Orléans, Centre national de la recherche scientifique UMR7355, Orléans, France
| | - Valdone Maciulyte
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - James M.A. Turner
- Sex Chromosome Biology Laboratory, The Francis Crick Institute, London, UK
| | - Karine Bernardeau
- Nantes Université, Centre hospitalier universitaire de Nantes, Centre national de la recherche scientifique, Institut National de La Santé et de La Recherche Médicale, BioCore, US16, Plateforme P2R, Structure Fédérative de Recherche François Bonamy, Nantes, France
| | - Xavier Montagutelli
- Mouse Genetics Laboratory, Institut Pasteur, Université Paris Cité, Paris, France
| | - Olivier Lantz
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
- Laboratoire D’immunologie Clinique, Institut Curie, Paris, France
- Centre D’investigation Clinique en Biothérapie Gustave-Roussy Institut Curie, Paris, France
| | - François Legoux
- Institut Curie, Paris Sciences et Lettres University, Institut National de La Santé et de La Recherche Médicale U932, Immunity and Cancer, Paris, France
- Institut de Génétique et Développement de Rennes, Université de Rennes, Institut National de La Santé et de La Recherche Médicale ERL1305, Centre national de la recherche scientifique UMR6290, Rennes, France
| |
Collapse
|
16
|
Lonez C, Breman E. Allogeneic CAR-T Therapy Technologies: Has the Promise Been Met? Cells 2024; 13:146. [PMID: 38247837 PMCID: PMC10814647 DOI: 10.3390/cells13020146] [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: 11/29/2023] [Revised: 01/09/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
This last decade, chimeric antigen receptor (CAR) T-cell therapy has become a real treatment option for patients with B-cell malignancies, while multiple efforts are being made to extend this therapy to other malignancies and broader patient populations. However, several limitations remain, including those associated with the time-consuming and highly personalized manufacturing of autologous CAR-Ts. Technologies to establish "off-the-shelf" allogeneic CAR-Ts with low alloreactivity are currently being developed, with a strong focus on gene-editing technologies. Although these technologies have many advantages, they have also strong limitations, including double-strand breaks in the DNA with multiple associated safety risks as well as the lack of modulation. As an alternative, non-gene-editing technologies provide an interesting approach to support the development of allogeneic CAR-Ts in the future, with possibilities of fine-tuning gene expression and easy development. Here, we will review the different ways allogeneic CAR-Ts can be manufactured and discuss which technologies are currently used. The biggest hurdles for successful therapy of allogeneic CAR-Ts will be summarized, and finally, an overview of the current clinical evidence for allogeneic CAR-Ts in comparison to its autologous counterpart will be given.
Collapse
|
17
|
Wu S, Yang X, Lou Y, Xiao X. MAIT cells in bacterial infectious diseases: heroes, villains, or both? Clin Exp Immunol 2023; 214:144-153. [PMID: 37624404 PMCID: PMC10714195 DOI: 10.1093/cei/uxad102] [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: 04/04/2023] [Revised: 08/20/2023] [Accepted: 08/21/2023] [Indexed: 08/26/2023] Open
Abstract
Due to the aggravation of bacterial drug resistance and the lag in the development of new antibiotics, it is crucial to develop novel therapeutic regimens for bacterial infectious diseases. Currently, immunotherapy is a promising regimen for the treatment of infectious diseases. Mucosal-associated invariant T (MAIT) cells, a subpopulation of innate-like T cells, are abundant in humans and can mount a rapid immune response to pathogens, thus becoming a potential target of immunotherapy for infectious diseases. At the site of infection, activated MAIT cells perform complex biological functions by secreting a variety of cytokines and cytotoxic substances. Many studies have shown that MAIT cells have immunoprotective effects because they can bridge innate and adaptive immune responses, leading to bacterial clearance, tissue repair, and homeostasis maintenance. MAIT cells also participate in cytokine storm generation, tissue fibrosis, and cancer progression, indicating that they play a role in immunopathology. In this article, we review recent studies of MAIT cells, discuss their dual roles in bacterial infectious diseases and provide some promising MAIT cell-targeting strategies for the treatment of bacterial infectious diseases.
Collapse
Affiliation(s)
- Sihong Wu
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xi Yang
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yongliang Lou
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xingxing Xiao
- Wenzhou Key Laboratory of Sanitary Microbiology, Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
18
|
Waterhölter A, Wunderlich M, Turner JE. MAIT cells in immune-mediated tissue injury and repair. Eur J Immunol 2023; 53:e2350483. [PMID: 37740567 DOI: 10.1002/eji.202350483] [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: 06/23/2023] [Revised: 09/20/2023] [Accepted: 09/21/2023] [Indexed: 09/24/2023]
Abstract
Mucosal-associated invariant T (MAIT) cells are T cells that express a semi-invariant αβ T-cell receptor (TCR), recognizing non-peptide antigens, such as microbial-derived vitamin B2 metabolites, presented by the nonpolymorphic MHC class I related-1 molecule. Like NKT cells and γδT cells, MAIT cells belong to the group of innate-like T cells that combine properties of the innate and adaptive immune systems. They account for up to 10% of the blood T-cell population in humans and are particularly abundant at mucosal sites. Beyond the emerging role of MAIT cells in antibacterial and antiviral defenses, increasing evidence suggests additional functions in noninfectious settings, including immune-mediated inflammatory diseases and tissue repair. Here, we discuss recent advances in the understanding of MAIT cell functions in sterile tissue inflammation, with a particular focus on autoimmunity, chronic inflammatory diseases, and tissue repair.
Collapse
Affiliation(s)
- Alex Waterhölter
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Wunderlich
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan-Eric Turner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
19
|
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
|
20
|
Gnirck AC, Philipp MS, Waterhölter A, Wunderlich M, Shaikh N, Adamiak V, Henneken L, Kautz T, Xiong T, Klaus D, Tomczyk P, Al-Bahra MM, Menche D, Walkenhorst M, Lantz O, Willing A, Friese MA, Huber TB, Krebs CF, Panzer U, Kurts C, Turner JE. Mucosal-associated invariant T cells contribute to suppression of inflammatory myeloid cells in immune-mediated kidney disease. Nat Commun 2023; 14:7372. [PMID: 37968302 PMCID: PMC10651937 DOI: 10.1038/s41467-023-43269-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 11/06/2023] [Indexed: 11/17/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells have been implicated in various inflammatory diseases of barrier organs, but so far, their role in kidney disease is unclear. Here we report that MAIT cells that recognize their prototypical ligand, the vitamin B2 intermediate 5-OP-RU presented by MR1, reside in human and mouse kidneys. Single cell RNAseq analysis reveals several intrarenal MAIT subsets, and one, carrying the genetic fingerprint of tissue-resident MAIT17 cells, is activated and expanded in a murine model of crescentic glomerulonephritis (cGN). An equivalent subset is also present in kidney biopsies of patients with anti-neutrophil cytoplasmatic antibody (ANCA)-associated cGN. MAIT cell-deficient MR1 mice show aggravated disease, whereas B6-MAITCAST mice, harboring higher MAIT cell numbers, are protected from cGN. The expanded MAIT17 cells express anti-inflammatory mediators known to suppress cGN, such as CTLA-4, PD-1, and TGF-β. Interactome analysis predicts CXCR6 - CXCL16-mediated cross-talk with renal mononuclear phagocytes, known to drive cGN progression. In line, we find that cGN is aggravated upon CXCL16 blockade. Finally, we present an optimized 5-OP-RU synthesis method which we apply to attenuating cGN in mice. In summary, we propose that CXCR6+ MAIT cells might play a protective role in cGN, implicating them as a potential target for anti-inflammatory therapies.
Collapse
Affiliation(s)
- Ann-Christin Gnirck
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Euroimmun, Lübeck, Germany
| | - Marie-Sophie Philipp
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
- Division of Immunology, Paul-Ehrlich-Institut Langen, Langen, Germany
| | - Alex Waterhölter
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Malte Wunderlich
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nikhat Shaikh
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Virginia Adamiak
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lena Henneken
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias Kautz
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institut für Transfusionsmedizin, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tingting Xiong
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Daniela Klaus
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Pascal Tomczyk
- Kekulé Institute of Organic Chemistry and Biochemistry, University of Bonn, Bonn, Germany
| | - Mohamad M Al-Bahra
- Kekulé Institute of Organic Chemistry and Biochemistry, University of Bonn, Bonn, Germany
| | - Dirk Menche
- Kekulé Institute of Organic Chemistry and Biochemistry, University of Bonn, Bonn, Germany
| | - Mark Walkenhorst
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Olivier Lantz
- Inserm U932, Laboratoire d'immunologie Clinique and Centre d'investigation Clinique en Biothérapie Gustave-Roussy, Institut Curie, Paris, France
| | - Anne Willing
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tobias B Huber
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian F Krebs
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ulf Panzer
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Christian Kurts
- Institute of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany.
- Department of Microbiology and Immunology, Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.
| | - Jan-Eric Turner
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Translational Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
- Hamburg Center for Kidney Health (HCKH), University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
| |
Collapse
|
21
|
Chandra S, Ascui G, Riffelmacher T, Chawla A, Ramírez-Suástegui C, Castelan VC, Seumois G, Simon H, Murray MP, Seo GY, Premlal ALR, Schmiedel B, Verstichel G, Li Y, Lin CH, Greenbaum J, Lamberti J, Murthy R, Nigro J, Cheroutre H, Ottensmeier CH, Hedrick SM, Lu LF, Vijayanand P, Kronenberg M. Transcriptomes and metabolism define mouse and human MAIT cell populations. Sci Immunol 2023; 8:eabn8531. [PMID: 37948512 PMCID: PMC11160507 DOI: 10.1126/sciimmunol.abn8531] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 10/05/2023] [Indexed: 11/12/2023]
Abstract
Mucosal-associated invariant T (MAIT) cells are a subset of T lymphocytes that respond to microbial metabolites. We defined MAIT cell populations in different organs and characterized the developmental pathway of mouse and human MAIT cells in the thymus using single-cell RNA sequencing and phenotypic and metabolic analyses. We showed that the predominant mouse subset, which produced IL-17 (MAIT17), and the subset that produced IFN-γ (MAIT1) had not only greatly different transcriptomes but also different metabolic states. MAIT17 cells in different organs exhibited increased lipid uptake, lipid storage, and mitochondrial potential compared with MAIT1 cells. All these properties were similar in the thymus and likely acquired there. Human MAIT cells in lung and blood were more homogeneous but still differed between tissues. Human MAIT cells had increased fatty acid uptake and lipid storage in blood and lung, similar to human CD8 T resident memory cells, but unlike mouse MAIT17 cells, they lacked increased mitochondrial potential. Although mouse and human MAIT cell transcriptomes showed similarities for immature cells in the thymus, they diverged more strikingly in the periphery. Analysis of pet store mice demonstrated decreased lung MAIT17 cells in these so-called "dirty" mice, indicative of an environmental influence on MAIT cell subsets and function.
Collapse
Affiliation(s)
- Shilpi Chandra
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Gabriel Ascui
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093 USA
| | - Thomas Riffelmacher
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, OX3 7FY UK
| | - Ashu Chawla
- Bioinformatics Core Facility, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Ciro Ramírez-Suástegui
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Viankail C. Castelan
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Gregory Seumois
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Hayley Simon
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Mallory P. Murray
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Goo-Young Seo
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | | | - Benjamin Schmiedel
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Greet Verstichel
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Yingcong Li
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92037 USA
| | - Chia-Hao Lin
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92037 USA
| | - Jason Greenbaum
- Bioinformatics Core Facility, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - John Lamberti
- Division of Cardiac Surgery, Rady Children’s Hospital, San Diego, CA 92123 USA
- Division of Pediatric Cardiac Surgery, Falk Cardiovascular Research Center, Stanford, CA 94305-5407 USA
| | - Raghav Murthy
- Division of Cardiac Surgery, Rady Children’s Hospital, San Diego, CA 92123 USA
- Division of Pediatric Cardiac Surgery, Children’s Heart Center Icahn School of Medicine at Mount Sinai, New York, NY 10029 USA
| | - John Nigro
- Division of Cardiac Surgery, Rady Children’s Hospital, San Diego, CA 92123 USA
| | - Hilde Cheroutre
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Christian H. Ottensmeier
- Liverpool Head and Neck Center, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK, L69 7ZB
| | - Stephen M. Hedrick
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92037 USA
- Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA, 92093 USA
| | - Li-Fan Lu
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92037 USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, 92093 USA
- Moores Cancer Center, University of California, San Diego, La Jolla, CA, 92093 USA
| | - Pandurangan Vijayanand
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
| | - Mitchell Kronenberg
- Center for Autoimmunity and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA 92037 USA
- Department of Molecular Biology, University of California, San Diego, La Jolla, CA 92037 USA
| |
Collapse
|
22
|
Sugimoto C, Fujita H, Wakao H. A flow-cytometry-based assay to assess the cytolytic activity against tumor cells by combination of mouse MAIT cells and natural killer cells. STAR Protoc 2023; 4:102620. [PMID: 39491553 PMCID: PMC10628808 DOI: 10.1016/j.xpro.2023.102620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/24/2023] [Accepted: 09/13/2023] [Indexed: 11/05/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are innate-like T cells responsible for mucosal immunity in the respiratory and intestinal tracts. Here we present a flow-cytometry-based assay to measure the cytolytic activity of murine MAIT cells and natural killer (NK) cells. We describe steps for differentiating MAIT-like cells from the induced pluripotent stem cells prepared from MAIT cells (reMAIT cells), NK cell isolation, co-culture with target tumor cells, and staining to distinguish dead cells from live cells. For complete details on the use and execution of this protocol, please refer to Sugimoto et al. (2022).1.
Collapse
Affiliation(s)
- Chie Sugimoto
- Host Defense Division, Research Center for Advanced Medical Science, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan.
| | - Hiroyoshi Fujita
- Host Defense Division, Research Center for Advanced Medical Science, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan
| | - Hiroshi Wakao
- Host Defense Division, Research Center for Advanced Medical Science, Dokkyo Medical University, Mibu, Tochigi 321-0293, Japan.
| |
Collapse
|
23
|
Lv M, Zhang Z, Cui Y. Unconventional T cells in brain homeostasis, injury and neurodegeneration. Front Immunol 2023; 14:1273459. [PMID: 37854609 PMCID: PMC10579804 DOI: 10.3389/fimmu.2023.1273459] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 09/20/2023] [Indexed: 10/20/2023] Open
Abstract
The interaction between peripheral immune cells and the brain is an important component of the neuroimmune axis. Unconventional T cells, which include natural killer T (NKT) cells, mucosal-associated invariant T (MAIT) cells, γδ T cells, and other poorly defined subsets, are a special group of T lymphocytes that recognize a wide range of nonpolymorphic ligands and are the connection between adaptive and innate immunity. Recently, an increasing number of complex functions of these unconventional T cells in brain homeostasis and various brain disorders have been revealed. In this review, we describe the classification and effector function of unconventional T cells, review the evidence for the involvement of unconventional T cells in the regulation of brain homeostasis, summarize the roles and mechanisms of unconventional T cells in the regulation of brain injury and neurodegeneration, and discuss immunotherapeutic potential as well as future research goals. Insight of these processes can shed light on the regulation of T cell immunity on brain homeostasis and diseases and provide new clues for therapeutic approaches targeting brain injury and neurodegeneration.
Collapse
Affiliation(s)
- Mengfei Lv
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| | - Zhaolong Zhang
- Department of Interventional Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yu Cui
- Institute of Neuroregeneration and Neurorehabilitation, Qingdao University, Qingdao, Shandong, China
- Qingdao Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
24
|
López-Rodríguez JC, Hancock SJ, Li K, Crotta S, Barrington C, Suárez-Bonnet A, Priestnall SL, Aubé J, Wack A, Klenerman P, Bengoechea JA, Barral P. Type I interferons drive MAIT cell functions against bacterial pneumonia. J Exp Med 2023; 220:e20230037. [PMID: 37516912 PMCID: PMC10373297 DOI: 10.1084/jem.20230037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 05/31/2023] [Accepted: 07/11/2023] [Indexed: 07/31/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are abundant in the lung and contribute to host defense against infections. During bacterial infections, MAIT cell activation has been proposed to require T cell receptor (TCR)-mediated recognition of antigens derived from the riboflavin synthesis pathway presented by the antigen-presenting molecule MR1. MAIT cells can also be activated by cytokines in an MR1-independent manner, yet the contribution of MR1-dependent vs. -independent signals to MAIT cell functions in vivo remains unclear. Here, we use Klebsiella pneumoniae as a model of bacterial pneumonia and demonstrate that MAIT cell activation is independent of MR1 and primarily driven by type I interferons (IFNs). During Klebsiella infection, type I IFNs stimulate activation of murine and human MAIT cells, induce a Th1/cytotoxic transcriptional program, and modulate MAIT cell location within the lungs. Consequently, adoptive transfer or boosting of pulmonary MAIT cells protect mice from Klebsiella infection, with protection being dependent on direct type I IFN signaling on MAIT cells. These findings reveal type I IFNs as new molecular targets to manipulate MAIT cell functions during bacterial infections.
Collapse
Affiliation(s)
- Juan Carlos López-Rodríguez
- The Peter Gorer Department of Immunobiology, King’s College London, London, UK
- The Francis Crick Institute, London, UK
| | - Steven J. Hancock
- Wellcome-Wolfson Institute for Experimental Medicine. School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, UK
| | - Kelin Li
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Alejandro Suárez-Bonnet
- The Francis Crick Institute, London, UK
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, UK
| | - Simon L. Priestnall
- The Francis Crick Institute, London, UK
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hatfield, UK
| | - Jeffrey Aubé
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, Oxford, UK
| | - Jose A. Bengoechea
- Wellcome-Wolfson Institute for Experimental Medicine. School of Medicine, Dentistry and Biomedical Sciences, Queen’s University Belfast, Belfast, UK
| | - Patricia Barral
- The Peter Gorer Department of Immunobiology, King’s College London, London, UK
- The Francis Crick Institute, London, UK
| |
Collapse
|
25
|
Zheng Y, Han F, Ho A, Xue Y, Wu Z, Chen X, Sandberg JK, Ma S, Leeansyah E. Role of MAIT cells in gastrointestinal tract bacterial infections in humans: More than a gut feeling. Mucosal Immunol 2023; 16:740-752. [PMID: 37353006 DOI: 10.1016/j.mucimm.2023.06.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023]
Abstract
Mucosa-associated invariant T (MAIT) cells are the largest population of unconventional T cells in humans. These antimicrobial T cells are poised with rapid effector responses following recognition of the cognate riboflavin (vitamin B2)-like metabolite antigens derived from microbial riboflavin biosynthetic pathway. Presentation of this unique class of small molecule metabolite antigens is mediated by the highly evolutionarily conserved major histocompatibility complex class I-related protein. In humans, MAIT cells are widely found along the upper and lower gastrointestinal tracts owing to their high expression of chemokine receptors and homing molecules directing them to these tissue sites. In this review, we discuss recent findings regarding the roles MAIT cells play in various gastrointestinal bacterial infections, and how their roles appear to differ depending on the etiological agents and the anatomical location. We further discuss the potential mechanisms by which MAIT cells contribute to pathogen control, orchestrate adaptive immunity, as well as their potential contribution to inflammation and tissue damage during gastrointestinal bacterial infections, and the ensuing tissue repair following resolution. Finally, we propose and discuss the use of the emerging three-dimensional organoid technology to test different hypotheses regarding the role of MAIT cells in gastrointestinal bacterial infections, inflammation, and immunity.
Collapse
Affiliation(s)
- 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
| | - Fei Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, 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
| | - Yiting Xue
- 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
| | - Zhengyu Wu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Xingchi Chen
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Johan K Sandberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Shaohua Ma
- 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
| | - Edwin Leeansyah
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China.
| |
Collapse
|
26
|
Corbett AJ, Ussher JE, Hinks TSC. Editorial: MAIT cells come of age. Front Immunol 2023; 14:1281881. [PMID: 37744347 PMCID: PMC10513092 DOI: 10.3389/fimmu.2023.1281881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Affiliation(s)
- Alexandra J. Corbett
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - James E. Ussher
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| | - Timothy S. C. Hinks
- Respiratory Medicine Unit and National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, United Kingdom
| |
Collapse
|
27
|
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: 10] [Impact Index Per Article: 5.0] [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
|
28
|
Nelson AG, Wang H, Dewar PM, Eddy EM, Li S, Lim XY, Patton T, Zhou Y, Pediongco TJ, Meehan LJ, Meehan BS, Mak JYW, Fairlie DP, Stent AW, Kjer-Nielsen L, McCluskey J, Eckle SBG, Corbett AJ, Souter MNT, Chen Z. Synthetic 5-amino-6-D-ribitylaminouracil paired with inflammatory stimuli facilitates MAIT cell expansion in vivo. Front Immunol 2023; 14:1109759. [PMID: 37720229 PMCID: PMC10500299 DOI: 10.3389/fimmu.2023.1109759] [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: 11/28/2022] [Accepted: 07/12/2023] [Indexed: 09/19/2023] Open
Abstract
Introduction Mucosal-associated invariant T (MAIT) cells are a population of innate-like T cells, which mediate host immunity to microbial infection by recognizing metabolite antigens derived from microbial riboflavin synthesis presented by the MHC-I-related protein 1 (MR1). Namely, the potent MAIT cell antigens, 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil (5-OP-RU) and 5-(2-oxoethylideneamino)-6-D-ribitylaminouracil (5-OE-RU), form via the condensation of the riboflavin precursor 5-amino-6-D-ribitylaminouracil (5-A-RU) with the reactive carbonyl species (RCS) methylglyoxal (MG) and glyoxal (G), respectively. Although MAIT cells are abundant in humans, they are rare in mice, and increasing their abundance using expansion protocols with antigen and adjuvant has been shown to facilitate their study in mouse models of infection and disease. Methods Here, we outline three methods to increase the abundance of MAIT cells in C57BL/6 mice using a combination of inflammatory stimuli, 5-A-RU and MG. Results Our data demonstrate that the administration of synthetic 5-A-RU in combination with one of three different inflammatory stimuli is sufficient to increase the frequency and absolute numbers of MAIT cells in C57BL/6 mice. The resultant boosted MAIT cells are functional and can provide protection against a lethal infection of Legionella longbeachae. Conclusion These results provide alternative methods for expanding MAIT cells with high doses of commercially available 5-A-RU (± MG) in the presence of various danger signals.
Collapse
Affiliation(s)
- Adam G. Nelson
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Huimeng Wang
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Phoebe M. Dewar
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Eleanor M. Eddy
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Songyi Li
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Xin Yi Lim
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Timothy Patton
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia
| | - Yuchen Zhou
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Troi J. Pediongco
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Lucy J. Meehan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Bronwyn S. Meehan
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Jeffrey Y. W. Mak
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | - David P. Fairlie
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, Australia
| | | | - Lars Kjer-Nielsen
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Sidonia B. G. Eckle
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Alexandra J. Corbett
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Michael N. T. Souter
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Zhenjun Chen
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| |
Collapse
|
29
|
Xu C, Li S, Fulford TS, Christo SN, Mackay LK, Gray DH, Uldrich AP, Pellicci DG, I Godfrey D, Koay HF. Expansion of MAIT cells in the combined absence of NKT and γδ-T cells. Mucosal Immunol 2023; 16:446-461. [PMID: 37182737 DOI: 10.1016/j.mucimm.2023.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/11/2023] [Accepted: 05/03/2023] [Indexed: 05/16/2023]
Abstract
Mucosal-associated invariant T (MAIT) cells, natural killer T (NKT) cells, and γδT cells are collectively referred to as 'unconventional T cells' due to their recognition of non-peptide antigens and restriction to MHC-I-like molecules. However, the factors controlling their widely variable frequencies between individuals and organs are poorly understood. We demonstrated that MAIT cells are increased in NKT or γδT cell-deficient mice and highly expand in mice lacking both cell types. TCRα repertoire analysis of γδT cell-deficient thymocytes revealed altered Trav segment usage relative to wild-type thymocytes, highlighting retention of the Tcra-Tcrd locus from the 129 mouse strain used to generate Tcrd-/- mice. This resulted in a moderate increase in distal Trav segment usage, including Trav1, potentially contributing to increased generation of Trav1-Traj33+ MAIT cells in the Tcrd-/- thymus. Importantly, adoptively transferred MAIT cells underwent increased homeostatic proliferation within NKT/gdT cell-deficient tissues, with MAIT cell subsets exhibiting tissue-specific homing patterns. Our data reveal a shared niche for unconventional T cells, where competition for common factors may be exploited to collectively modulate these cells in the immune response. Lastly, our findings emphasise careful assessment of studies using NKT or γδT cell-deficient mice when investigating the role of unconventional T cells in disease.
Collapse
Affiliation(s)
- Calvin Xu
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Shihan Li
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Thomas S Fulford
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Susan N Christo
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Laura K Mackay
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Daniel Hd Gray
- Walter and Eliza Hall Institute of Medical Research, Parkville, Australia; Department of Medical Biology, University of Melbourne, Parkville, Australia
| | - Adam P Uldrich
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia
| | - Daniel G Pellicci
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia; Murdoch Children's Research Institute, Melbourne, Australia; Department of Paediatrics, University of Melbourne, Parkville, Australia.
| | - Dale I Godfrey
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia.
| | - Hui-Fern Koay
- Department of Microbiology and Immunology at the Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Parkville, Australia.
| |
Collapse
|
30
|
Lv Z, Luo F, Chu Y. Strategies for overcoming bottlenecks in allogeneic CAR-T cell therapy. Front Immunol 2023; 14:1199145. [PMID: 37554322 PMCID: PMC10405079 DOI: 10.3389/fimmu.2023.1199145] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/05/2023] [Indexed: 08/10/2023] Open
Abstract
Patient-derived autologous chimeric antigen receptor (CAR)-T cell therapy is a revolutionary breakthrough in immunotherapy and has made impressive progress in both preclinical and clinical studies. However, autologous CAR-T cells still have notable drawbacks in clinical manufacture, such as long production time, variable cell potency and possible manufacturing failures. Allogeneic CAR-T cell therapy is significantly superior to autologous CAR-T cell therapy in these aspects. The use of allogeneic CAR-T cell therapy may provide simplified manufacturing process and allow the creation of 'off-the-shelf' products, facilitating the treatments of various types of tumors at less delivery time. Nevertheless, severe graft-versus-host disease (GvHD) or host-mediated allorejection may occur in the allogeneic setting, implying that addressing these two critical issues is urgent for the clinical application of allogeneic CAR-T cell therapy. In this review, we summarize the current approaches to overcome GvHD and host rejection, which empower allogeneic CAR-T cell therapy with a broader future.
Collapse
Affiliation(s)
- Zixin Lv
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Biotherapy Research Center, Fudan University, Shanghai, China
| | - Feifei Luo
- Biotherapy Research Center, Fudan University, Shanghai, China
- Department of Digestive Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Yiwei Chu
- Department of Immunology, School of Basic Medical Sciences, and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
- Biotherapy Research Center, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Wang NI, Ninkov M, Haeryfar SMM. Classic costimulatory interactions in MAIT cell responses: from gene expression to immune regulation. Clin Exp Immunol 2023; 213:50-66. [PMID: 37279566 PMCID: PMC10324557 DOI: 10.1093/cei/uxad061] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 05/17/2023] [Accepted: 06/01/2023] [Indexed: 06/08/2023] Open
Abstract
Mucosa-associated invariant T (MAIT) cells are evolutionarily conserved, innate-like T lymphocytes with enormous immunomodulatory potentials. Due to their strategic localization, their invariant T cell receptor (iTCR) specificity for major histocompatibility complex-related protein 1 (MR1) ligands of commensal and pathogenic bacterial origin, and their sensitivity to infection-elicited cytokines, MAIT cells are best known for their antimicrobial characteristics. However, they are thought to also play important parts in the contexts of cancer, autoimmunity, vaccine-induced immunity, and tissue repair. While cognate MR1 ligands and cytokine cues govern MAIT cell maturation, polarization, and peripheral activation, other signal transduction pathways, including those mediated by costimulatory interactions, regulate MAIT cell responses. Activated MAIT cells exhibit cytolytic activities and secrete potent inflammatory cytokines of their own, thus transregulating the biological behaviors of several other cell types, including dendritic cells, macrophages, natural killer cells, conventional T cells, and B cells, with significant implications in health and disease. Therefore, an in-depth understanding of how costimulatory pathways control MAIT cell responses may introduce new targets for optimized MR1/MAIT cell-based interventions. Herein, we compare and contrast MAIT cells and mainstream T cells for their expression of classic costimulatory molecules belonging to the immunoglobulin superfamily and the tumor necrosis factor (TNF)/TNF receptor superfamily, based not only on the available literature but also on our transcriptomic analyses. We discuss how these molecules participate in MAIT cells' development and activities. Finally, we introduce several pressing questions vis-à-vis MAIT cell costimulation and offer new directions for future research in this area.
Collapse
Affiliation(s)
- Nicole I Wang
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
| | - Marina Ninkov
- Department of Microbiology and Immunology, Western University, London, 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
|
32
|
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: 0] [Impact Index Per Article: 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
|
33
|
Riffelmacher T, Paynich Murray M, Wientjens C, Chandra S, Cedillo-Castelán V, Chou TF, McArdle S, Dillingham C, Devereaux J, Nilsen A, Brunel S, Lewinsohn DM, Hasty J, Seumois G, Benedict CA, Vijayanand P, Kronenberg M. Divergent metabolic programmes control two populations of MAIT cells that protect the lung. Nat Cell Biol 2023; 25:877-891. [PMID: 37231163 PMCID: PMC10264248 DOI: 10.1038/s41556-023-01152-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 04/18/2023] [Indexed: 05/27/2023]
Abstract
Although mucosal-associated invariant T (MAIT) cells provide rapid, innate-like responses, they are not pre-set, and memory-like responses have been described for MAIT cells following infections. The importance of metabolism for controlling these responses, however, is unknown. Here, following pulmonary immunization with a Salmonella vaccine strain, mouse MAIT cells expanded as separate CD127-Klrg1+ and CD127+Klrg1- antigen-adapted populations that differed in terms of their transcriptome, function and localization in lung tissue. These populations remained altered from steady state for months as stable, separate MAIT cell lineages with enhanced effector programmes and divergent metabolism. CD127+ MAIT cells engaged in an energetic, mitochondrial metabolic programme, which was critical for their maintenance and IL-17A synthesis. This programme was supported by high fatty acid uptake and mitochondrial oxidation and relied on highly polarized mitochondria and autophagy. After vaccination, CD127+ MAIT cells protected mice against Streptococcus pneumoniae infection. In contrast, Klrg1+ MAIT cells had dormant but ready-to-respond mitochondria and depended instead on Hif1a-driven glycolysis to survive and produce IFN-γ. They responded antigen independently and participated in protection from influenza virus. These metabolic dependencies may enable tuning of memory-like MAIT cell responses for vaccination and immunotherapies.
Collapse
Affiliation(s)
- Thomas Riffelmacher
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
| | | | | | | | | | | | - Sara McArdle
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | | - Aaron Nilsen
- Oregon Health and Science University, Portland, OR, USA
| | - Simon Brunel
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Jeff Hasty
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA
| | | | | | | | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA, USA.
- Department of Molecular Biology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
34
|
Pankhurst TE, Buick KH, Lange JL, Marshall AJ, Button KR, Palmer OR, Farrand KJ, Montgomerie I, Bird TW, Mason NC, Kuang J, Compton BJ, Comoletti D, Salio M, Cerundolo V, Quiñones-Mateu ME, Painter GF, Hermans IF, Connor LM. MAIT cells activate dendritic cells to promote T FH cell differentiation and induce humoral immunity. Cell Rep 2023; 42:112310. [PMID: 36989114 PMCID: PMC10045373 DOI: 10.1016/j.celrep.2023.112310] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 02/02/2023] [Accepted: 03/13/2023] [Indexed: 03/29/2023] Open
Abstract
Protective immune responses against respiratory pathogens, such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and influenza virus, are initiated by the mucosal immune system. However, most licensed vaccines are administered parenterally and are largely ineffective at inducing mucosal immunity. The development of safe and effective mucosal vaccines has been hampered by the lack of a suitable mucosal adjuvant. In this study we explore a class of adjuvant that harnesses mucosal-associated invariant T (MAIT) cells. We show evidence that intranasal immunization of MAIT cell agonists co-administered with protein, including the spike receptor binding domain from SARS-CoV-2 virus and hemagglutinin from influenza virus, induce protective humoral immunity and immunoglobulin A production. MAIT cell adjuvant activity is mediated by CD40L-dependent activation of dendritic cells and subsequent priming of T follicular helper cells. In summary, we show that MAIT cells are promising vaccine targets that can be utilized as cellular adjuvants in mucosal vaccines.
Collapse
Affiliation(s)
- Theresa E Pankhurst
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Kaitlin H Buick
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Joshua L Lange
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Andrew J Marshall
- Ferrier Research Institute, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Kaileen R Button
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Olga R Palmer
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Kathryn J Farrand
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Isabelle Montgomerie
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Thomas W Bird
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Ngarangi C Mason
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Joanna Kuang
- Department of Microbiology and Immunology, University of Otago, Dunedin 9016, New Zealand
| | - Benjamin J Compton
- Ferrier Research Institute, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Davide Comoletti
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Mariolina Salio
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | - Vincenzo Cerundolo
- Medical Research Council Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford OX3 9DS, UK
| | | | - Gavin F Painter
- Ferrier Research Institute, Victoria University of Wellington, Wellington 6012, New Zealand
| | - Ian F Hermans
- Malaghan Institute of Medical Research, Wellington 6242, New Zealand
| | - Lisa M Connor
- School of Biological Sciences, Victoria University of Wellington, Wellington 6012, New Zealand; Malaghan Institute of Medical Research, Wellington 6242, New Zealand.
| |
Collapse
|
35
|
Sharma M, Niu L, Zhang X, Huang S. Comparative transcriptomes reveal pro-survival and cytotoxic programs of mucosal-associated invariant T cells upon Bacillus Calmette-Guérin stimulation. Front Cell Infect Microbiol 2023; 13:1134119. [PMID: 37091679 PMCID: PMC10116416 DOI: 10.3389/fcimb.2023.1134119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 03/15/2023] [Indexed: 04/08/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are protective against tuberculous and non-tuberculous mycobacterial infections with poorly understood mechanisms. Despite an innate-like nature, MAIT cell responses remain heterogeneous in bacterial infections. To comprehensively characterize MAIT activation programs responding to different bacteria, we stimulated MAIT cells with E. coli to compare with Bacillus Calmette-Guérin (BCG), which remains the only licensed vaccine and a feasible tool for investigating anti-mycobacterial immunity in humans. Upon sequencing mRNA from the activated and inactivated CD8+ MAIT cells, results demonstrated the altered MAIT cell gene profiles by each bacterium with upregulated expression of activation markers, transcription factors, cytokines, and cytolytic mediators crucial in anti-mycobacterial responses. Compared with E. coli, BCG altered more MAIT cell genes to enhance cell survival and cytolysis. Flow cytometry analyses similarly displayed a more upregulated protein expression of B-cell lymphoma 2 and T-box transcription factor Eomesodermin in BCG compared to E.coli stimulations. Thus, the transcriptomic program and protein expression of MAIT cells together displayed enhanced pro-survival and cytotoxic programs in response to BCG stimulation, supporting BCG induces cell-mediated effector responses of MAIT cells to fight mycobacterial infections.
Collapse
Affiliation(s)
| | | | | | - Shouxiong Huang
- Department of Environmental and Public Health Sciences, College of Medicine, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
36
|
Mabire M, Hegde P, Hammoutene A, Wan J, Caër C, Sayegh RA, Cadoux M, Allaire M, Weiss E, Thibault-Sogorb T, Lantz O, Goodhardt M, Paradis V, de la Grange P, Gilgenkrantz H, Lotersztajn S. MAIT cell inhibition promotes liver fibrosis regression via macrophage phenotype reprogramming. Nat Commun 2023; 14:1830. [PMID: 37005415 PMCID: PMC10067815 DOI: 10.1038/s41467-023-37453-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 03/16/2023] [Indexed: 04/04/2023] Open
Abstract
Recent data have shown that liver fibrosis can regress even at later stages of cirrhosis and shifting the immune response from pro-inflammatory towards a resolutive profile is considered as a promising option. The immune regulatory networks that govern the shift of the inflammatory phenotype and thus potential reversal of liver fibrosis are lesser known. Here we show that in precision-cut human liver slices obtained from patients with end-stage fibrosis and in mouse models, inhibiting Mucosal-Associated Invariant T (MAIT) cells using pharmacological or antibody-driven approaches, limits fibrosis progression and even regresses fibrosis, following chronic toxic- or non-alcoholic steatohepatitis (NASH)-induced liver injury. Mechanistic studies, combining RNA sequencing, in vivo functional studies (performed in male mice) and co-culture experiments indicate that disruption of the MAIT cell-monocyte/macrophage interaction results in resolution of fibrosis both by increasing the frequency of restorative Ly6Clo at the expenses of pro-fibrogenic Ly6Chi monocyte-derived macrophages and promoting an autophagic phenotype in both subsets. Thus, our data show that MAIT cell activation and the consequential phenotype shift of liver macrophages are important pathogenic features of liver fibrosis and could be targeted by anti-fibrogenic therapy.
Collapse
Affiliation(s)
- Morgane Mabire
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Pushpa Hegde
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Adel Hammoutene
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Jinghong Wan
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Charles Caër
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Rola Al Sayegh
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Mathilde Cadoux
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Manon Allaire
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Emmanuel Weiss
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
- Département d'Anesthésie et Réanimation, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, 92110, Clichy, France
| | - Tristan Thibault-Sogorb
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
- Département d'Anesthésie et Réanimation, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, 92110, Clichy, France
| | | | - Michèle Goodhardt
- Université Paris Cité, INSERM UMRS 976, Institut de Recherche Saint Louis, F-75010, Paris, France
| | - Valérie Paradis
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
- Département de Pathologie, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris, 92110, Clichy, France
| | | | - Hélène Gilgenkrantz
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France
| | - Sophie Lotersztajn
- Université Paris Cité, INSERM, UMR-S1149, Centre de Recherche sur l'Inflammation (CRI), Laboratoire d'Excellence Inflamex, F-75018, Paris, France.
| |
Collapse
|
37
|
Purohit SK, Corbett AJ, Slobedman B, Abendroth A. Varicella Zoster Virus infects mucosal associated Invariant T cells. Front Immunol 2023; 14:1121714. [PMID: 37006246 PMCID: PMC10063790 DOI: 10.3389/fimmu.2023.1121714] [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: 12/12/2022] [Accepted: 03/06/2023] [Indexed: 03/19/2023] Open
Abstract
IntroductionMucosal Associated Invariant T (MAIT) cells are innate-like T cells that respond to conserved pathogen-derived vitamin B metabolites presented by the MHC class I related-1 molecule (MR1) antigen presentation pathway. Whilst viruses do not synthesize these metabolites, we have reported that varicella zoster virus (VZV) profoundly suppresses MR1 expression, implicating this virus in manipulation of the MR1:MAIT cell axis. During primary infection, the lymphotropism of VZV is likely to be instrumental in hematogenous dissemination of virus to gain access to cutaneous sites where it clinically manifests as varicella (chickenpox). However, MAIT cells, which are found in the blood and at mucosal and other organ sites, have yet to be examined in the context of VZV infection. The goal of this study was to examine any direct impact of VZV on MAIT cells.MethodsUsing flow cytometry, we interrogated whether primary blood derived MAIT cells are permissive to infection by VZV whilst further analysing differential levels of infection between various MAIT cell subpopulations. Changes in cell surface extravasation, skin homing, activation and proliferation markers after VZV infection of MAIT cells was also assessed via flow cytometry. Finally the capacity of MAIT cells to transfer infectious virus was tested through an infectious center assay and imaged via fluorescence microscopy.ResultsWe identify primary blood-derived MAIT cells as being permissive to VZV infection. A consequence of VZV infection of MAIT cells was their capacity to transfer infectious virus to other permissive cells, consistent with MAIT cells supporting productive infection. When subgrouping MAIT cells by their co- expression of a variety cell surface markers, there was a higher proportion of VZV infected MAIT cells co-expressing CD4+ and CD4+/CD8+ MAIT cells compared to the more phenotypically dominant CD8+ MAIT cells, whereas infection was not associated with differences in co-expression of CD56 (MAIT cell subset with enhanced responsiveness to innate cytokine stimulation), CD27 (co-stimulatory) or PD-1 (immune checkpoint). Infected MAIT cells retained high expression of CCR2, CCR5, CCR6, CLA and CCR4, indicating a potentially intact capacity for transendothelial migration, extravasation and trafficking to skin sites. Infected MAIT cells also displayed increased expression of CD69 (early activation) and CD71 (proliferation) markers.DiscussionThese data identify MAIT cells as being permissive to VZV infection and identify impacts of such infection on co- expressed functional markers.
Collapse
Affiliation(s)
- Shivam K. Purohit
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
| | - Alexandra J. Corbett
- Department of Microbiology and Immunology, The University of Melbourne, at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC, Australia
| | - Barry Slobedman
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- *Correspondence: Allison Abendroth, ; Barry Slobedman,
| | - Allison Abendroth
- Infection, Immunity and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Charles Perkins Centre, University of Sydney, Sydney, NSW, Australia
- *Correspondence: Allison Abendroth, ; Barry Slobedman,
| |
Collapse
|
38
|
Witt KD. Role of MHC class I pathways in Mycobacterium tuberculosis antigen presentation. Front Cell Infect Microbiol 2023; 13:1107884. [PMID: 37009503 PMCID: PMC10050577 DOI: 10.3389/fcimb.2023.1107884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Accepted: 02/23/2023] [Indexed: 03/17/2023] Open
Abstract
MHC class I antigen processing is an underappreciated area of nonviral host–pathogen interactions, bridging both immunology and cell biology, where the pathogen’s natural life cycle involves little presence in the cytoplasm. The effective response to MHC-I foreign antigen presentation is not only cell death but also phenotypic changes in other cells and stimulation of the memory cells ready for the next antigen reoccurrence. This review looks at the MHC-I antigen processing pathway and potential alternative sources of the antigens, focusing on Mycobacterium tuberculosis (Mtb) as an intracellular pathogen that co-evolved with humans and developed an array of decoy strategies to survive in a hostile environment by manipulating host immunity to its own advantage. As that happens via the selective antigen presentation process, reinforcement of the effective antigen recognition on MHC-I molecules may stimulate subsets of effector cells that act earlier and more locally. Vaccines against tuberculosis (TB) could potentially eliminate this disease, yet their development has been slow, and success is limited in the context of this global disease’s spread. This review’s conclusions set out potential directions for MHC-I-focused approaches for the next generation of vaccines.
Collapse
Affiliation(s)
- Karolina D. Witt
- Pandemic Sciences Institute, University of Oxford, Oxford, United Kingdom
- Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
- *Correspondence: Karolina D. Witt,
| |
Collapse
|
39
|
Li YR, Zhou K, Wilson M, Kramer A, Zhu Y, Dawson N, Yang L. Mucosal-associated invariant T cells for cancer immunotherapy. Mol Ther 2023; 31:631-646. [PMID: 36463401 PMCID: PMC10014234 DOI: 10.1016/j.ymthe.2022.11.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 11/07/2022] [Accepted: 11/29/2022] [Indexed: 12/09/2022] Open
Abstract
Human mucosal-associated invariant T (MAIT) cells are characterized by their expression of an invariant TCR α chain Vα7.2-Jα33/Jα20/Jα12 paired with a restricted TCR β chain. MAIT cells recognize microbial peptides presented by the highly conserved MHC class I-like molecule MR1 and bridge the innate and acquired immune systems to mediate augmented immune responses. Upon activation, MAIT cells rapidly proliferate, produce a variety of cytokines and cytotoxic molecules, and trigger efficient antitumor immunity. Administration of a representative MAIT cell ligand 5-OP-RU effectively activates MAIT cells and enhances their antitumor capacity. In this review, we introduce MAIT cell biology and their importance in antitumor immunity, summarize the current development of peripheral blood mononuclear cell-derived and stem cell-derived MAIT cell products for cancer treatment, and discuss the potential of genetic engineering of MAIT cells for off-the-shelf cancer immunotherapy.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kuangyi Zhou
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Matthew Wilson
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Adam Kramer
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Yichen Zhu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Niels Dawson
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, 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; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
40
|
New insights into MAIT cells in autoimmune diseases. Biomed Pharmacother 2023; 159:114250. [PMID: 36652733 DOI: 10.1016/j.biopha.2023.114250] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/17/2023] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are resident T cells that express semi-invariant TCR chains and are restricted by monomorphic major histocompatibility complex (MHC) class I-related molecules (MR1). MAIT cells can be activated by microbial-specific metabolites (MR1-dependent mode) or cytokines (MR1-independent mode). Activated MAIT cells produce chemokines, cytotoxic molecules (granzyme B and perforin), and proinflammatory cytokines (IFN-γ, TNF-α, and IL-17), to clear pathogens and target infected cells involved in the pro-inflammatory, migratory, and cytolytic properties of MAIT cells. MAIT cells produce pro-inflammatory cytokines in the target organs of autoimmune diseases and contribute to the development and progression of autoimmune diseases. This article reviews the biological characteristics, activation mechanism, dynamic migration, and dual functions of MAIT cells, and focuses on the mechanism and potential application of MAIT cells in the early diagnosis, disease activity monitoring, and therapeutic targets of autoimmune diseases, to lay a foundation for future research.
Collapse
|
41
|
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
|
42
|
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: 23] [Impact Index Per Article: 11.5] [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
|
43
|
Patton T, Zhao Z, Lim XY, Eddy E, Wang H, Nelson AG, Ennis B, Eckle SBG, Souter MNT, Pediongco TJ, Koay HF, Zhang JG, Djajawi TM, Louis C, Lalaoui N, Jacquelot N, Lew AM, Pellicci DG, McCluskey J, Zhan Y, Chen Z, Lawlor KE, Corbett AJ. RIPK3 controls MAIT cell accumulation during development but not during infection. Cell Death Dis 2023; 14:111. [PMID: 36774342 PMCID: PMC9922319 DOI: 10.1038/s41419-023-05619-0] [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: 08/07/2022] [Revised: 01/25/2023] [Accepted: 01/25/2023] [Indexed: 02/13/2023]
Abstract
Cell death mechanisms in T lymphocytes vary according to their developmental stage, cell subset and activation status. The cell death control mechanisms of mucosal-associated invariant T (MAIT) cells, a specialized T cell population, are largely unknown. Here we report that MAIT cells express key necroptotic machinery; receptor-interacting protein kinase 3 (RIPK3) and mixed lineage kinase domain-like (MLKL) protein, in abundance. Despite this, we discovered that the loss of RIPK3, but not necroptotic effector MLKL or apoptotic caspase-8, specifically increased MAIT cell abundance at steady-state in the thymus, spleen, liver and lungs, in a cell-intrinsic manner. In contrast, over the course of infection with Francisella tularensis, RIPK3 deficiency did not impact the magnitude of the expansion nor contraction of MAIT cell pools. These findings suggest that, distinct from conventional T cells, the accumulation of MAIT cells is restrained by RIPK3 signalling, likely prior to thymic egress, in a manner independent of canonical apoptotic and necroptotic cell death pathways.
Collapse
Affiliation(s)
- Timothy Patton
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Zhe Zhao
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Xin Yi Lim
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Eleanor Eddy
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Huimeng Wang
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Adam G Nelson
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Bronte Ennis
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sidonia B G Eckle
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Michael N T Souter
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Troi J Pediongco
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Hui-Fern Koay
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Jian-Guo Zhang
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Tirta M Djajawi
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Cynthia Louis
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Najoua Lalaoui
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Nicolas Jacquelot
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andrew M Lew
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Daniel G Pellicci
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
- Murdoch Children's Research Institute, Royal Children's Hospital Parkville, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - James McCluskey
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Yifan Zhan
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Department of Drug Discovery, Shanghai Huaota Biopharm, Shanghai, China
| | - Zhenjun Chen
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Kate E Lawlor
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Alexandra J Corbett
- Department of Immunology and Microbiology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia.
| |
Collapse
|
44
|
Treiner E. Mucosal-associated invariant T cells in hematological malignancies: Current knowledge, pending questions. Front Immunol 2023; 14:1160943. [PMID: 37020559 PMCID: PMC10067713 DOI: 10.3389/fimmu.2023.1160943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 03/06/2023] [Indexed: 04/07/2023] Open
Abstract
Non-classical HLA restricted T cell subsets such as γδ T and NK-T cells are showing promises for immune-based therapy of hematological malignancies. Mucosal-Associated Invariant T cells (MAIT) belong to this family of innate-like T cell subsets and are the focus of many studies on infectious diseases, owing to their unusual recognition of bacterial/fungal metabolites. Their ability to produce type 1 cytokines (IFNγ, TNFα) as well as cytotoxic effector molecules endows them with potential anti-tumor functions. However, their contribution to tumor surveillance in solid cancers is unclear, and only few studies have specifically focused on MAIT cells in blood cancers. In this review, we wish to recapitulate our current knowledge on MAIT cells biology in hematological neoplasms, at diagnosis and/or during treatment, as well as tentative approaches to target them as therapeutic tools. We also wish to take this opportunity to briefly elaborate on what we think are important question to address in this field, as well as potential limitations to overcome in order to make MAIT cells the basis of future, novel therapies for hematological cancers.
Collapse
Affiliation(s)
- Emmanuel Treiner
- Infinity, Inserm UMR1291, Toulouse, France
- University Toulouse 3, Toulouse, France
- Laboratory of Immunology, Toulouse University Hospital, Toulouse, France
- *Correspondence: Emmanuel Treiner,
| |
Collapse
|
45
|
Mullaney JA, Roy NC, Halliday C, Young W, Altermann E, Kruger MC, Dilger RN, McNabb WC. Effects of early postnatal life nutritional interventions on immune-microbiome interactions in the gastrointestinal tract and implications for brain development and function. Front Microbiol 2022; 13:960492. [PMID: 36504799 PMCID: PMC9726769 DOI: 10.3389/fmicb.2022.960492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 11/01/2022] [Indexed: 11/24/2022] Open
Abstract
The gastrointestinal (GI) microbiota has co-evolved with the host in an intricate relationship for mutual benefit, however, inappropriate development of this relationship can have detrimental effects. The developing GI microbiota plays a vital role during the first 1,000 days of postnatal life, during which occurs parallel development and maturation of the GI tract, immune system, and brain. Several factors such as mode of delivery, gestational age at birth, exposure to antibiotics, host genetics, and nutrition affect the establishment and resultant composition of the GI microbiota, and therefore play a role in shaping host development. Nutrition during the first 1,000 days is considered to have the most potential in shaping microbiota structure and function, influencing its interactions with the immune system in the GI tract and consequent impact on brain development. The importance of the microbiota-GI-brain (MGB) axis is also increasingly recognized for its importance in these developmental changes. This narrative review focuses on the importance of the GI microbiota and the impact of nutrition on MGB axis during the immune system and brain developmental period in early postnatal life of infants.
Collapse
Affiliation(s)
- Jane A. Mullaney
- Riddet Institute, Massey University, Palmerston North, New Zealand,AgResearch, Palmerston North, New Zealand,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nicole C. Roy
- Riddet Institute, Massey University, Palmerston North, New Zealand,High-Value Nutrition National Science Challenge, Auckland, New Zealand,Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Christine Halliday
- Riddet Institute, Massey University, Palmerston North, New Zealand,AgResearch, Palmerston North, New Zealand,School of Food and Advanced Technology, College of Sciences, Massey University, Palmerston North, New Zealand
| | - Wayne Young
- Riddet Institute, Massey University, Palmerston North, New Zealand,AgResearch, Palmerston North, New Zealand,High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Eric Altermann
- Riddet Institute, Massey University, Palmerston North, New Zealand,High-Value Nutrition National Science Challenge, Auckland, New Zealand,School of Veterinary Science, Massey University, Palmerston North, New Zealand
| | - Marlena C. Kruger
- School of Health Sciences, College of Health, Massey University, Palmerston North, New Zealand
| | - Ryan N. Dilger
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Warren C. McNabb
- Riddet Institute, Massey University, Palmerston North, New Zealand,High-Value Nutrition National Science Challenge, Auckland, New Zealand,*Correspondence: Warren C. McNabb,
| |
Collapse
|
46
|
Abstract
Mucosal Associated Invariant T cells (MAIT) exert potent antimicrobial activity through direct recognition of metabolite-MR1 complexes and indirect activation by inflammatory cytokines. Additionally, via licensing of antigen presenting cells, MAIT cells orchestrate humoral and cellular adaptive immunity. Our recent understanding of molecular mechanisms of MAIT cell activation, and of the signals required to differentiate them in polarised subsets, pave the way for harnessing their functionality through small molecules or adoptive cell therapy.
Collapse
Affiliation(s)
- Mariolina Salio
- Immunocore LTD, 92 Park Drive, Milton Park, Abingdon, Oxfordshire OX14 4RY, United Kingdom.
| |
Collapse
|
47
|
Abstract
Mucosal associated invariant T (MAIT) cells are innate T cells that recognize bacterial metabolites and secrete cytokines and cytolytic enzymes to destroy infected target cells. This makes MAIT cells promising targets for immunotherapy to combat bacterial infections. Here, we analyzed the effects of an immunotherapeutic agent, the IL-15 superagonist N-803, on MAIT cell activation, trafficking, and cytolytic function in macaques. We found that N-803 could activate MAIT cells in vitro and increase their ability to produce IFN-γ in response to bacterial stimulation. To expand upon this, we examined the phenotypes and functions of MAIT cells present in samples collected from PBMC, airways (bronchoalveolar lavage [BAL] fluid), and lymph nodes (LN) from rhesus macaques that were treated in vivo with N-803. N-803 treatment led to a transient 6 to 7-fold decrease in the total number of MAIT cells in the peripheral blood, relative to pre N-803 time points. Concurrent with the decrease in cells in the peripheral blood, we observed a rapid decline in the frequency of CXCR3+CCR6+ MAITs. This corresponded with an increase in the frequency of CCR6+ MAITs in the BAL fluid, and higher frequencies of ki-67+ and granzyme B+ MAITs in the blood, LN, and BAL fluid. Finally, N-803 improved the ability of MAIT cells collected from PBMC and airways to produce IFN-γ in response to bacterial stimulation. Overall, N-803 shows the potential to transiently alter the phenotypes and functions of MAIT cells, which could be combined with other strategies to combat bacterial infections.
Collapse
|
48
|
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: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [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
|
49
|
Meermeier EW, Zheng CL, Tran JG, Soma S, Worley AH, Weiss DI, Modlin RL, Swarbrick G, Karamooz E, Khuzwayo S, Wong EB, Gold MC, Lewinsohn DM. Human lung-resident mucosal-associated invariant T cells are abundant, express antimicrobial proteins, and are cytokine responsive. Commun Biol 2022; 5:942. [PMID: 36085311 PMCID: PMC9463188 DOI: 10.1038/s42003-022-03823-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 08/09/2022] [Indexed: 12/02/2022] Open
Abstract
Mucosal-associated Invariant T (MAIT) cells are an innate-like T cell subset that recognize a broad array of microbial pathogens, including respiratory pathogens. Here we investigate the transcriptional profile of MAIT cells localized to the human lung, and postulate that MAIT cells may play a role in maintaining homeostasis at this mucosal barrier. Using the MR1/5-OP-RU tetramer, we identified MAIT cells and non-MAIT CD8+ T cells in lung tissue not suitable for transplant from human donors. We used RNA-sequencing of MAIT cells compared to non-MAIT CD8+ T cells to define the transcriptome of MAIT cells in the human lung. We show that, as a population, lung MAIT cells are polycytotoxic, secrete the directly antimicrobial molecule IL-26, express genes associated with persistence, and selectively express cytokine and chemokine- related molecules distinct from other lung-resident CD8+ T cells, such as interferon-γ- and IL-12- receptors. These data highlight MAIT cells' predisposition to rapid pro-inflammatory cytokine responsiveness and antimicrobial mechanisms in human lung tissue, concordant with findings of blood-derived counterparts, and support a function for MAIT cells as early sensors in the defense of respiratory barrier function.
Collapse
Affiliation(s)
- Erin W Meermeier
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
- Department of Medicine, Division of Hematology/Oncology, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Christina L Zheng
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Jessica G Tran
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - Shogo Soma
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Aneta H Worley
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - David I Weiss
- David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Robert L Modlin
- Division of Dermatology, Department of Medicine, David Geffen School of Medicine at University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Gwendolyn Swarbrick
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - Elham Karamooz
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - Sharon Khuzwayo
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Emily B Wong
- Africa Health Research Institute, Durban, South Africa
- School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
- Division of Infectious Diseases, Massachusetts General Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Division of Infection and Immunity, University College London, London, UK
| | - Marielle C Gold
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA
- VA Portland Health Care System, Portland, OR, 97239, USA
| | - David M Lewinsohn
- Department of Pulmonary and Critical Care Medicine, Oregon Health & Science University, Portland, OR, 97239, USA.
- VA Portland Health Care System, Portland, OR, 97239, USA.
| |
Collapse
|
50
|
Jensen O, Trivedi S, Li K, Aubé J, Hale JS, Ryan ET, Leung DT. Use of a MAIT-Activating Ligand, 5-OP-RU, as a Mucosal Adjuvant in a Murine Model of Vibrio cholerae O1 Vaccination. Pathog Immun 2022; 7:122-144. [PMID: 36072570 PMCID: PMC9438945 DOI: 10.20411/pai.v7i1.525] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 07/14/2022] [Indexed: 11/23/2022] Open
Abstract
Background Mucosal-associated invariant T (MAIT) cells are innate-like T cells enriched in the mucosa with capacity for B-cell help. We hypothesize that targeting MAIT cells, using a MAIT-activating ligand as an adjuvant, could improve mucosal vaccine responses to bacterial pathogens such as Vibrio cholerae. Methods We utilized murine models of V. cholerae vaccination to test the adjuvant potential of the MAIT-activating ligand, 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil (5-OP-RU). We measured V. cholerae-specific antibody and antibody-secreting cell responses and used flow cytometry to examine MAIT-cell and B-cell phenotype, in blood, bronchoalveolar lavage fluid (BALF), and mucosal tissues, following intranasal vaccination with live V. cholerae O1 or a V. cholerae O1 polysaccharide conjugate vaccine. Results We report significant expansion of MAIT cells in the lungs (P < 0.001) and BALF (P < 0.001) of 5-OP-RU treated mice, and higher mucosal (BALF, P = 0.045) but not systemic (serum, P = 0.21) V. cholerae O-specific-polysaccharide IgG responses in our conjugate vaccine model when adjuvanted with low-dose 5-OP-RU. In contrast, despite significant MAIT cell expansion, no significant differences in V. cholerae-specific humoral responses were found in our live V. cholerae vaccination model. Conclusions Using a murine model, we demonstrate the potential, as well as the limitations, of targeting MAIT cells to improve antibody responses to mucosal cholera vaccines. Our study highlights the need for future research optimizing MAIT-cell targeting for improving mucosal vaccines.
Collapse
Affiliation(s)
- Owen Jensen
- Division of Infectious Diseases, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Shubhanshi Trivedi
- Division of Infectious Diseases, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah
| | - Kelin Li
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Jeffrey Aubé
- Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - J. Scott Hale
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah
| | - Edward T. Ryan
- Division of Infectious Disease, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Immunology and Infectious diseases, Harvard School of Public Health, Boston, Massachusetts
| | - Daniel T. Leung
- Division of Infectious Diseases, Department of Internal Medicine, University of Utah School of Medicine, Salt Lake City, Utah
- Division of Microbiology & Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|