1
|
Wang H, Kc P, Zhang K, Materne C, Lhomme M, Galier S, Ichou F, Neves C, Lehuen A, Haas JT, Salem JE, Guerin M, Lesnik P. MAIT Cells Promote Cholesterol Excretion Pathways Mitigating Atherosclerosis. Circ Res 2025; 136:968-981. [PMID: 40135347 DOI: 10.1161/circresaha.124.325841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/07/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025]
Abstract
BACKGROUND Previous clinical studies have indicated reduced circulating mucosal-associated invariant T (MAIT) cells in individuals with coronary artery disease. However, the precise role and underlying mechanisms of MAIT cells in this context remain unclear. Immune homeostasis plays a pivotal role in the development of atherosclerosis. This study explores the impact of MAIT cells on atherosclerosis. METHODS Vα19+/- Ldlr-/- mice, characterized by a high MAIT cell frequency, and MAIT cell deficient MR1-/- (major histocompatibility complex-related molecule 1) Ldlr-/- mice and their respective controls were used. Starting at 6 weeks of age, mice were subjected to a 1% cholesterol diet for 16 weeks. Additionally, the study analyzed circulating MAIT cell frequency and cholesterol levels in 68 patients with hypercholesterolemia. RESULTS In Vα19+/- Ldlr-/- mice, increased MAIT cells demonstrated a protective effect against atherosclerosis by reducing VLDL-C (very-low-density lipoprotein cholesterol) levels through heightened cholesterol excretion. This effect was accompanied by elevated jejunal ABCB1a, ABCG5, and ABCG8 expression, mediated by augmented levels of Liver X receptor transcription and activation, likely through intestinal IL-22 (interleukin-22) signaling. Conversely, cholesterol reduction mediated by intestinal cholesterol excretion was blocked by inhibition of MAIT cells. Moreover, MAIT cell-deficient MR1-/- Ldlr-/- mice exhibited elevated total cholesterol levels and increased atherosclerotic lesions. In patients with hypercholesterolemia, circulating MAIT cell frequency displayed negative correlations with VLDL-C levels and positive correlations with HDL-C (high-density lipoprotein cholesterol) levels. CONCLUSIONS Our findings demonstrate a new mechanism for plasma VLDL-C clearance by MAIT cell-mediated cholesterol excretion. The results provide further evidence that immunity is involved in cholesterol homeostasis. Targeting intestinal immunity to regulate cholesterol homeostasis holds promise as a new cholesterol-lowering modality to prevent atherosclerotic cardiovascular disease.
Collapse
Affiliation(s)
- Hua Wang
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Pukar Kc
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Kaidi Zhang
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Clément Materne
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Marie Lhomme
- Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), ICAN OMICS, Paris, France (M.L., F.I.)
| | - Sophie Galier
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Farid Ichou
- Foundation for Innovation in Cardiometabolism and Nutrition (ICAN), ICAN OMICS, Paris, France (M.L., F.I.)
| | - Carolina Neves
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Agnès Lehuen
- Université Paris Cité, Institut Cochin, Inserm U1016, Centre National de la Recherche Scientifique UMR 8104, Inflamex Laboratory, Paris, France (A.L.)
| | - Joel T Haas
- Université de Lille, Inserm, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, Lille, France (J.T.H.)
| | - Joe-Elie Salem
- INSERM, CIC-1901 Paris-Est, Assistance Publique - Hôpitaux de Paris, Pitié-Salpêtrière Hospital, Department of Pharmacology, Paris, France (J.-E.S.)
| | - Maryse Guerin
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| | - Philippe Lesnik
- Sorbonne Université, National Institute of Health and Medical Research (INSERM) U1166, Paris, France (H.W., P.K.C., K.Z., C.M., S.G., C.N., M.G., P.L.)
| |
Collapse
|
2
|
Ronca V, Gerussi A, Collins P, Parente A, Oo YH, Invernizzi P. The liver as a central "hub" of the immune system: pathophysiological implications. Physiol Rev 2025; 105:493-539. [PMID: 39297676 DOI: 10.1152/physrev.00004.2023] [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: 05/18/2023] [Revised: 09/05/2024] [Accepted: 09/08/2024] [Indexed: 01/16/2025] Open
Abstract
The purpose of this review is to describe the immune function of the liver, guiding the reader from the homeostatic tolerogenic status to the aberrant activation demonstrated in chronic liver disease. An extensive description of the pathways behind the inflammatory modulation of the healthy liver will be provided focusing on the complex immune cell network residing within the liver. The limit of tolerance will be presented in the context of organ transplantation, seizing the limits of homeostatic mechanisms that fail in accepting the graft, progressing eventually toward rejection. The triggers and mechanisms behind chronic activation in metabolic liver conditions and viral hepatitis will be discussed. The last part of the review will be dedicated to one of the greatest paradoxes for a tolerogenic organ, developing autoimmunity. Through the description of the three most common autoimmune liver diseases, the autoimmune reaction against hepatocytes and biliary epithelial cells will be dissected.
Collapse
Affiliation(s)
- Vincenzo Ronca
- Centre for Liver and Gastro Research and National Institute for Health and Care Research (NIHR) Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Liver Unit, Queen Elizabeth Hospital University Hospital Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Rare Diseases, European Reference Network Centre-Rare Liver, Birmingham, United Kingdom
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Humanitas Research Hospital, Milan, Italy
| | - Alessio Gerussi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Paul Collins
- VIB-UGent Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Faculty of Sciences, Ghent University, Ghent, Belgium
| | - Alessandro Parente
- Liver Unit, Queen Elizabeth Hospital University Hospital Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Institute of Liver Studies, King's College Hospital NHS Foundation Trust, London, United Kingdom
| | - Ye Htun Oo
- Centre for Liver and Gastro Research and National Institute for Health and Care Research (NIHR) Biomedical Research Centre, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, United Kingdom
- Liver Unit, Queen Elizabeth Hospital University Hospital Birmingham National Health Service (NHS) Foundation Trust, Birmingham, United Kingdom
- Centre for Rare Diseases, European Reference Network Centre-Rare Liver, Birmingham, United Kingdom
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, European Reference Network on Hepatological Diseases (ERN RARE-LIVER), IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
- Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
3
|
Salou M, Paiva RA, Lantz O. Development and Functions of MAIT Cells. Annu Rev Immunol 2025; 43:253-283. [PMID: 39879553 DOI: 10.1146/annurev-immunol-082323-025943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Mucosal-associated invariant T (MAIT) cells are evolutionarily conserved T cells that recognize microbial metabolites. They are abundant in humans and conserved during mammalian evolution, which suggests that they have important nonredundant functions. In this article, we discuss the evolutionary conservation of MAIT cells and describe their original developmental process. MAIT cells exert a wide variety of effector functions, from killing infected cells and promoting inflammation to repairing tissues. We provide insights into these functions and discuss how they result from the context of stimulation encountered by MAIT cells in different tissues and pathological settings. We describe how MAIT cell numbers and features are modified in disease states, focusing mainly on in vivo models. Lastly, we discuss emerging strategies to manipulate MAIT cells for therapeutic purposes.
Collapse
Affiliation(s)
- Marion Salou
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
| | - Rafael A Paiva
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
| | - Olivier Lantz
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
- Laboratoire d'Immunologie Clinique, Institut Curie, Paris, France
- Centre d'Investigation Clinique en Biothérapie, Gustave-Roussy and Institut Curie (CIC-BT1428), Paris, France
| |
Collapse
|
4
|
Liu R, Pei L, Liu J, Peng L, Guo C, Li Y, Duan Z, Du Y, Shang D, Li S, Zhang Y, Song B. Circulating mucosal-associated invariant T cells predict early neurological deterioration in acute ischemic stroke. J Stroke Cerebrovasc Dis 2025; 34:108301. [PMID: 40157657 DOI: 10.1016/j.jstrokecerebrovasdis.2025.108301] [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: 10/22/2024] [Revised: 03/24/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Mucosal-associated invariant T (MAIT) cells are innate-like T cells that rapidly produce cytokines such as tumor necrosis factor-α (TNF-α) and interleukin-17 (IL-17) upon activation. The immune response is crucial in stroke-related injury. However, few studies have investigated the role of MAIT cells in ischemic brain injury. This study assessed the predictive value of circulating MAIT cells in acute ischemic stroke (AIS) and early neurological deterioration (END). METHODS We prospectively and continuously enrolled AIS patients within 72 h of stroke onset and included controls. END was defined as a ≥ 2-point increase in the National Institutes of Health Stroke Scale score within the first 72 h. Receiver operating characteristic curves were used to evaluate the predictive value of MAIT cells for END. RESULTS This study included 188 AIS patients and 135 controls, with 50 (26.6 %) AIS patients experiencing END. After adjusting for all potential confounders, circulating MAIT cell frequencies were lower in AIS patients than in controls (odds ratio [OR]: 0.83, 95 % confidence interval [CI]: 0.70-0.97, P = 0.02). IL-17 and TNF-α levels were significantly higher in AIS patients and negatively correlated with MAIT cell frequencies (R = -0.26, P < 0.05; R = -0.19, P < 0.05). Multivariate logistic regression analysis revealed that MAIT cell frequencies were lower in patients with END compared to those without END (OR: 0.74, 95 % CI: 0.55-0.96, P = 0.03). The area under the curve for MAIT cells in END prediction was 0.641 (95 % CI: 0.548-0.725, P < 0.05). CONCLUSIONS MAIT cell frequency was reduced in AIS patients and may serve as a predictive marker for END. Modulating these cells could be a novel AIS treatment strategy.
Collapse
Affiliation(s)
- Ruihua Liu
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Lulu Pei
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jiacheng Liu
- Department of Neurology, Xinxiang Medical University, Xinxiang, China
| | - Liang Peng
- Department of Laboratory, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Cairu Guo
- Department of Central Laboratory, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Yan Li
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Zhihui Duan
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Yanjiao Du
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Dandan Shang
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Shao Li
- Department of Neurology, Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, China
| | - Yunting Zhang
- Department of Neurology, Xinxiang Medical University, Xinxiang, China
| | - Bo Song
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
5
|
Suckling RJ, Pamukcu C, Simmons RA, Fonseca D, Grant E, Harrison R, Shaikh SA, Khanolkar RC, Ghadbane H, Creese A, Hock M, Gligoris TG, Lepore M, Karuppiah V, Salio M. Molecular basis underpinning MR1 allomorph recognition by an MR1-restricted T cell receptor. Front Immunol 2025; 16:1547664. [PMID: 40207221 PMCID: PMC11979126 DOI: 10.3389/fimmu.2025.1547664] [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: 12/18/2024] [Accepted: 02/21/2025] [Indexed: 04/11/2025] Open
Abstract
Introduction The MHC-class-I-related molecule MR1 presents small metabolites of microbial and self-origin to T cells bearing semi-invariant or variant T cell receptors. One such T cell receptor, MC.7.G5, was previously shown to confer broad MR1-restricted reactivity to tumor cells but not normal cells, sparking interest in the development of non-MHC-restricted immunotherapy approaches. Methods/Results Here we provide cellular, biophysical, and crystallographic evidence that the MC.7.G5 TCR does not have pan-cancer specificity but is restricted to a rare allomorph of MR1, bearing the R9H mutation. Discussion Our results underscore the importance of in-depth characterization of MR1-reactive TCRs against targets expressing the full repertoire of MR1 allomorphs.
Collapse
|
6
|
Stern L, Emanuel Z, Traves R, Willis K, Purohit SK, Samer C, Mak JYW, Fairlie DP, Tscharke DC, Corbett AJ, Abendroth A, Slobedman B. Herpes simplex virus type 1 impairs mucosal-associated invariant T cells. mBio 2025:e0388724. [PMID: 40135871 DOI: 10.1128/mbio.03887-24] [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: 01/13/2025] [Accepted: 02/04/2025] [Indexed: 03/27/2025] Open
Abstract
Herpes simplex virus type 1 (HSV-1) is a highly successful pathogen that infects mucosal sites and adopts an arsenal of strategies to manipulate host immunity. Mucosal-associated invariant T (MAIT) cells are abundant innate-like T lymphocytes that recognize bacterial and fungal-derived vitamin B-related metabolites presented by major histocompatibility complex class I-related protein 1 (MR1). MAIT cells can also be activated in an MR1-independent manner via cytokine stimulation, predominantly by IL-12 and IL-18. MAIT cell alterations have been identified as being associated with a number of viral infections, but direct interactions between viruses and MAIT cells are poorly understood. It is unknown whether HSV-1 can infect MAIT cells and modulate their functions. Here, we show that HSV-1 can infect primary human MAIT cells, including CD4±/CD8± and CD56± MAIT cell subpopulations. Furthermore, HSV-1 infection profoundly inhibits the functional capacity of MAIT cells to respond to T cell receptor (TCR)-dependent stimulation by the MAIT cell activating ligand 5-(2-oxopropylideneamino)-6-D-ribitylaminouracil (5-OP-RU) and to cytokine stimulation by IL-12/IL-18. HSV-1-infected MAIT cells display reduced cytotoxic potential, diminished synthesis of effector cytokines, and decreased expression of key cytokine receptors including IL-18R. In addition, MAIT cells exposed to HSV-1-infected fibroblasts but which remained uninfected (viral GFP-negative) also exhibit a suppressed effector response to TCR-dependent stimulation. The functional suppression of HSV-1-exposed MAIT cells was not mediated by a soluble factor within the supernatant, suggesting direct contact of MAIT cells with HSV-1-infected fibroblasts is required. Overall, this study reveals that HSV-1 can infect MAIT cells and substantially impair MAIT cell effector functions. IMPORTANCE Mucosal-associated invariant T cells (MAIT cells) are "unconventional" immune cells that are becoming increasingly appreciated to play important roles in a variety of viral infections. Herpes simplex virus (HSV) causes significant human disease and is a master manipulator of multiple immune functions, but how this virus may control MAIT cells is poorly understood. We discovered that HSV can infect human MAIT cells and impair their functional capacity and also show that MAIT cells exposed to HSV, but which do not show evidence of infection, are similarly impaired. This study therefore identifies an additional immunomodulatory function of HSV.
Collapse
Affiliation(s)
- Lauren Stern
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Zoe Emanuel
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Renee Traves
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Katherine Willis
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Shivam K Purohit
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Carolyn Samer
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Jeffrey Y W Mak
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - David P Fairlie
- ARC Centre of Excellence for Innovations in Peptide and Protein Science, Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - David C Tscharke
- John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Alexandra J Corbett
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Allison Abendroth
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Barry Slobedman
- Infection, Immunity, and Inflammation, School of Medical Sciences, Faculty of Medicine and Health, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
7
|
Wang S, Liu Y, Zou X, Pan M, Wan Q, Chu X. Exploring the pathogenesis of RA through the gut-articular axis-dysbiosis a potential factor. Clin Anat 2025; 38:134-145. [PMID: 39189295 DOI: 10.1002/ca.24215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 08/04/2024] [Indexed: 08/28/2024]
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease with a complex etiology. It has been suggested that the pathogenesis of RA begins in the mucosa and then transitions to the joints when many factors interact, including microbial dysbiosis, inflammatory responses, and immune abnormalities at the mucosal site. Data from RA animals and patients suggest there are changes in the mucosal microflora before the onset of RA, and that dysbiosis of the mucosal ecology continues to play a role in the development of arthritis. Microbial dysbiosis of the mucosa reduces the normal barrier function of the intestinal tract, promotes inflammatory reactions in the mucosal areas of the intestines, and then activates the intestinal immune cells abnormally to produce a large number of auto-reactive antibodies that exacerbate arthritis. Current findings do not clarify whether dysbiosis is only a potential trigger for the development of RA. If it is possible to intervene in such microbial changes before the onset of RA, could the clinical symptoms of arthritis be prevented or reduced? Finding new ways to regulate gut flora composition to maintain gut barrier function is an ongoing challenge for the prevention and treatment of RA.
Collapse
Affiliation(s)
- Shuai Wang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Yue Liu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Xingyu Zou
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Mengjun Pan
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qing Wan
- Tongling Institutes for Food and Drug Control, Tongling, China
| | - Xiaoqin Chu
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, China
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, China
- Engineering Technology Research Center of Modern Pharmaceutical Preparation, Anhui Province, China
| |
Collapse
|
8
|
Zhang X, Li S, Lason W, Greco M, Klenerman P, Hinks TSC. MAIT cells protect against sterile lung injury. Cell Rep 2025; 44:115275. [PMID: 39918959 DOI: 10.1016/j.celrep.2025.115275] [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: 09/12/2024] [Revised: 12/06/2024] [Accepted: 01/16/2025] [Indexed: 02/09/2025] Open
Abstract
Mucosal-associated invariant T (MAIT) cells, the most abundant unconventional T cells in the lung, can exhibit a wide range of functional responses to different triggers via their T cell receptor (TCR) and/or cytokines. Their role, especially in sterile lung injury, is unknown. Using single-cell RNA sequencing (scRNA-seq), spectral analysis, and adoptive transfer in a bleomycin-induced sterile lung injury, we found that bleomycin activates murine pulmonary MAIT cells and is associated with a protective role against bleomycin-induced lung injury. MAIT cells drive the accumulation of type 1 conventional dendritic cells (cDC1s), limiting tissue damage in a DNGR-1-dependent manner. Human scRNA-seq data revealed that MAIT cells were activated, with increased cDC populations in idiopathic pulmonary fibrosis patients. Thus, MAIT cells enhance defense against sterile lung injury by fostering cDC1-driven anti-fibrotic pathways.
Collapse
Affiliation(s)
- Xiawei Zhang
- Respiratory Medicine Unit, Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Shuailin Li
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford OX3 7DQ, UK
| | - Wojciech Lason
- Respiratory Medicine Unit, Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK
| | - Maria Greco
- MRC Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research and Translational Gastroenterology Unit, Nuffield Department of Clinical Medicine, University of Oxford, Oxford OX1 3SY, UK
| | - Timothy S C Hinks
- Respiratory Medicine Unit, Experimental Medicine Division, Nuffield Department of Medicine, University of Oxford, John Radcliffe Hospital, Oxford OX3 9DU, UK.
| |
Collapse
|
9
|
Camard L, Stephen T, Yahia-Cherbal H, Guillemot V, Mella S, Baillet V, Lopez-Maestre H, Capocefalo D, Cantini L, Leloup C, Marsande J, Garro K, Sienes Bailo J, Dangien A, Pietrosemoli N, Hasan M, Wang H, Eckle SB, Fourie AM, Greving C, Joyce-Shaikh B, Parker R, Cua DJ, Bianchi E, Rogge L. IL-23 tunes inflammatory functions of human mucosal-associated invariant T cells. iScience 2025; 28:111898. [PMID: 40008359 PMCID: PMC11850163 DOI: 10.1016/j.isci.2025.111898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 11/15/2024] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
IL-23 signaling plays a key role in the pathogenesis of chronic inflammatory and infectious diseases, yet the cellular targets and signaling pathways affected by this cytokine remain poorly understood. We show that IL-23 receptors are expressed on the large majority of human mucosal-associated invariant T (MAIT), but not of conventional T cells. Protein and transcriptional profiling at the population and single cell level demonstrates that stimulation with IL-23 or the structurally related cytokine IL-12 drives distinct functional profiles, revealing a high level of plasticity of MAIT cells. IL-23, in particular, affects key molecules and pathways related to autoimmunity and cytotoxic functions. Integrated analysis of transcriptomes and chromatin accessibility, supported by CRISPR-Cas9 mediated deletion, shows that AP-1 transcription factors constitute a key regulatory node of the IL-23 pathway in MAIT cells. In conclusion, our findings indicate that MAIT cells are key mediators of IL-23 functions in immunity to infections and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Laetitia Camard
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Tharshana Stephen
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
- scBiomarkers, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Hanane Yahia-Cherbal
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Vincent Guillemot
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Sébastien Mella
- scBiomarkers, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Victoire Baillet
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Hélène Lopez-Maestre
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Daniele Capocefalo
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics Group, 75015 Paris, France
| | - Laura Cantini
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics Group, 75015 Paris, France
| | - Claire Leloup
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Julie Marsande
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Katherine Garro
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Juan Sienes Bailo
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Ambre Dangien
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
- Department of Dermatology, Hôpital Cochin, AP-HP, AP-HP Centre-Université de Paris, 75014 Paris, France
| | - Natalia Pietrosemoli
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Milena Hasan
- scBiomarkers, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Huimeng Wang
- Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, 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 3000, Australia
| | - Anne M. Fourie
- Janssen Research & Development, LLC, San Diego, CA 92121, USA
| | - Carrie Greving
- Janssen Research & Development, LLC, San Diego, CA 92121, USA
| | | | - Raphaelle Parker
- Janssen Research & Development, Janssen-Cilag, 92130 Issy les Moulineaux, France
| | - Daniel J. Cua
- Janssen Research & Development, LLC, Spring House, PA 19002, USA
| | - Elisabetta Bianchi
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Lars Rogge
- Immunoregulation Unit, Department of Immunology, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| |
Collapse
|
10
|
Loureiro JP, Vacchini A, Berloffa G, Devan J, Schaefer V, Nosi V, Colombo R, Beshirova A, Montanelli G, Meyer B, Sharpe T, Chancellor A, Recher M, Mori L, De Libero G. Recognition of MR1-antigen complexes by TCR Vγ9Vδ2. Front Immunol 2025; 16:1519128. [PMID: 40040716 PMCID: PMC11876030 DOI: 10.3389/fimmu.2025.1519128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/30/2025] [Indexed: 03/06/2025] Open
Abstract
The TCR-mediated activation of T cells expressing the TCR Vγ9Vδ2 relies on an innate-like mechanism involving the butyrophilin 3A1, 3A2 and 2A1 molecules and phospho-antigens, without the participation of classical antigen-presenting molecules. Whether TCR Vγ9Vδ2 cells also recognize complexes composed of antigens and antigen-presenting molecules in an adaptive-like manner is unknown. Here, we identify MR1-autoreactive cells expressing the TCR Vγ9Vδ2. This MR1-restricted response is antigen- and CDR3δ-dependent and butyrophilin-independent. TCR gene transfer reconstitutes MR1-antigen recognition, and engineered TCR Vγ9Vδ2 tetramers interact with soluble MR1-antigen complexes in an antigen-dependent manner. These cells are present in healthy individuals with low frequency and are mostly CD8+ or CD4-CD8 double negative. We also describe a patient with autoimmune symptoms and TCR γδ lymphocytosis in which ~10% of circulating T cells are MR1-self-reactive and express a TCR Vγ9Vδ2. These cells release pro-inflammatory cytokines, suggesting a possible participation in disease pathogenesis. Thus, MR1-self-antigen complexes can interact with some TCRs Vγ9Vδ2, promoting full cell activation and potentially contributing to diseases.
Collapse
Affiliation(s)
- José Pedro Loureiro
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Alessandro Vacchini
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Giuliano Berloffa
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Jan Devan
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Verena Schaefer
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Vladimir Nosi
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Rodrigo Colombo
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Aisha Beshirova
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Giulia Montanelli
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Benedikt Meyer
- Immunodeficiency Laboratory, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | | | - Andrew Chancellor
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Mike Recher
- Immunodeficiency Laboratory, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Lucia Mori
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Gennaro De Libero
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| |
Collapse
|
11
|
Sun Y, Sen S, Parmar R, Arakawa-Hoyt J, Cappelletti M, Rossetti M, Gjertson DW, Sigdel TK, Sarwal MM, Schaenman JM, Bunnapradist S, Lanier LL, Pickering H, Reed EF. Cytotoxic KLRG1+ IL-7R- effector CD8+ T cells distinguish kidney transplant recipients controlling cytomegalovirus reactivation. Front Immunol 2025; 16:1542531. [PMID: 40028342 PMCID: PMC11868092 DOI: 10.3389/fimmu.2025.1542531] [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: 12/10/2024] [Accepted: 01/27/2025] [Indexed: 03/05/2025] Open
Abstract
Introduction Cytomegalovirus (CMV) viremia remains a major contributor to clinical complications in solid organ transplant (SOT) patients, including organ injury, morbidity and mortality. Given their critical role in antiviral defense, CD8+ T cells are essential for protective immunity against CMV. Methods Using single-cell RNA sequencing, we investigated the transcriptional signatures and developmental lineages of CD8+ T cells in eight immunosuppressed kidney transplant recipients (KTRs) who received organs from CMV-seropositive donors. Results were validated in a cohort of 62 KTRs using immunophenotyping. Results Our data revealed a significant influence of CMV serostatus on transcriptional variance of CD8+ memory T cells, associating with the first principal component from a global analysis of CD8+ T cells (p =0.0406), forming a continuum with five principal differentiation trajectories driven by CMV primary infection or reactivation. Following CMV primary infection, CD8+ T cells were hallmarked by restrained effector-memory differentiation. CD8+ T cells during CMV reactivation diverged non-linearly into senescent-like cells with signatures of arrested cell cycle, diminished translational activity and downregulated ZNF683 and longitudinally expanding effector cells with robust cytotoxic potential and upregulated ZNF683, acting as a reservoir for long-lived effector cells supporting long-term protection. Notably, CD28lo KLRG1hi IL-7R (CD127)lo HLA-DRhi CD8+ T cells present prior to the detection of viremia in CMV-seropositive patients emerged as a key feature distinguishing patients who did or did not undergo CMV reactivation after prophylaxis discontinuation (p =0.0163). Frequencies of these cells were also positively correlated with CMV-stimulated secretion of IFN-γ (p =0.0494), TNF-α (p =0.0358), MIP-1α (p =0.0262), MIP-1β (p =0.0043). Discussion These results provide insights into the transcriptional regulation that influences the generation of CD8+ T cell immunity to CMV and may inform strategics for monitoring host immune response to CMV to better identify and introduce therapeutic intervention to patients at risk of developing clinically significant CMV viremia.
Collapse
Affiliation(s)
- Yumeng Sun
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Subha Sen
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rajesh Parmar
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Janice Arakawa-Hoyt
- Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States
| | - Monica Cappelletti
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Maura Rossetti
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - David W. Gjertson
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Tara K. Sigdel
- Department of Surgery, Division of Multi Organ Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Minnie M. Sarwal
- Department of Surgery, Division of Multi Organ Transplantation, University of California, San Francisco, San Francisco, CA, United States
| | - Joanna M. Schaenman
- Division of Infectious Diseases, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Suphamai Bunnapradist
- Division of Nephrology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Lewis L. Lanier
- Department of Microbiology and Immunology, Parker Institute for Cancer Immunotherapy, University of California, San Francisco, San Francisco, CA, United States
| | - Harry Pickering
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Elaine F. Reed
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
12
|
Chancellor A, Constantin D, Berloffa G, Yang Q, Nosi V, Loureiro JP, Colombo R, Jakob RP, Joss D, Pfeffer M, De Simone G, Morabito A, Schaefer V, Vacchini A, Brunelli L, Montagna D, Heim M, Zippelius A, Davoli E, Häussinger D, Maier T, Mori L, De Libero G. The carbonyl nucleobase adduct M 3Ade is a potent antigen for adaptive polyclonal MR1-restricted T cells. Immunity 2025; 58:431-447.e10. [PMID: 39701104 DOI: 10.1016/j.immuni.2024.11.019] [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: 10/06/2023] [Revised: 07/04/2024] [Accepted: 11/21/2024] [Indexed: 12/21/2024]
Abstract
The major histocompatibility complex (MHC) class I-related molecule MHC-class-I-related protein 1 (MR1) presents metabolites to distinct MR1-restricted T cell subsets, including mucosal-associated invariant T (MAIT) and MR1T cells. However, self-reactive MR1T cells and the nature of recognized antigens remain underexplored. Here, we report a cell endogenous carbonyl adduct of adenine (8-(9H-purin-6-yl)-2-oxa-8-azabicyclo[3.3.1]nona-3,6-diene-4,6-dicarbaldehyde [M3Ade]) sequestered in the A' pocket of MR1. M3Ade induced in vitro MR1-mediated stimulation of MR1T cell clones that bound MR1-M3Ade tetramers. MR1-M3Ade tetramers identified heterogeneous MR1-reactive T cells ex vivo in healthy donors, individuals with acute myeloid leukemia, and tumor-infiltrating lymphocytes from non-small cell lung adenocarcinoma and hepatocarcinoma. These cells displayed phenotypic, transcriptional, and functional diversity at distinct differentiation stages, indicating their adaptive nature. They were also polyclonal, with some preferential T cell receptor (TCRαβ) pair usage. Thus, M3Ade is an MR1-presented self-metabolite that enables stimulation and tracking of human-MR1T cells from blood and tissue, aiding our understanding of their roles in health and disease.
Collapse
Affiliation(s)
- Andrew Chancellor
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland.
| | - Daniel Constantin
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Giuliano Berloffa
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Qinmei Yang
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Vladimir Nosi
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland; Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy
| | - José Pedro Loureiro
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Rodrigo Colombo
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Roman P Jakob
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Daniel Joss
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Michael Pfeffer
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Giulia De Simone
- Department of Environmental Health Science, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milano, Italy
| | - Aurelia Morabito
- Department of Environmental Health Science, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milano, Italy
| | - Verena Schaefer
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Alessandro Vacchini
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Laura Brunelli
- Department of Environmental Health Science, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milano, Italy
| | - Daniela Montagna
- Department of Sciences Clinic-Surgical, Diagnostic and Pediatric, University of Pavia and Pediatric Clinic, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Markus Heim
- Hepatology Laboratory, Department of Biomedicine, University of Basel and University Hospital Basel, 4031 Basel, Switzerland
| | - Alfred Zippelius
- Cancer Immunology, Department of Biomedicine, University of Basel and University Hospital Basel, 4031 Basel, Switzerland
| | - Enrico Davoli
- Department of Environmental Health Science, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, 20156 Milano, Italy
| | - Daniel Häussinger
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Timm Maier
- Biozentrum, University of Basel, 4056 Basel, Switzerland
| | - Lucia Mori
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland
| | - Gennaro De Libero
- Experimental Immunology, Department of Biomedicine, University Hospital Basel, University of Basel, 4031 Basel, Switzerland.
| |
Collapse
|
13
|
Wu Z, Chen X, Han F, Leeansyah E. MAIT cell homing in intestinal homeostasis and inflammation. SCIENCE ADVANCES 2025; 11:eadu4172. [PMID: 39919191 PMCID: PMC11804934 DOI: 10.1126/sciadv.adu4172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/08/2025] [Indexed: 02/09/2025]
Abstract
Mucosa-associated invariant T (MAIT) cells are a large population of unconventional T cells widely distributed in the human gastrointestinal tract. Their homing to the gut is central to maintaining mucosal homeostasis and immunity. This review discusses the potential mechanisms that guide MAIT cells to the intestinal mucosa during homeostasis and inflammation, emphasizing the roles of chemokines, chemokine receptors, and tissue adhesion molecules. The potential influence of the gut microbiota on MAIT cell homing to different regions of the human gut is also discussed. Last, we introduce how organoid technology offers a potentially valuable approach to advance our understanding of MAIT cell tissue homing by providing a more physiologically relevant model that mimics the human gut tissue. These models may enable a detailed investigation of the gut-specific homing mechanisms of MAIT cells. By understanding the regulation of MAIT cell homing to the human gut, potential avenues for therapeutic interventions targeting gut inflammatory conditions such as inflammatory bowel diseases (IBD) may emerge.
Collapse
Affiliation(s)
- Zhengyu Wu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Xingchi Chen
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Fei Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Edwin Leeansyah
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| |
Collapse
|
14
|
Zhang Y, Yang Z, Tan X, Jiang N, Cao G, Yang Q. MAIT cell deficiency exacerbates neuroinflammation in P301S human tau transgenic mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.03.631124. [PMID: 39803476 PMCID: PMC11722295 DOI: 10.1101/2025.01.03.631124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/18/2025]
Abstract
The role of immune cells in neurodegeneration remains incompletely understood. Our recent study revealed the presence of mucosal-associated invariant T (MAIT) cells in the meninges, where they express antioxidant molecules to maintain meningeal barrier integrity. Accumulation of misfolded tau proteins are a hallmark of neurodegenerative diseases. The role of MAIT cells in tau-related neuroinflammation and neurodegeneration, however, remains unclear. Here we report that the meninges of P301 mutant human tau transgenic mice had increased numbers of MAIT cells, which retained their expression of antioxidant molecules. Mr1 -/- P301S mice that lacked MAIT cells exhibited increased tau pathology and hippocampus atrophy compared to control Mr1 +/+ P301S mice. Adoptive transfer of MAIT cells reduced tau pathology and hippocampus atrophy in Mr1 -/- P301S mice. Meningeal barrier integrity was compromised in Mr/ -/- P301S mice, but not in control Mr1 +/+ P301S mice. A distinctive microglia subset with proinflammatory gene expression profile (M-inflammatory) was enriched in the hippocampus of Mr1 -/- P301S mice. The transcriptomes of the remaining microglia in these mice also shifted towards a proinflammatory state, with increased expression of inflammatory cytokines, chemokines, and genes related with ribosome biogenesis and immune responses to toxic substances. The transfer of MAIT cells restored meningeal barrier integrity and suppressed microglial inflammation in the Mr1 -/- P301S mice. Together, our data indicate an important role for MAIT cells in regulating tau-pathology-related neuroinflammation and neurodegeneration.
Collapse
|
15
|
Lagattuta KA, Kohlgruber AC, Abdelfattah NS, Nathan A, Rumker L, Birnbaum ME, Elledge SJ, Raychaudhuri S. The T cell receptor sequence influences the likelihood of T cell memory formation. Cell Rep 2025; 44:115098. [PMID: 39731734 PMCID: PMC11785489 DOI: 10.1016/j.celrep.2024.115098] [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: 07/31/2024] [Revised: 09/19/2024] [Accepted: 12/02/2024] [Indexed: 12/30/2024] Open
Abstract
The amino acid sequence of the T cell receptor (TCR) varies between T cells of an individual's immune system. Particular TCR residues nearly guarantee mucosal-associated invariant T (MAIT) and natural killer T (NKT) cell transcriptional fates. To define how the TCR sequence affects T cell fates, we analyze the paired αβTCR sequence and transcriptome of 961,531 single cells. We find that hydrophobic complementarity-determining region (CDR)3 residues promote regulatory T cell fates in both the CD8 and CD4 lineages. Most strikingly, we find a set of TCR sequence features that promote the T cell transition from naive to memory. We quantify the extent of these features through our TCR scoring function "TCR-mem." Using TCR transduction experiments, we demonstrate that increased TCR-mem promotes T cell activation, even among T cells that recognize the same antigen. Our results reveal a common set of TCR sequence features that enable T cell activation and immunological memory.
Collapse
MESH Headings
- Immunologic Memory
- Animals
- Mice
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Memory T Cells/immunology
- Memory T Cells/metabolism
- Complementarity Determining Regions/genetics
- Humans
- Amino Acid Sequence
- Lymphocyte Activation/immunology
- Mice, Inbred C57BL
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Kaitlyn A Lagattuta
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ayano C Kohlgruber
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA; Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Nouran S Abdelfattah
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Aparna Nathan
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Laurie Rumker
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael E Birnbaum
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Department of Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Stephen J Elledge
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Li J, Xiao C, Li C, He J. Tissue-resident immune cells: from defining characteristics to roles in diseases. Signal Transduct Target Ther 2025; 10:12. [PMID: 39820040 PMCID: PMC11755756 DOI: 10.1038/s41392-024-02050-5] [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: 06/04/2024] [Revised: 09/28/2024] [Accepted: 11/04/2024] [Indexed: 01/19/2025] Open
Abstract
Tissue-resident immune cells (TRICs) are a highly heterogeneous and plastic subpopulation of immune cells that reside in lymphoid or peripheral tissues without recirculation. These cells are endowed with notably distinct capabilities, setting them apart from their circulating leukocyte counterparts. Many studies demonstrate their complex roles in both health and disease, involving the regulation of homeostasis, protection, and destruction. The advancement of tissue-resolution technologies, such as single-cell sequencing and spatiotemporal omics, provides deeper insights into the cell morphology, characteristic markers, and dynamic transcriptional profiles of TRICs. Currently, the reported TRIC population includes tissue-resident T cells, tissue-resident memory B (BRM) cells, tissue-resident innate lymphocytes, tissue-resident macrophages, tissue-resident neutrophils (TRNs), and tissue-resident mast cells, but unignorably the existence of TRNs is controversial. Previous studies focus on one of them in specific tissues or diseases, however, the origins, developmental trajectories, and intercellular cross-talks of every TRIC type are not fully summarized. In addition, a systemic overview of TRICs in disease progression and the development of parallel therapeutic strategies is lacking. Here, we describe the development and function characteristics of all TRIC types and their major roles in health and diseases. We shed light on how to harness TRICs to offer new therapeutic targets and present burning questions in this field.
Collapse
Affiliation(s)
- Jia Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chu Xiao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
17
|
Mi Q, Wu X, Chen Y, Meng W. MAIT cells modulating the oral lichen planus immune microenvironment: a cellular crosstalk perspective. Inflamm Res 2025; 74:10. [PMID: 39762617 DOI: 10.1007/s00011-024-01990-6] [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: 10/14/2024] [Revised: 12/22/2024] [Accepted: 12/24/2024] [Indexed: 01/11/2025] Open
Abstract
Mucosal-associated invariant T (MAIT) cells, a type of T lymphocytes with innate-like characteristics, are crucial in bridging innate and adaptive immunity. When activated, MAIT cells release various inflammatory molecules and swiftly respond to antigens. Notably, numerous studies highlight the significant impact of MAIT cells on tumors and various immune disorders by influencing the immune microenvironment. Oral lichen planus (OLP) is an immune-mediated inflammatory condition mainly involving T lymphocytes. Previous research primarily focused on T cells alone, neglecting the broader immune environment. However, there is a current growing recognition of the complex interactions among multiple immune cells and inflammatory factors in patients with OLP. This immune microenvironment comprises T lymphocytes, fibroblasts, keratinocytes, dendritic cells, macrophages, inflammation-related cytokines, and chemokines, orchestrating intricate interactions that contribute to OLP initiation and persistence. Therefore, this review consolidates current research on the interplay between MAIT cells and other immune cells within the OLP microenvironment. We also delve into potential mechanisms through which MAIT cells regulate inflammation in patients with OLP, aiming to further explore the role of MAIT cells in these patients.
Collapse
Affiliation(s)
- Qian Mi
- Departments of Oral Medicine, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xiaoli Wu
- Departments of Oral Medicine, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Yuhe Chen
- Departments of Oral Medicine, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Wenxia Meng
- Departments of Oral Medicine, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong Province, China.
| |
Collapse
|
18
|
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
|
19
|
Dolton G, Thomas H, Tan LR, Rius Rafael C, Doetsch S, Ionescu GA, Cardo LF, Crowther MD, Behiry E, Morin T, Caillaud ME, Srai D, Parolini L, Hasan MS, Fuller A, Topley K, Wall A, Hopkins JR, Omidvar N, Alvares C, Zabkiewicz J, Frater J, Szomolay B, Sewell AK. MHC-related protein 1-restricted recognition of cancer via a semi-invariant TCR-α chain. J Clin Invest 2025; 135:e181895. [PMID: 39744940 PMCID: PMC11684821 DOI: 10.1172/jci181895] [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: 04/11/2024] [Accepted: 10/29/2024] [Indexed: 01/16/2025] Open
Abstract
The T cell antigen presentation platform MR1 consists of 6 allomorphs in humans that differ by no more than 5 amino acids. The principal function of this highly conserved molecule involves presenting microbial metabolites to the abundant mucosal-associated invariant T (MAIT) cell subset. Recent developments suggest that the role of MR1 extends to presenting antigens from cancer cells, a function dependent on the K43 residue in the MR1 antigen binding cleft. Here, we successfully cultured cancer-activated, MR1-restricted T cells from multiple donors and confirmed that they recognized a wide range of cancer types expressing the most common MR1*01 and/or MR1*02 allomorphs (over 95% of the population), while remaining inert to healthy cells including healthy B cells and monocytes. Curiously, in all but one donor these T cells were found to incorporate a conserved TCR-α chain motif, CAXYGGSQGNLIF (where X represents 3-5 amino acids), because of pairing between 10 different TRAV genes and the TRAJ42 gene segment. This semi-invariance in the TCR-α chain is reminiscent of MAIT cells and suggests recognition of a conserved antigen bound to K43.
Collapse
MESH Headings
- Humans
- Minor Histocompatibility Antigens/genetics
- Minor Histocompatibility Antigens/immunology
- Minor Histocompatibility Antigens/metabolism
- Histocompatibility Antigens Class I/immunology
- Histocompatibility Antigens Class I/genetics
- Histocompatibility Antigens Class I/metabolism
- Neoplasms/immunology
- Neoplasms/genetics
- Neoplasms/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Mucosal-Associated Invariant T Cells/immunology
- Mucosal-Associated Invariant T Cells/metabolism
- Antigen Presentation
- Antigens, Neoplasm/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/metabolism
- Amino Acid Motifs
- Cell Line, Tumor
Collapse
Affiliation(s)
- Garry Dolton
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Hannah Thomas
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Li Rong Tan
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Cristina Rius Rafael
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Stephanie Doetsch
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Giulia-Andreea Ionescu
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Lucia F. Cardo
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Michael D. Crowther
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Enas Behiry
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Théo Morin
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Marine E. Caillaud
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Devinder Srai
- Nuffield Department of Medicine and Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Lucia Parolini
- Nuffield Department of Medicine and Department of Chemistry, University of Oxford, Oxford, United Kingdom
| | - Md Samiul Hasan
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Anna Fuller
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Katie Topley
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Aaron Wall
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Jade R. Hopkins
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Nader Omidvar
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Caroline Alvares
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Joanna Zabkiewicz
- Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - John Frater
- Nuffield Department of Medicine and NIHR Biomedical Research Centre University of Oxford, Oxford, United Kingdom
| | - Barbara Szomolay
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunology Research Institute, Cardiff University Cardiff, United Kingdom
| | - Andrew K. Sewell
- Division of Infection and Immunity, Cardiff University School of Medicine, Cardiff, United Kingdom
- Systems Immunology Research Institute, Cardiff University Cardiff, United Kingdom
- Division of Infection and Immunity, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
20
|
Li Y, Ong JWX, See YM, Yee JY, Tang C, Zheng S, Ng BT, Lee BTK, Rotzschke O, Andiappan AK, Lee J. Immunophenotyping schizophrenia subtypes stratified by antipsychotic response. Brain Behav Immun 2025; 123:656-671. [PMID: 39414177 DOI: 10.1016/j.bbi.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/02/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024] Open
Abstract
Immune dysfunction has been proposed to play a role in the pathophysiology behind the development and persistence of psychosis. Current immunophenotyping studies are limited by small sample sizes and the number of immune markers investigated. Pharmacological subtypes in schizophrenia based on antipsychotic response have been proposed, but few studies have investigated immunophenotypes in treatment-resistant schizophrenia. In this study, we perform comprehensive immunophenotyping on 196 subjects comprising 147 schizophrenia patients stratified by antipsychotic response (49 antipsychotic-responsive, 70 clozapine-responsive, 28 clozapine-resistant) and 49 healthy controls. We aim to identify significant immune cell populations associated with schizophrenia and increasing treatment resistance, as potential modulators of underlying psychosis and/or treatment response. Patients with schizophrenia were recruited and assessed on the Clinical Global Impression - Schizophrenia (CGI-SCH). Treatment response was defined as a rating of three (mild severity) or less on the CGI-SCH positive symptom item after at least 8 weeks of adequate antipsychotic or clozapine treatment. Peripheral blood mononuclear cells were collected and flow cytometry was performed to identify 66 immune cell populations. Differences in cell population proportions were compared between schizophrenia cases and controls, and across all 4 groups, with post-hoc pairwise comparisons. Mucosal-associated invariant T (MAIT) cells (specifically CD8 + and DN double-negative subsets), total, exhausted and memory CD8 + T cells, VD1 + ϒδ T cells, plasmablasts, IgG + B cells and conventional dendritic cells 2 (cDC2) were among the top cell populations downregulated in schizophrenia. We observed increased downregulation with increasing treatment resistance. Conversely, naïve and exhausted CD4 + T cells, CD4/CD8 ratio and CCR5 + CCR2 + HLA DR + Myeloid cells were found to be upregulated in schizophrenia - we observed increased upregulation with increasing treatment resistance. We show significant immunophenotypic differences between schizophrenia cases and healthy controls, and consistent trend differences across varying degrees of antipsychotic resistance. We also examined immune cell populations not previously reported in schizophrenia. Future studies may explore immune markers identified as potential biomarkers of treatment resistance, and clarify on the relationship between immunological changes and pharmacological subtypes in schizophrenia.
Collapse
Affiliation(s)
- Yanhui Li
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Jocelyn Wen Xin Ong
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore. 8A Biomedical Grove Level 3 & 4. Immunos Building Singapore 138648, Singapore
| | - Yuen Mei See
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Jie Yin Yee
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Charmaine Tang
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Shushan Zheng
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Boon Tat Ng
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore
| | - Bernett Teck Kwong Lee
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore. 1 Mandalay Rd, Singapore 308232, Singapore
| | - Olaf Rotzschke
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore. 8A Biomedical Grove Level 3 & 4. Immunos Building Singapore 138648, Singapore
| | - Anand Kumar Andiappan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A*STAR), Singapore. 8A Biomedical Grove Level 3 & 4. Immunos Building Singapore 138648, Singapore
| | - Jimmy Lee
- Institute of Mental Health, Singapore. 10 Buangkok View, Buangkok Green Medical Park, Singapore 539747, Singapore; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore. 1 Mandalay Rd, Singapore 308232, Singapore.
| |
Collapse
|
21
|
Afify R, Lipsius K, Wyatt-Johnson SJ, Brutkiewicz RR. Myeloid antigen-presenting cells in neurodegenerative diseases: a focus on classical and non-classical MHC molecules. Front Neurosci 2024; 18:1488382. [PMID: 39720231 PMCID: PMC11667120 DOI: 10.3389/fnins.2024.1488382] [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: 08/29/2024] [Accepted: 11/20/2024] [Indexed: 12/26/2024] Open
Abstract
In recent years, increasing evidence has highlighted the critical role of myeloid cells, specifically those that present antigen (APCs) in health and disease. These shape the progression and development of neurodegenerative disorders, where considerable interplay between the immune system and neurons influences the course of disease pathogenesis. Antigen-presenting myeloid cells display different classes of major histocompatibility complex (MHC) and MHC-like proteins on their surface for presenting various types of antigens to a wide variety of T cells. While most studies focus on the role of myeloid MHC class I and II molecules in health and disease, there is still much that remains unknown about non-polymorphic MHC-like molecules such as CD1d and MR1. Thus, in this review, we will summarize the recent findings regarding the contributions of both classical and non-classical MHC molecules, particularly on myeloid microglial APCs, in neurodegenerative diseases. This will offer a better understanding of altered mechanisms that may pave the way for the development of novel therapeutic strategies targeting immune cell-MHC interactions, to mitigate neurodegeneration and its associated pathology.
Collapse
Affiliation(s)
| | | | | | - Randy R. Brutkiewicz
- Department of Microbiology and Immunology and Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN, United States
| |
Collapse
|
22
|
Yang Z, Luo B, Li M, He Z, Ren C, Chen X, Kang X, Chen H, Xu E, Guan W, Xia X. The effector function of mucosal associated invariant T cells alters with aging and is regulated by RORγt. Front Immunol 2024; 15:1504806. [PMID: 39669566 PMCID: PMC11634854 DOI: 10.3389/fimmu.2024.1504806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 11/01/2024] [Indexed: 12/14/2024] Open
Abstract
Introduction Mucosal-associated invariant T (MAIT) cells are a predominant subset of innate-like T cells in humans, characterized by diverse gene expression profiles and functional capabilities. However, the factors influencing the transcriptomes and effector functions of MAIT cells, particularly at mucosal barriers, remain largely unclear. Methods In this study, we employed single-cell RNA sequencing (scRNA-seq) and functional assays to investigate the transcriptomic and functional characteristics of intestinal MAIT cells in mouse models during aging. We also extended scRNA-seq analysis to human intestinal MAIT cells to compare their gene expression patterns with those observed in aged mice. Results Our findings demonstrated that the transcriptomes and functional capabilities of intestinal MAIT cells shifted from MAIT17 to MAIT1 profiles with aging in mouse models, with notable changes in the production of cytotoxic molecules. Further scRNA-seq analysis of human intestinal MAIT cells revealed a segregation into MAIT1 and MAIT17 subsets, displaying gene expression patterns that mirrored those seen in aged mouse models. The transcription factor RORγt was expressed in both MAIT1 and MAIT17 cells, acting to repress IFNγ production while promoting IL17 expression. Moreover, reduced expression of RORC and Il17A was correlated with poorer survival outcomes in colorectal cancer patients. Discussion These results suggest that aging induces a functional shift between MAIT1 and MAIT17 cells, which may be influenced by transcriptional regulators like RORγt. The observed alterations in MAIT cell activity could potentially impact disease prognosis, particularly in colorectal cancer. This study provides new insights into the dynamics of MAIT cell responses at mucosal barriers, highlighting possible therapeutic targets for modulating MAIT cell functions in aging and disease.
Collapse
Affiliation(s)
- Zhi Yang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Banxin Luo
- Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Minhuan Li
- Department of Andrology, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Ziyun He
- Department of Gastrointestinal Surgery, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Chuanfu Ren
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
| | - Xin Chen
- Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xing Kang
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Hong Chen
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - En Xu
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Wenxian Guan
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Nanjing Drum Tower Hospital, Drum Tower Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
- Department of General Surgery, Taikang Xianlin DrumTower Hospital, The Affiliated Hospital of Wuhan University Medical School, Nanjing, China
| | - Xuefeng Xia
- Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
- Department of General Surgery, Drum Tower Clinical Medical College of Nanjing Medical University, Nanjing, China
- Department of General Surgery, Taikang Xianlin DrumTower Hospital, The Affiliated Hospital of Wuhan University Medical School, Nanjing, China
| |
Collapse
|
23
|
Takasaki R, Ito E, Nagae M, Takahashi Y, Matsuoka T, Yasue W, Arichi N, Ohno H, Yamasaki S, Inuki S. Development of Ribityllumazine Analogue as Mucosal-Associated Invariant T Cell Ligands. J Am Chem Soc 2024; 146:29964-29976. [PMID: 39432319 DOI: 10.1021/jacs.4c12997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells are a subset of innate-like T cells abundant in human tissues that play a significant role in defense against bacterial and viral infections and in tissue repair. MAIT cells are activated by recognizing microbial-derived small-molecule ligands presented by the MHC class I related-1 protein. Although several MAIT cell modulators have been identified in the past decade, potent and chemically stable ligands remain limited. Herein, we carried out a structure-activity relationship study of ribityllumazine derivatives and found a chemically stable MAIT cell ligand with a pteridine core and a 2-oxopropyl group as the Lys-reactive group. The ligand showed high potency in a cocultivation assay using model cell lines of antigen-presenting cells and MAIT cells. The X-ray crystallographic analysis revealed the binding mode of the ligand to MR1 and the T cell receptor, indicating that it forms a covalent bond with MR1 via Schiff base formation. Furthermore, we found that the ligand stimulated proliferation of human MAIT cells in human peripheral blood mononuclear cells and showed an adjuvant effect in mice. Our developed ligand is one of the most potent among chemically stable MAIT cell ligands, contributing to accelerating therapeutic applications of MAIT cells.
Collapse
Affiliation(s)
- Ryosuke Takasaki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Emi Ito
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan
| | - Masamichi Nagae
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan
| | - Yuki Takahashi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Takuro Matsuoka
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Wakana Yasue
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Norihito Arichi
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Hiroaki Ohno
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
| | - Sho Yamasaki
- Research Institute for Microbial Diseases, Osaka University, Suita, Osaka 565-0871, Japan
- Immunology Frontier Research Center (IFReC), Osaka University, Suita, Osaka 565-0871, Japan
| | - Shinsuke Inuki
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Kyoto 606-8501, Japan
- Graduate School of Biomedical Sciences, Tokushima University, Tokushima, Tokushima 770-8505, Japan
| |
Collapse
|
24
|
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
|
25
|
Cornforth TV, Moyo N, Cole S, Lam EPS, Lobry T, Wolchinsky R, Lloyd A, Ward K, Denham EM, Masi G, Qing Yun PT, Moore C, Dhaouadi S, Besra GS, Veerapen N, Illing PT, Vivian JP, Raynes JM, Le Nours J, Purcell AW, Kundu S, Silk JD, Williams L, Papa S, Rossjohn J, Howie D, Dukes J. Conserved allomorphs of MR1 drive the specificity of MR1-restricted TCRs. Front Oncol 2024; 14:1419528. [PMID: 39445059 PMCID: PMC11496959 DOI: 10.3389/fonc.2024.1419528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/05/2024] [Indexed: 10/25/2024] Open
Abstract
Background Major histocompatibility complex class-1-related protein (MR1), unlike human leukocyte antigen (HLA) class-1, was until recently considered to be monomorphic. MR1 presents metabolites in the context of host responses to bacterial infection. MR1-restricted TCRs specific to tumor cells have been described, raising interest in their potential therapeutic application for cancer treatment. The diversity of MR1-ligand biology has broadened with the observation that single nucleotide variants (SNVs) exist within MR1 and that allelic variants can impact host immunity. Methods The TCR from a MR1-restricted T-cell clone, MC.7.G5, with reported cancer specificity and pan-cancer activity, was cloned and expressed in Jurkat E6.1 TCRαβ- β2M- CD8+ NF-κB:CFP NFAT:eGFP AP-1:mCherry cells or in human donor T cells. Functional activity of 7G5.TCR-T was demonstrated using cytotoxicity assays and by measuring cytokine release after co-culture with cancer cell lines with or without loading of previously described MR1 ligands. MR1 allele sequencing was undertaken after the amplification of the MR1 gene region by PCR. In vivo studies were undertaken at Labcorp Drug Development (Ann Arbor, MI, USA) or Epistem Ltd (Manchester, UK). Results The TCR cloned from MC.7.G5 retained MR1-restricted functional cytotoxicity as 7G5.TCR-T. However, activity was not pan-cancer, as initially reported with the clone MC.7.G5. Recognition was restricted to cells expressing a SNV of MR1 (MR1*04) and was not cancer-specific. 7G5.TCR-T and 7G5-like TCR-T cells reacted to both cancer and healthy cells endogenously expressing MR1*04 SNVs, which encode R9H and H17R substitutions. This allelic specificity could be overcome by expressing supraphysiological levels of the wild-type MR1 (MR1*01) in cell lines. Conclusions Healthy individuals harbor T cells reactive to MR1 variants displaying self-ligands expressed in cancer and benign tissues. Described "cancer-specific" MR1-restricted TCRs need further validation, covering conserved allomorphs of MR1. Ligands require identification to ensure targeting MR1 is restricted to those specific to cancer and not normal tissues. For the wider field of immunology and transplant biology, the observation that MR1*04 may behave as an alloantigen warrants further study. .
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | - Gurdyal S. Besra
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Natacha Veerapen
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, United Kingdom
| | - Patricia T. Illing
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Julian P. Vivian
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jeremy M. Raynes
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Jérôme Le Nours
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | - Anthony W. Purcell
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
| | | | | | | | - Sophie Papa
- Enara Bio Ltd., Oxford, United Kingdom
- School of Cancer and Pharmaceutical Sciences, King’s College London, Guy’s Hospital, London, United Kingdom
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, VIC, Australia
- Institute of Infection and Immunity, Cardiff University, School of Medicine, Cardiff, United Kingdom
| | | | | |
Collapse
|
26
|
López-Rodríguez JC, Barral P. Mucosal associated invariant T cells: Powerhouses of the lung. Immunol Lett 2024; 269:106910. [PMID: 39128630 PMCID: PMC11835791 DOI: 10.1016/j.imlet.2024.106910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 07/29/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The lungs face constant environmental challenges from harmless molecules, airborne pathogens and harmful agents that can damage the tissue. The lungs' immune system includes numerous tissue-resident lymphocytes that contribute to maintain tissue homeostasis and to the early initiation of immune responses. Amongst tissue-resident lymphocytes, Mucosal Associated Invariant T (MAIT) cells are present in human and murine lungs and emerging evidence supports their contribution to immune responses during infections, chronic inflammatory disorders and cancer. This review explores the mechanisms underpinning MAIT cell functions in the airways, their impact on lung immunity and the potential for targeting pulmonary MAIT cells in a therapeutic context.
Collapse
Affiliation(s)
- J C López-Rodríguez
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK.
| | - P Barral
- Centre for Inflammation Biology and Cancer Immunology, The Peter Gorer Department of Immunobiology, King's College London, London, UK; The Francis Crick Institute, London, UK.
| |
Collapse
|
27
|
Galaverna F, Flamini S, De Luca CD, Pili I, Boccieri E, Benini F, Quagliarella F, Rosignoli C, Rosichini M, Genah S, Catanoso M, Cardinale A, Volpe G, Coccetti M, Pitisci A, Li Pira G, Carta R, Lucarelli B, Del Bufalo F, Bertaina V, Becilli M, Pagliara D, Algeri M, Merli P, Locatelli F, Velardi E. Mucosal-associated invariant T cells are functionally impaired in pediatric and young adult patients following allogeneic hematopoietic stem cell transplantation and their recovery correlates with clinical outcomes. Haematologica 2024; 109:3222-3236. [PMID: 38813718 PMCID: PMC11443409 DOI: 10.3324/haematol.2023.284649] [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: 11/16/2023] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Abstract
Mucosal-associated invariant T (MAIT) cells are innate-like T cells implicated in the response to fungal and bacterial infections. Their contribution to restoring T-cell immunity and influencing hematopoietic stem cell transplant (HSCT) outcomes remains poorly understood. We retrospectively studied MAIT-cell recovery in 145 consecutive children and young adults with hematologic malignancies undergoing allogeneic (allo)-HSCT between April 2019 and May 2022, from unrelated matched donor (MUD, N=52), with standard graft-versus-host-disease (GvHD) prophylaxis, or HLA-haploidentical (Haplo, N=93) donor after in vitro αβT/CD19-cell depletion, without post-HSCT pharmacological prophylaxis. With a median follow-up of 33 months (range, 12-49 months), overall survival (OS), disease-free survival (DFS), and non-relapse mortality (NRM) were 79.5%, 72%, and 7%, respectively; GvHD-free relapse-free survival (GRFS) was 63%, while cumulative incidence of relapse was 23%. While αβT cells were reconstituted 1-2 years post HSCT, MAIT cells showed delayed recovery and prolonged functional impairment, characterized by expression of activation (CD25, CD38), exhaustion (PD1, TIM3) and senescence (CD57) markers, and suboptimal ex vivo response. OS, DFS, and NRM were not affected by MAIT cells. Interestingly, higher MAIT cells at day +30 correlated with higher incidence of grade II-IV acute GvHD (19% vs. 7%, P=0.06). Furthermore, a greater MAIT-cell count tended to be associated with a higher incidence of chronic GvHD (cGvHD) (17% vs. 6%, P=0.07) resulting in lower GRFS (55% vs. 73%, P=0.05). Higher MAIT cells also correlated with greater cytomegalovirus (CMV) reactivation and lower late blood stream infections (BSI) (44% vs. 24%, P=0.02 and 9% vs. 18%, P=0.08, respectively). Future studies are needed to confirm the impact of early MAIT-cell recovery on cGvHD, CMV reactivation, and late BSI.
Collapse
Affiliation(s)
- Federica Galaverna
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Sara Flamini
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Carmen Dolores De Luca
- Department of Maternal and Child Health, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome
| | - Ilaria Pili
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Emilia Boccieri
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Francesca Benini
- Department of Maternal and Child Health, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome
| | - Francesco Quagliarella
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Chiara Rosignoli
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Marco Rosichini
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome
| | - Shirley Genah
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Marialuigia Catanoso
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Antonella Cardinale
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Gabriele Volpe
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Marianna Coccetti
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Angela Pitisci
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Giuseppina Li Pira
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Roberto Carta
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Barbarella Lucarelli
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Francesca Del Bufalo
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Valentina Bertaina
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Marco Becilli
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Daria Pagliara
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Mattia Algeri
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Health Sciences, Magna Graecia University, Catanzaro
| | - Pietro Merli
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome
| | - Franco Locatelli
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Maternal and Child Health, Catholic University of the Sacred Heart, Largo Francesco Vito, 1, 00168 Rome.
| | - Enrico Velardi
- Research Area of Hematology and Oncology, Cell and Gene Therapy, Bambino Gesù Children's Hospital, IRCCS, Rome.
| |
Collapse
|
28
|
Abacar K, Macleod T, Direskeneli H, McGonagle D. How underappreciated autoinflammatory (innate immunity) mechanisms dominate disparate autoimmune disorders. Front Immunol 2024; 15:1439371. [PMID: 39372419 PMCID: PMC11449752 DOI: 10.3389/fimmu.2024.1439371] [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: 05/27/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
Historically inflammation against self was considered autoimmune which stems back to the seminal observations by Ehrlich who described serum factors, now known to be autoantibodies produced by B lineage cells that mediate "horror autotoxicus". The 20th century elucidation of B- and T-cell adaptive immune responses cemented the understanding of the key role of adaptive immune responses in mediating pathology against self. However, Mechnikov shared the Nobel Prize for the discovery of phagocytosis, the most rudimentary aspect of innate immunity. Fast forward some 100 years and an immunogenetic understanding of innate immunity led to the categorising of innate immunopathology under the umbrella term 'auto inflammation' and terminology such as "horror autoinflammaticus" to highlight the schism from the classical adaptive immune understanding of autoimmunity. These concepts lead to calls for a two-tiered classification of inflammation against self, but just as innate and adaptive immunity are functionally integrated, so is immunopathology in many settings and the concept of an autoimmune to autoinflammation continuum emerged with overlaps between both. Herein we describe several historically designated disorders of adaptive immunity where innate immunity is key, including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), systemic juvenile idiopathic arthritis (sJIA) and adult-onset Still's disease (AOSD) where the immunopathology phenotype is strongly linked to major histocompatibility complex (MHC) class II associations and responds to drugs that target T-cells. We also consider MHC-I-opathies including psoriasis and Behcet's disease(BD) that are increasingly viewed as archetype CD8 T-cell related disorders. We also briefly review the key role of barrier dysfunction in eczema and ulcerative colitis (UC) where innate tissue permeability barrier dysfunction and microbial dysbiosis contributes to prominent adaptive immune pathological mechanisms. We also highlight the emerging roles of intermediate populations of lymphocytes including gamma delta (γδ) and mucosal-associated invariant T (MAIT) cells that represent a blend of adaptive immune plasticity and innate immune rapid responders that may also determine site specific patterns of inflammation.
Collapse
Affiliation(s)
- Kerem Abacar
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Tom Macleod
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
| | - Haner Direskeneli
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds, United Kingdom
| | - Dennis McGonagle
- Department of Internal Medicine, Division of Rheumatology, Marmara University School of Medicine, Istanbul, Türkiye
- National Institute for Health Research, Leeds Biomedical Research Centre, Leeds Teaching Hospitals, Leeds, United Kingdom
| |
Collapse
|
29
|
Cheng OJ, Lebish EJ, Jensen O, Jacenik D, Trivedi S, Cacioppo JG, Aubé J, Beswick EJ, Leung DT. Mucosal-associated invariant T cells modulate innate immune cells and inhibit colon cancer growth. Scand J Immunol 2024; 100:e13391. [PMID: 38773691 PMCID: PMC11315626 DOI: 10.1111/sji.13391] [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: 01/27/2024] [Revised: 04/16/2024] [Accepted: 05/05/2024] [Indexed: 05/24/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells are innate-like T cells that can be activated by microbial antigens and cytokines and are abundant in mucosal tissues including the colon. MAIT cells have cytotoxic and pro-inflammatory functions and have potentials for use as adoptive cell therapy. However, studies into their anti-cancer activity, including their role in colon cancer, are limited. Using an animal model of colon cancer, we showed that peritumoral injection of in vivo-expanded MAIT cells into RAG1-/- mice with MC38-derived tumours inhibits tumour growth compared to control. Multiplex cytokine analyses showed that tumours from the MAIT cell-treated group have higher expression of markers for eosinophil-activating cytokines, suggesting a potential association between eosinophil recruitment and tumour inhibition. In a human peripheral leukocyte co-culture model, we showed that leukocytes stimulated with MAIT ligand showed an increase in eotaxin-1 production and activation of eosinophils, associated with increased cancer cell killing. In conclusion, we showed that MAIT cells have a protective role in a murine colon cancer model, associated with modulation of the immune response to cancer, potentially involving eosinophil-associated mechanisms. Our results highlight the potential of MAIT cells for non-donor restricted colon cancer immunotherapy.
Collapse
Affiliation(s)
- Olivia J. Cheng
- Division of Microbiology & Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States
- Division of Infectious Disease, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Eric J. Lebish
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Owen Jensen
- Division of Microbiology & Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States
- Division of Infectious Disease, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Damian Jacenik
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
- Department of Cytobiochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Shubhanshi Trivedi
- Division of Infectious Disease, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| | - Jackson G. Cacioppo
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Jeffrey Aubé
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States
| | - Ellen J. Beswick
- Division of Gastroenterology, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
- Division of Digestive Diseases and Nutrition, Department of Internal Medicine, University of Kentucky, Lexington, Kentucky, United States
| | - Daniel T. Leung
- Division of Microbiology & Immunology, Department of Pathology, University of Utah, Salt Lake City, Utah, United States
- Division of Infectious Disease, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, United States
| |
Collapse
|
30
|
Siewert LK, Fromm K, Dehio C, Pinschewer DD. Cutting Edge: Redundant Roles for MHC Class II-, CD1d-, and MR1-restricted T Cells in Clearing Bartonella Infection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:553-558. [PMID: 38984869 PMCID: PMC11335324 DOI: 10.4049/jimmunol.2400045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024]
Abstract
The importance of unconventional T cells for mucosal immunity is firmly established but for systemic bacterial infection remains less well defined. In this study, we explored the role of various T cell subsets in murine Bartonella infection, which establishes persistent bacteremia unless controlled by antibacterial Abs. We found that αβ T cells are essential for Ab production against and clearance of B. taylorii, whereas MHC class I (MHC-I)- or MHC class II (MHC-II)-deficient mice eliminated B. taylorii infection with normal kinetics. Similarly, animals lacking either CD1d or MR1 suppressed bacteremia with normal kinetics. Interestingly, mice with a combined deficiency of either MHC-II and CD1d or MHC-II and MR1 failed to clear the infection, indicating that the combination of CD1d- and MR1-restricted T cells can compensate for the lack of MHC-II in this model. Our data document a previously underappreciated contribution of unconventional T cells to the control of systemic bacterial infection, supposedly as helper cells for antibacterial Ab production.
Collapse
Affiliation(s)
- Lena K. Siewert
- Biozentrum, University of Basel, Basel, Switzerland
- Division of Experimental Virology, Department Biomedicine–Haus Petersplatz, University of Basel, Basel, Switzerland
| | - Katja Fromm
- Biozentrum, University of Basel, Basel, Switzerland
| | | | - Daniel D. Pinschewer
- Division of Experimental Virology, Department Biomedicine–Haus Petersplatz, University of Basel, Basel, Switzerland
| |
Collapse
|
31
|
Touni AA, Sohn R, Cosgrove C, Shivde RS, Dellacecca ER, Abdel-Aziz RTA, Cedercreutz K, Green SJ, Abdel-Wahab H, Le Poole IC. Topical antibiotics limit depigmentation in a mouse model of vitiligo. Pigment Cell Melanoma Res 2024; 37:583-596. [PMID: 38439216 DOI: 10.1111/pcmr.13164] [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: 07/17/2023] [Revised: 01/04/2024] [Accepted: 02/12/2024] [Indexed: 03/06/2024]
Abstract
Oral neomycin administration impacts the gut microbiome and delays vitiligo development in mice, and topical antibiotics may likewise allow the microbiome to preserve skin health and delay depigmentation. Here, we examined the effects of 6-week topical antibiotic treatment on vitiligo-prone pmel-1 mice. Bacitracin, Neosporin, or Vaseline were applied to one denuded flank, while the contralateral flank was treated with Vaseline in all mice. Ventral depigmentation was quantified weekly. We found that topical Neosporin treatment significantly reduced depigmentation and exhibited effects beyond the treated area, while Bacitracin ointment had no effect. Stool samples collected from four representative mice/group during treatment revealed that Neosporin treatment aligned with reduced abundance of the Alistipes genus in the gut, while relevant changes to the skin microbiome at end point were less apparent. Either antibiotic treatment led to reduced expression of MR1, potentially limiting mucosal-associated invariant T-cell activation, while Neosporin-treated skin selectively revealed significantly reduced CD8+ T-cell abundance. The latter finding aligned with reduced expression of multiple inflammatory markers and markedly increased regulatory T-cell density. Our studies on favorable skin and oral antibiotic treatment share the neomycin compound, and in either case, microbial changes were most apparent in stool samples. Taken together, neomycin-containing antibiotic applications can mediate skin Treg infiltration to limit vitiligo development. Our study highlights the therapeutic potential of short-term antibiotic applications to limit depigmentation vitiligo.
Collapse
Affiliation(s)
- Ahmed Ahmed Touni
- Department of Dermatology, Faculty of Medicine, Minia University, Minia, Egypt
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Rachel Sohn
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Cormac Cosgrove
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Rohan S Shivde
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Emilia R Dellacecca
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Kettil Cedercreutz
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Stefan J Green
- Department of Internal Medicine and Genomics and Microbiome Core Facility, Rush University, Chicago, Illinois, USA
| | - Hossam Abdel-Wahab
- Department of Dermatology, Faculty of Medicine, Minia University, Minia, Egypt
| | - I Caroline Le Poole
- Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
- Department of Microbiology and Immunology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
32
|
Borges-Fernandes LO, de Lima Moreira M, Pereira VHS, Pascoal-Xavier MA, Lopes Ribeiro Á, da Costa-Rocha IA, Lopes LR, Moreira GTC, Araújo MSDS, Teixeira-Carvalho A, Brito-de-Sousa JP, de Carvalho AL, Mourão MVA, Campos FA, Borges M, Carneiro M, Tsuji M, Martins-Filho OA, Coelho-dos-Reis JGA, Peruhype-Magalhães V. MR1 blockade drives differential impact on integrative signatures based on circuits of circulating immune cells and soluble mediators in visceral leishmaniasis. Front Immunol 2024; 15:1373498. [PMID: 39192975 PMCID: PMC11347828 DOI: 10.3389/fimmu.2024.1373498] [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: 01/19/2024] [Accepted: 07/15/2024] [Indexed: 08/29/2024] Open
Abstract
Introduction Visceral leishmaniasis (VL) is an important tropical and neglected disease and represents a serious global health problem. The initial interaction between the phagocytes and the parasite is crucial to determine the pathogen's capacity to initiate infection and it shapes the subsequent immune response that will develop. While type-1 T-cells induce IL-6, IL-1β, TNF-α, and IL-12 production by monocytes/macrophages to fight the infection, type-2 T-cells are associated with a regulatory phenotype (IL-10 and TGF-β) and successful infection establishment. Recently, our group demonstrated the role of an important Th1/Th17 T-cell population, the mucosal-associated invariant T (MAIT) cells, in VL. MAIT cells can respond to L. infantum by producing TNF-α and IFN-γ upon MR1-dependent activation. Objective and methods Here, we describe the impact of the MR1-blockage on L. infantum internalization on the functional profile of circulating neutrophils and monocytes as well as the impact of the MR1-blockage on the soluble mediator signatures of in vitro whole blood cultures. Results Overall, our data showed that VL patients presents higher percentage of activated neutrophils than asymptomatic and non-infected controls. In addition, MR1 blockade led to lower TNF-α and TGF-β production by non-activated neutrophils from asymptomatic individuals. Moreover, TNF-α and IL-10 production by monocytes was higher in VL patients. In the analysis of soluble mediators produced in vitro, MR1-blockade induced a decrease of IFN-γ and an increase of IL-10, IL-27 and IL-33 in the cell cultures of AS group, a cytokine pattern associated with type 2 deleterious response. Discussion and conclusion These data corroborate the hypothesis that MR1-restricted responses are associated to a protective role during Leishmania infection.
Collapse
Affiliation(s)
| | - Marcela de Lima Moreira
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
| | | | - 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
- Basic and Applied Virology Laboratory, Department of Microbiology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Ludmila Rosa Lopes
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
| | | | | | - 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
| | - Moriya Tsuji
- Aaron Diamond AIDS Research Center, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
- Division of Infectious Disease, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY, United States
| | | | - Jordana Grazziela Alves Coelho-dos-Reis
- René Rachou Institute, Oswaldo Cruz Foundation (FIOCRUZ-MINAS), Belo Horizonte, Minas Gerais, Brazil
- Basic and Applied Virology Laboratory, Department of Microbiology, Institute for Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | |
Collapse
|
33
|
Mak JYW, Rivero RJD, Hoang HN, Lim XY, Deng J, McWilliam HEG, Villadangos JA, McCluskey J, Corbett AJ, Fairlie DP. Potent Immunomodulators Developed from an Unstable Bacterial Metabolite of Vitamin B2 Biosynthesis. Angew Chem Int Ed Engl 2024; 63:e202400632. [PMID: 38679861 DOI: 10.1002/anie.202400632] [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: 01/10/2024] [Revised: 04/22/2024] [Accepted: 04/26/2024] [Indexed: 05/01/2024]
Abstract
Bacterial synthesis of vitamin B2 generates a by-product, 5-(2-oxopropylideneamino)-d-ribityl-aminouracil (5-OP-RU), with potent immunological properties in mammals, but it is rapidly degraded in water. This natural product covalently bonds to the key immunological protein MR1 in the endoplasmic reticulum of antigen presenting cells (APCs), enabling MR1 refolding and trafficking to the cell surface, where it interacts with T cell receptors (TCRs) on mucosal associated invariant T lymphocytes (MAIT cells), activating their immunological and antimicrobial properties. Here, we strategically modify this natural product to understand the molecular basis of its recognition by MR1. This culminated in the discovery of new water-stable compounds with extremely powerful and distinctive immunological functions. We report their capacity to bind MR1 inside APCs, triggering its expression on the cell surface (EC50 17 nM), and their potent activation (EC50 56 pM) or inhibition (IC50 80 nM) of interacting MAIT cells. We further derivatize compounds with diazirine-alkyne, biotin, or fluorophore (Cy5 or AF647) labels for detecting, monitoring, and studying cellular MR1. Computer modeling casts new light on the molecular mechanism of activation, revealing that potent activators are first captured in a tyrosine- and serine-lined cleft in MR1 via specific pi-interactions and H-bonds, before more tightly attaching via a covalent bond to Lys43 in MR1. This chemical study advances our molecular understanding of how bacterial metabolites are captured by MR1, influence cell surface expression of MR1, interact with T cells to induce immunity, and offers novel clues for developing new vaccine adjuvants, immunotherapeutics, and anticancer drugs.
Collapse
Affiliation(s)
- 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, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Ryan J D Rivero
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Huy N Hoang
- Centre for Chemistry and Drug Discovery and ARC Centre of Excellence for Innovations in Peptide and Protein Science Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, 4072, Australia
| | - Xin Yi Lim
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Jieru Deng
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Hamish E G McWilliam
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Jose A Villadangos
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
- Department of Biochemistry and Pharmacology Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, 3010, Australia
| | - James McCluskey
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, Australia
| | - Alexandra J Corbett
- Department of Microbiology and Immunology, The University of Melbourne, at the Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, 3000, 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, The University of Queensland, Brisbane, Queensland, 4072, Australia
| |
Collapse
|
34
|
Chen S, Wu X, Yang Y, Xu X, Xiong X, Meng W. Increased pathogenicity and pro-inflammatory capabilities of mucosal-associated invariant T cells involved in Oral Lichen Planus. BMC Oral Health 2024; 24:829. [PMID: 39039547 PMCID: PMC11264365 DOI: 10.1186/s12903-024-04621-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024] Open
Abstract
BACKGROUND Mucosal-associated invariant T (MAIT) cells assume pivotal roles in numerous autoimmune inflammatory maladies. However, scant knowledge exists regarding their involvement in the pathological progression of oral lichen planus (OLP). The focus of our study was to explore whether MAIT cells were altered across distinct clinical types of OLP. METHODS The frequency, phenotype, and partial functions of MAIT cells were performed by flow cytometry, using peripheral blood from 18 adults with non-erosive OLP and 22 adults with erosive OLP compared with 15 healthy adults. We also studied the changes in MAIT cells in 15 OLP patients receiving and 10 not receiving corticosteroids. Surface proteins including CD4, CD8, CD69, CD103, CD38, HLA-DR, Tim-3, Programmed Death Molecule-1 (PD-1), and related factors released by MAIT cells such as Granzyme B (GzB), interferon (IFN)-γ, tumour necrosis factor (TNF)-α, interleukin (IL)-17A, and IL-22 were detected. RESULTS Within non-erosive OLP patients, MAIT cells manifested an activated phenotype, evident in an elevated frequency of CD69+ CD38+ MAIT cells (p < 0.01). Conversely, erosive OLP patients displayed an activation and depletion phenotype in MAIT cells, typified by elevated CD69 (p < 0.01), CD103 (p < 0.05), and PD-1 expression (p < 0.01). Additionally, MAIT cells exhibited heightened cytokine production, encompassing GzB, IFN-γ, and IL-17A in erosive OLP patients. Notably, the proportion of CD103+ MAIT cells (p < 0.05) and GzB secretion (p < 0.01) by MAIT cells diminished, while the proportion of CD8+ MAIT cells (p < 0.05) rose in OLP patients with corticosteroid therapy. CONCLUSIONS MAIT cells exhibit increased pathogenicity and pro-inflammatory capabilities in OLP. Corticosteroid therapy influences the expression of certain phenotypes and functions of MAIT cells in the peripheral blood of OLP patients.
Collapse
Affiliation(s)
- Siting Chen
- Departments of Oral Medicine, Stomatological Hospital, School of Stomatology, Southern Medical University NO.366, Jiangnan Road, Guangzhou, Guangdong Province, 510280, P.R. China
| | - Xiaoli Wu
- Departments of Oral Medicine, Stomatological Hospital, School of Stomatology, Southern Medical University NO.366, Jiangnan Road, Guangzhou, Guangdong Province, 510280, P.R. China
| | - Yinshen Yang
- Departments of Oral Medicine, Stomatological Hospital, School of Stomatology, Southern Medical University NO.366, Jiangnan Road, Guangzhou, Guangdong Province, 510280, P.R. China
| | - Xiaoheng Xu
- Departments of Oral Medicine, Stomatological Hospital, School of Stomatology, Southern Medical University NO.366, Jiangnan Road, Guangzhou, Guangdong Province, 510280, P.R. China
| | - Xiaoqin Xiong
- Departments of Oral Medicine, Stomatological Hospital, School of Stomatology, Southern Medical University NO.366, Jiangnan Road, Guangzhou, Guangdong Province, 510280, P.R. China
| | - Wenxia Meng
- Departments of Oral Medicine, Stomatological Hospital, School of Stomatology, Southern Medical University NO.366, Jiangnan Road, Guangzhou, Guangdong Province, 510280, P.R. China.
| |
Collapse
|
35
|
Gao M, Zhao X. Insights into the tissue repair features of MAIT cells. Front Immunol 2024; 15:1432651. [PMID: 39086492 PMCID: PMC11289772 DOI: 10.3389/fimmu.2024.1432651] [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: 05/14/2024] [Accepted: 07/04/2024] [Indexed: 08/02/2024] Open
Abstract
Mucosa-associated invariant T (MAIT) cells are a subset of innate-like non-conventional T cells characterized by multifunctionality. In addition to their well-recognized antimicrobial activity, increasing attention is being drawn towards their roles in tissue homeostasis and repair. However, the precise mechanisms underlying these functions remain incompletely understood and are still subject to ongoing exploration. Currently, it appears that the tissue localization of MAIT cells and the nature of the diseases or stimuli, whether acute or chronic, may induce a dynamic interplay between their pro-inflammatory and anti-inflammatory, or pathogenic and reparative functions. Therefore, elucidating the conditions and mechanisms of MAIT cells' reparative functions is crucial for fully maximizing their protective effects and advancing future MAIT-related therapies. In this review, we will comprehensively discuss the establishment and potential mechanisms of their tissue repair functions as well as the translational application prospects and current challenges in this field.
Collapse
Affiliation(s)
- Mengge Gao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
| | - Xiaosu Zhao
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Medical Sciences, Beijing, China
- Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| |
Collapse
|
36
|
Brown AJ, White J, Shaw L, Gross J, Slabodkin A, Kushner E, Greiff V, Matsuda J, Gapin L, Scott-Browne J, Kappler J, Marrack P. MHC heterozygosity limits T cell receptor variability in CD4 T cells. Sci Immunol 2024; 9:eado5295. [PMID: 38996008 DOI: 10.1126/sciimmunol.ado5295] [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: 02/07/2024] [Accepted: 06/20/2024] [Indexed: 07/14/2024]
Abstract
αβ T cell receptor (TCR) V(D)J genes code for billions of TCR combinations. However, only some appear on peripheral T cells in any individual because, to mature, thymocytes must react with low affinity but not high affinity with thymus expressed major histocompatibility (MHC)/peptides. MHC proteins are very polymorphic. Different alleles bind different peptides. Therefore, any individual might express many different MHC alleles to ensure that some peptides from an invader are bound to MHC and activate T cells. However, most individuals express limited numbers of MHC alleles. To explore this, we compared the TCR repertoires of naïve CD4 T cells in mice expressing one or two MHC alleles. Unexpectedly, the TCRs in heterozygotes were less diverse that those in the sum of their MHC homozygous relatives. Our results suggest that thymus negative selection cancels out the advantages of increased thymic positive selection in the MHC heterozygotes.
Collapse
MESH Headings
- Animals
- Mice
- CD4-Positive T-Lymphocytes/immunology
- Heterozygote
- Major Histocompatibility Complex/immunology
- Major Histocompatibility Complex/genetics
- Mice, Inbred C57BL
- Mice, Transgenic
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Thymus Gland/immunology
Collapse
Affiliation(s)
- Alexander J Brown
- Department of Immunology and Genomic Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - Janice White
- Department of Immunology and Genomic Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Laura Shaw
- Department of Immunology and Genomic Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Jimmy Gross
- Department of Immunology and Genomic Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Andrei Slabodkin
- Department of Immunology, University of Oslo and Oslo University Hospital, Postboks 4950 Nydalen OUS HF Rikshospitalet, 0424 Oslo, Norway
| | - Ella Kushner
- Department of Immunology and Genomic Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Victor Greiff
- Department of Immunology, University of Oslo and Oslo University Hospital, Postboks 4950 Nydalen OUS HF Rikshospitalet, 0424 Oslo, Norway
| | - Jennifer Matsuda
- Department of Immunology and Genomic Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
| | - Laurent Gapin
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - James Scott-Browne
- Department of Immunology and Genomic Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| | - John Kappler
- Department of Immunology and Genomic Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
- Barbara Davis Center for Childhood Diabetes, University of Colorado Anschutz Medical Campus, 1775 Aurora Ct, Aurora, CO 80045, USA
| | - Philippa Marrack
- Department of Immunology and Genomic Medicine, National Jewish Health, 1400 Jackson St, Denver, CO 80206, USA
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, 12800 East 19th Avenue, Aurora, CO 80045, USA
| |
Collapse
|
37
|
Tian K, Jing D, Lan J, Lv M, Wang T. Commensal microbiome and gastrointestinal mucosal immunity: Harmony and conflict with our closest neighbor. Immun Inflamm Dis 2024; 12:e1316. [PMID: 39023417 PMCID: PMC11256888 DOI: 10.1002/iid3.1316] [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: 01/31/2024] [Revised: 05/06/2024] [Accepted: 06/03/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND The gastrointestinal tract contains a wide range of microorganisms that have evolved alongside the immune system of the host. The intestinal mucosa maintains balance within the intestines by utilizing the mucosal immune system, which is controlled by the complex gut mucosal immune network. OBJECTIVE This review aims to comprehensively introduce current knowledge of the gut mucosal immune system, focusing on its interaction with commensal bacteria. RESULTS The gut mucosal immune network includes gut-associated lymphoid tissue, mucosal immune cells, cytokines, and chemokines. The connection between microbiota and the immune system occurs through the engagement of bacterial components with pattern recognition receptors found in the intestinal epithelium and antigen-presenting cells. This interaction leads to the activation of both innate and adaptive immune responses. The interaction between the microbial community and the host is vital for maintaining the balance and health of the host's mucosal system. CONCLUSION The gut mucosal immune network maintains a delicate equilibrium between active immunity, which defends against infections and damaging non-self antigens, and immunological tolerance, which allows for the presence of commensal microbiota and dietary antigens. This balance is crucial for the maintenance of intestinal health and homeostasis. Disturbance of gut homeostasis leads to enduring or severe gastrointestinal ailments, such as colorectal cancer and inflammatory bowel disease. Utilizing these factors can aid in the development of cutting-edge mucosal vaccines that have the ability to elicit strong protective immune responses at the primary sites of pathogen invasion.
Collapse
Affiliation(s)
- Kexin Tian
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| | - Dehong Jing
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| | - Junzhe Lan
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| | - Mingming Lv
- Department of BreastWomen's Hospital of Nanjing Medical University, Nanjing Maternity, and Child Health Care HospitalNanjingChina
| | - Tingting Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
- Jiangsu Key Laboratory of Molecular Medicine, Division of Immunology, Medical SchoolNanjing UniversityNanjingChina
| |
Collapse
|
38
|
Pyaram K, Chang CH. NKT Cells and Other Innate T Cells: The Immune Cells That Do Not Follow the Rules. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:3-5. [PMID: 38885470 DOI: 10.4049/jimmunol.2400243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 05/01/2024] [Indexed: 06/20/2024]
Abstract
This Pillars of Immunology article is a commentary on “A subset of CD4+ thymocytes selected by MHC class I molecules,” a pivotal article by A. Bendelac, N. Killeen, D.R. Littman, and R.H. Schwartz published in Science in 1994, marking the discovery of NKT cells and paving the way for the identification and characterization of other innate T cells. https://doi.org/10.1126/science.7907820.
Collapse
Affiliation(s)
- Kalyani Pyaram
- Department of Cancer Biology, University of Kansas Medical Center, Kansas City, KS
| | | |
Collapse
|
39
|
Yang AYP, Wistuba-Hamprecht K, Greten TF, Ruf B. Innate-like T cells in liver disease. Trends Immunol 2024; 45:535-548. [PMID: 38879436 DOI: 10.1016/j.it.2024.05.008] [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: 04/28/2024] [Revised: 05/27/2024] [Accepted: 05/27/2024] [Indexed: 07/14/2024]
Abstract
Mammalian innate-like T cells (ILTCs), including mucosal-associated invariant T (MAIT), natural killer T (NKT), and γδ T cells, are abundant tissue-resident lymphocytes that have recently emerged as orchestrators of hepatic inflammation, tissue repair, and immune homeostasis. This review explores the involvement of different ILTC subsets in liver diseases. We explore the mechanisms underlying the pro- and anti-inflammatory effector functions of ILTCs in a context-dependent manner. We highlight latest findings regarding the dynamic interplay between ILTC functional subsets and other immune and parenchymal cells which may inform candidate immunomodulatory strategies to achieve improved clinical outcomes in liver diseases. We present new insights into how distinct gene expression programs in hepatic ILTCs are induced, maintained, and reprogrammed in a context- and disease stage-dependent manner.
Collapse
Affiliation(s)
- Albert Ying-Po Yang
- Department of Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany; M3 Research Center for Malignome, Metabolome, and Microbiome, Faculty of Medicine, University of Tübingen, Tübingen, Germany
| | - Kilian Wistuba-Hamprecht
- Department of Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany; M3 Research Center for Malignome, Metabolome, and Microbiome, Faculty of Medicine, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) - Image-Guided and Functionally Instructed Tumor Therapies, University of Tübingen, Tübingen, Germany; Skin Cancer Unit, German Cancer Research Center (DKFZ), Heidelberg, Germany; Department of Dermatology, Venereology, and Allergology, University Medical Center Mannheim, Ruprecht-Karl University of Heidelberg, Mannheim, Germany; DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Tim F Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Center for Cancer Research (CCR) Liver Cancer Program, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin Ruf
- Department of Internal Medicine I, University Hospital Tübingen, Eberhard Karls University of Tübingen, Tübingen, Germany; M3 Research Center for Malignome, Metabolome, and Microbiome, Faculty of Medicine, University of Tübingen, Tübingen, Germany; Cluster of Excellence iFIT (EXC 2180) - Image-Guided and Functionally Instructed Tumor Therapies, University of Tübingen, Tübingen, Germany.
| |
Collapse
|
40
|
El Morr Y, Fürstenheim M, Mestdagh M, Franciszkiewicz K, Salou M, Morvan C, Dupré T, Vorobev A, Jneid B, Premel V, Darbois A, Perrin L, Mondot S, Colombeau L, Bugaut H, du Halgouet A, Richon S, Procopio E, Maurin M, Philippe C, Rodriguez R, Lantz O, Legoux F. MAIT cells monitor intestinal dysbiosis and contribute to host protection during colitis. Sci Immunol 2024; 9:eadi8954. [PMID: 38905325 PMCID: PMC7616241 DOI: 10.1126/sciimmunol.adi8954] [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: 05/25/2023] [Accepted: 05/29/2024] [Indexed: 06/23/2024]
Abstract
Intestinal inflammation shifts microbiota composition and metabolism. How the host monitors and responds to such changes remains unclear. Here, we describe a protective mechanism by which mucosal-associated invariant T (MAIT) cells detect microbiota metabolites produced upon intestinal inflammation and promote tissue repair. At steady state, MAIT ligands derived from the riboflavin biosynthesis pathway were produced by aerotolerant bacteria residing in the colonic mucosa. Experimental colitis triggered luminal expansion of riboflavin-producing bacteria, leading to increased production of MAIT ligands. Modulation of intestinal oxygen levels suggested a role for oxygen in inducing MAIT ligand production. MAIT ligands produced in the colon rapidly crossed the intestinal barrier and activated MAIT cells, which expressed tissue-repair genes and produced barrier-promoting mediators during colitis. Mice lacking MAIT cells were more susceptible to colitis and colitis-driven colorectal cancer. Thus, MAIT cells are sensitive to a bacterial metabolic pathway indicative of intestinal inflammation.
Collapse
Affiliation(s)
- Yara El Morr
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | - Mariela Fürstenheim
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
- Université Paris Cité, Paris, France
| | - Martin Mestdagh
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | | | - Marion Salou
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | - Claire Morvan
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, F-75015Paris, France
| | - Thierry Dupré
- Laboratoire de Biochimie, Hôpital Bichat AP-HP, Université de Paris, Paris, France
| | - Alexey Vorobev
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | - Bakhos Jneid
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | - Virginie Premel
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | - Aurélie Darbois
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | - Laetitia Perrin
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | - Stanislas Mondot
- Institut Micalis, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Ludovic Colombeau
- CNRS UMR 3666, INSERM U1143, Chemical Biology of Cancer Laboratory, PSL University, Institut Curie, 75005Paris, France
| | - Hélène Bugaut
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | | | - Sophie Richon
- Institut Curie, PSL Research University, CNRS UMR144, Paris, France
| | - Emanuele Procopio
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | - Mathieu Maurin
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
| | - Catherine Philippe
- Institut Micalis, INRAE, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Raphael Rodriguez
- CNRS UMR 3666, INSERM U1143, Chemical Biology of Cancer Laboratory, PSL University, Institut Curie, 75005Paris, France
| | - Olivier Lantz
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
- Laboratoire d’immunologie clinique, Institut Curie, 75005Paris, France
- Centre d’investigation Clinique en Biothérapie Gustave-Roussy Institut Curie (CIC-BT1428), Paris, France
| | - François Legoux
- Institut Curie, PSL University, Inserm U932, Immunity and Cancer, Paris, France
- INSERM ERL1305, CNRS UMR6290, Université de Rennes, Institut de Génétique & Développement de Rennes, Rennes, France
| |
Collapse
|
41
|
Zheng Y, Han F, Wu Z, Wang B, Chen X, Boulouis C, Jiang Y, Ho A, He D, Sia WR, Mak JYW, Fairlie DP, Wang LF, Sandberg JK, Lobie PE, Ma S, Leeansyah E. MAIT cell activation and recruitment in inflammation and tissue damage in acute appendicitis. SCIENCE ADVANCES 2024; 10:eadn6331. [PMID: 38865451 PMCID: PMC11168461 DOI: 10.1126/sciadv.adn6331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 05/08/2024] [Indexed: 06/14/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells are antimicrobial T cells abundant in the gut, but mechanisms for their migration into tissues during inflammation are poorly understood. Here, we used acute pediatric appendicitis (APA), a model of acute intestinal inflammation, to examine these migration mechanisms. MAIT cells were lower in numbers in circulation of patients with APA but were enriched in the inflamed appendix with increased production of proinflammatory cytokines. Using the patient-derived appendix organoid (PDAO) model, we found that circulating MAIT cells treated with inflammatory cytokines elevated in APA up-regulated chemokine receptors, including CCR1, CCR3, and CCR4. They exhibited enhanced infiltration of Escherichia coli-pulsed PDAO in a CCR1-, CCR2-, and CCR4-dependent manner. Close interactions of MAIT cells with infected organoids led to the PDAO structural destruction and death. These findings reveal a previously unidentified mechanism of MAIT cell tissue homing, their participation in tissue damage in APA, and their intricate relationship with mucosal tissues during acute intestinal inflammation in humans.
Collapse
Affiliation(s)
- Yichao Zheng
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Precision Medicine and Healthcare Research Centre, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen 518055, China
| | - Fei Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Zhengyu Wu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Bingjie Wang
- Department of Pediatric Surgery, Zhangzhou Affiliated Hospital of Fujian Medical University, Zhangzhou 363000, China
| | - Xingchi Chen
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Caroline Boulouis
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Yuebin Jiang
- Department of Pathology, Zhangzhou Municipal Hospital of Fujian Province, Zhangzhou 363000, China
| | - Amanda Ho
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Precision Medicine and Healthcare Research Centre, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen 518055, China
| | - Dan He
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Precision Medicine and Healthcare Research Centre, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen 518055, China
| | - Wan Rong Sia
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - 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, 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, The University of Queensland, Brisbane, Queensland, Australia
| | - Lin-Fa Wang
- Programme in Emerging Infectious Diseases, Duke-National University of Singapore Medical School, Singapore, Singapore
| | - Johan K. Sandberg
- Center for Infectious Medicine, Department of Medicine, Karolinska Institutet, 14152 Stockholm, Sweden
| | - Peter E. Lobie
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Precision Medicine and Healthcare Research Centre, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen 518055, China
| | - Shaohua Ma
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Precision Medicine and Healthcare Research Centre, Tsinghua-Berkeley Shenzhen Institute, Tsinghua University, Shenzhen 518055, China
| | - Edwin Leeansyah
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| |
Collapse
|
42
|
Karlova Zubata I, Smetanova Brozova J, Karel T, Bacova B, Novak J. High pre-transplant Mucosal Associated Invariant T Cell (MAIT) count predicts favorable course of myeloid aplasia. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2024; 168:139-146. [PMID: 36896825 DOI: 10.5507/bp.2023.011] [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: 09/07/2022] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
AIMS Mucosal Associated Invariant T (MAIT) cells are unconventional T cells with anti-infective potential. MAIT cells detect and fight against microbes on mucosal surfaces and in peripheral tissues. Previous works suggested that MAIT cells survive exposure to cytotoxic drugs in these locations. We sought to determine if they maintain their anti-infective functions after myeloablative chemotherapy. METHODS We correlated the amount of MAIT cells (measured by flow cytometry) in the peripheral blood of 100 adult patients before the start of myeloablative conditioning plus autologous stem cell transplantation with the clinical and laboratory outcomes of aplasia. RESULTS The amount of MAIT cells negatively correlated with peak C-reactive protein level and the amount of red blood cell transfusion units resulting in earlier discharge of patients with the highest amount of MAIT cells. CONCLUSION This work suggests the anti-infectious potential of MAIT cells is maintained during myeloid aplasia.
Collapse
Affiliation(s)
| | - Jitka Smetanova Brozova
- Central Laboratories of the Faculty Hospital Kralovske Vinohrady, Srobarova 50, 100 34, Prague 10, Czech Republic
| | - Tomas Karel
- Department of Statistics and Probability, Faculty of Informatics and Statistics, University of Economics and Business in Prague, Namesti W. Churchilla 1938/4, 130 67, Prague 3, Czech Republic
| | - Barbora Bacova
- Central Laboratories of the Faculty Hospital Kralovske Vinohrady, Srobarova 50, 100 34, Prague 10, Czech Republic
- Department of Immunology, 3rd Faculty of Medicine, Charles University, Ruska 87, 100 00, Prague 10, Czech Republic
| | - Jan Novak
- Department of Haematology, 3
- Department of Immunology, 3rd Faculty of Medicine, Charles University, Ruska 87, 100 00, Prague 10, Czech Republic
| |
Collapse
|
43
|
Siblany L, Stocker N, Ricard L, Brissot E, Duléry R, Banet A, Sestili S, Belhocine R, Van de Wyngaert Z, Bonnin A, Capes A, Ledraa T, Beurier P, Fadel K, Mohty M, Gaugler B, Malard F. Unconventional T Cells Influence Clinical Outcome After Allogeneic Hematopoietic Cell Transplantation. J Clin Immunol 2024; 44:139. [PMID: 38822857 DOI: 10.1007/s10875-024-01741-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 05/20/2024] [Indexed: 06/03/2024]
Abstract
We evaluated the impact of early recovery of mucosal-associated invariant T cells (MAIT) and gamma-delta (γδ) T cells, especially Vδ2+ T cells, on the clinical outcomes of 76 patients who underwent allogeneic hematopoietic cell transplantation (allo-HCT). MAIT cells were identified at day 20-30 post-transplant using flow cytometry and defined as CD3+ TCRVα7.2+CD161+. Two subsets of Vδ2+ T cells were analyzed according to the expression of CD26. The cytotoxicity profile of MAIT and Vδ2+ T cells was analyzed according to the intracellular expression of perforin and granzyme B, and intracellular IFN-γ was evaluated after in vitro activation. CD26+Vδ2+ T cells displayed higher intracellular levels of IFN-γ, whereas CD26- Vδ2+ T were found to be more cytotoxic. Moreover, MAIT cell frequency was correlated with the frequency of Vδ2+ T cells with a better correlation observed with Vδ2+CD26+ than with the Vδ2+CD26- T cell subset. By using the composite endpoint graft-versus-host disease (GvHD)-free, relapse-free survival (GRFS) as the primary endpoint, we found that patients with a higher MAIT cell frequency at day 20-30 after allo-HCT had a significantly increased GRFS and a better overall survival (OS) and disease-free survival (DFS). Moreover, patients with a low CD69 expression by MAIT cells had an increased cumulative incidence of grade 2-4 acute GvHD (aGvHD). These results suggest that MAIT cell reconstitution may provide mitigating effects early after allo-HCT depending on their activation markers and functional status. Patients with a high frequency of Vδ2+CD26+ T cells had a significantly higher GRFS, OS and DFS, but there was no impact on cumulative incidence of grade 2-4 aGVHD, non-relapse mortality and relapse. These results revealed that the impact of Vδ2+ T cells on the success of allo-HCT may vary according to the frequency of the CD26+ subset.
Collapse
Affiliation(s)
- Lama Siblany
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Nicolas Stocker
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Laure Ricard
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Eolia Brissot
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Rémy Duléry
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Anne Banet
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Simona Sestili
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Ramdane Belhocine
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Zoé Van de Wyngaert
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Agnès Bonnin
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Antoine Capes
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Tounes Ledraa
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Pauline Beurier
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
| | - Karen Fadel
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Mohamad Mohty
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Béatrice Gaugler
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France
| | - Florent Malard
- INSERM, Centre de Recherche Saint-Antoine (CRSA), Sorbonne Université, F-75012, Paris, France.
- AP-HP, Hôpital Saint-Antoine, Service d'Hématologie Clinique et Thérapie Cellulaire, Sorbonne Université, F-75012, Paris, France.
| |
Collapse
|
44
|
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] [Grants] [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
|
45
|
Zhang B, Chen P, Zhu J, Lu Y. The quantity, function and anti-tumor effect of Mucosal associated invariant T cells in patients with bladder cancer. Int Immunopharmacol 2024; 133:111892. [PMID: 38663315 DOI: 10.1016/j.intimp.2024.111892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND Bladder cancer (BC), a prevalent malignancy in the urinary system, often poses challenges for effective treatment. Immunotherapy, harnessing the immune system, has exhibited promise in early-stage clinical trials. Mucosal associated invariant T (MAIT) cells, a subset of immune cells implicated in various diseases, including certain cancer, have yet to be explored in BC patients. We aimed to investigate the quantity, function, and anti-tumor effects of MAIT cells in BC patients. METHODS A total of 75 newly diagnosed BC patients and 183 healthy volunteers were included. Blood samples were collected and analyzed to evaluate the quantity and function of MAIT cells. Surgical resection provided BC tissues for further analysis, and the clinical features of BC tumors were collected and their relationship with MAIT cells was explored. RESULTS MAIT cells were identified in both healthy individuals and BC patients. The proportion of MAIT cells in the peripheral blood of BC patients did not significantly differ from that of healthy controls. However, the study revealed a correlation between the proportion of IFN-γ producing MAIT cells and tumor number and invasion in BC patients. Furthermore, MAIT cells exhibited cytotoxic effects on BC cells in vitro and in vivo. CONCLUSIONS This study sheds light on the role of MAIT cells in BC. While the quantity of MAIT cells showed no significant change in BC patients, their functional attributes and association with tumor characteristics suggest their potential as an immunotherapy target in BC treatment.
Collapse
Affiliation(s)
- Baodan Zhang
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Pengcheng Chen
- The Center for Microbes, Development and Health, Key Laboratory of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai 200031, China
| | - Jie Zhu
- Department of Psychiatry, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yongyong Lu
- Department of Urology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
46
|
Wyatt-Johnson SK, Kersey HN, Brutkiewicz RR. Enrichment of liver MAIT cells in a mouse model of Alzheimer's disease. J Neuroimmunol 2024; 390:578332. [PMID: 38537322 PMCID: PMC11382344 DOI: 10.1016/j.jneuroim.2024.578332] [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: 01/30/2024] [Revised: 03/14/2024] [Accepted: 03/20/2024] [Indexed: 05/13/2024]
Abstract
Emerging evidence has supported a role for the immune system and liver in Alzheimer's disease (AD). However, our understanding of how hepatic immune cells are altered in AD is limited. We previously found that brain mucosal-associated invariant T (MAIT) cell numbers are increased in AD. Furthermore, loss of MAIT cells and their antigen-presenting molecule, MR1, reduced amyloid-β accumulation in the brain. MAIT cells are also significantly present in the liver. Therefore, we sought to analyze MAIT and other immune cells in the AD liver. Increased frequency of activated MAIT cells (but not conventional T cells) were found in 8-month-old 5XFAD mouse livers. Therefore, these data raise the possibility that there is a role for peripheral MAIT cells in AD pathology.
Collapse
Affiliation(s)
- Season K Wyatt-Johnson
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America.
| | - Holly N Kersey
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America
| | - Randy R Brutkiewicz
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, United States of America.
| |
Collapse
|
47
|
Sobel AL, Melamed J, Haas D, LeBlanc G, Cirone A, Constantinides MG. Antibiotic use in early life subsequently impairs MAIT cell-mediated immunity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.10.593643. [PMID: 38798453 PMCID: PMC11118404 DOI: 10.1101/2024.05.10.593643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells are predominantly located in barrier tissues where they rapidly respond to pathogens and commensals by recognizing microbial derivatives of riboflavin synthesis. Early-life exposure to these metabolites imprints the abundance of MAIT cells within tissues, so we hypothesized that antibiotic use during this period may abrogate their development. We identified antibiotics that deplete riboflavin-synthesizing commensals and revealed an early period of susceptibility during which antibiotic administration impaired MAIT cell development. The reduction in MAIT cell abundance rendered mice more susceptible to pneumonia, while MAIT cell-deficient mice were unaffected by early-life antibiotics. Concomitant administration of a riboflavin-synthesizing commensal during antibiotic treatment was sufficient to restore MAIT cell development and immunity. Our work demonstrates that transient depletion of riboflavin-synthesizing commensals in early life can adversely affect responses to subsequent infections.
Collapse
|
48
|
Vacchini A, Chancellor A, Yang Q, Colombo R, Spagnuolo J, Berloffa G, Joss D, Øyås O, Lecchi C, De Simone G, Beshirova A, Nosi V, Loureiro JP, Morabito A, De Gregorio C, Pfeffer M, Schaefer V, Prota G, Zippelius A, Stelling J, Häussinger D, Brunelli L, Villalta P, Lepore M, Davoli E, Balbo S, Mori L, De Libero G. Nucleobase adducts bind MR1 and stimulate MR1-restricted T cells. Sci Immunol 2024; 9:eadn0126. [PMID: 38728413 DOI: 10.1126/sciimmunol.adn0126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 04/18/2024] [Indexed: 05/12/2024]
Abstract
MR1T cells are a recently found class of T cells that recognize antigens presented by the major histocompatibility complex-I-related molecule MR1 in the absence of microbial infection. The nature of the self-antigens that stimulate MR1T cells remains unclear, hampering our understanding of their physiological role and therapeutic potential. By combining genetic, pharmacological, and biochemical approaches, we found that carbonyl stress and changes in nucleobase metabolism in target cells promote MR1T cell activation. Stimulatory compounds formed by carbonyl adducts of nucleobases were detected within MR1 molecules produced by tumor cells, and their abundance and antigenicity were enhanced by drugs that induce carbonyl accumulation. Our data reveal carbonyl-nucleobase adducts as MR1T cell antigens. Recognizing cells under carbonyl stress allows MR1T cells to monitor cellular metabolic changes with physiological and therapeutic implications.
Collapse
Affiliation(s)
- Alessandro Vacchini
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Andrew Chancellor
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Qinmei Yang
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Rodrigo Colombo
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Julian Spagnuolo
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Giuliano Berloffa
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Daniel Joss
- Department of Chemistry, University of Basel, Basel 4056, Switzerland
| | - Ove Øyås
- Department of Biosystems Science and Engineering and SIB Swiss Institute of Bioinformatics, ETH Zurich, Basel 4058, Switzerland
| | - Chiara Lecchi
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Giulia De Simone
- Department of Environmental Health Science, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano 20156, Italy
| | - Aisha Beshirova
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Vladimir Nosi
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - José Pedro Loureiro
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Aurelia Morabito
- Department of Environmental Health Science, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano 20156, Italy
| | - Corinne De Gregorio
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Michael Pfeffer
- Department of Chemistry, University of Basel, Basel 4056, Switzerland
| | - Verena Schaefer
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Gennaro Prota
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Alfred Zippelius
- Cancer Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Jörg Stelling
- Department of Biosystems Science and Engineering and SIB Swiss Institute of Bioinformatics, ETH Zurich, Basel 4058, Switzerland
| | - Daniel Häussinger
- Department of Chemistry, University of Basel, Basel 4056, Switzerland
| | - Laura Brunelli
- Department of Environmental Health Science, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano 20156, Italy
| | - Peter Villalta
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Medicinal Chemistry, University of Minnesota, Minneapolis, MN 55455, USA
| | - Marco Lepore
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Enrico Davoli
- Department of Environmental Health Science, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milano 20156, Italy
| | - Silvia Balbo
- Masonic Cancer Center, University of Minnesota, Minneapolis, MN 55455, USA
| | - Lucia Mori
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| | - Gennaro De Libero
- Experimental Immunology, Department of Biomedicine, University Hospital and University of Basel, Basel 4031, Switzerland
| |
Collapse
|
49
|
Ignacio A, Czyz S, McCoy KD. Early life microbiome influences on development of the mucosal innate immune system. Semin Immunol 2024; 73:101885. [PMID: 38788491 DOI: 10.1016/j.smim.2024.101885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 05/11/2024] [Accepted: 05/16/2024] [Indexed: 05/26/2024]
Abstract
The gut microbiota is well known to possess immunomodulatory capacities, influencing a multitude of cellular signalling pathways to maintain host homeostasis. Although the formation of the immune system initiates before birth in a sterile environment, an emerging body of literature indicates that the neonatal immune system is influenced by a first wave of external stimuli that includes signals from the maternal microbiota. A second wave of stimulus begins after birth and must be tightly regulated during the neonatal period when colonization of the host occurs concomitantly with the maturation of the immune system, requiring a fine adjustment between establishing tolerance towards the commensal microbiota and preserving inflammatory responses against pathogenic invaders. Besides integrating cues from commensal microbes, the neonatal immune system must also regulate responses triggered by other environmental signals, such as dietary antigens, which become more complex with the introduction of solid food during the weaning period. This "window of opportunity" in early life is thought to be crucial for the proper development of the immune system, setting the tone of subsequent immune responses in adulthood and modulating the risk of developing chronic and metabolic inflammatory diseases. Here we review the importance of host-microbiota interactions for the development and maturation of the immune system, particularly in the early-life period, highlighting the known mechanisms involved in such communication. This discussion is focused on recent data demonstrating microbiota-mediated education of innate immune cells and its role in the development of lymphoid tissues.
Collapse
Affiliation(s)
- Aline Ignacio
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Sonia Czyz
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kathy D McCoy
- Department of Physiology and Pharmacology, Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
50
|
Konecny AJ, Huang Y, Setty M, Prlic M. Signals that control MAIT cell function in healthy and inflamed human tissues. Immunol Rev 2024; 323:138-149. [PMID: 38520075 DOI: 10.1111/imr.13325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/25/2024]
Abstract
Mucosal-associated invariant T (MAIT) cells have a semi-invariant T-cell receptor that allows recognition of antigen in the context of the MHC class I-related (MR1) protein. Metabolic intermediates of the riboflavin synthesis pathway have been identified as MR1-restricted antigens with agonist properties. As riboflavin synthesis occurs in many bacterial species, but not human cells, it has been proposed that the main purpose of MAIT cells is antibacterial surveillance and protection. The majority of human MAIT cells secrete interferon-gamma (IFNg) upon activation, while some MAIT cells in tissues can also express IL-17. Given that MAIT cells are present in human barrier tissues colonized by a microbiome, MAIT cells must somehow be able to distinguish colonization from infection to ensure effector functions are only elicited when necessary. Importantly, MAIT cells have additional functional properties, including the potential to contribute to restoring tissue homeostasis by expression of CTLA-4 and secretion of the cytokine IL-22. A recent study provided compelling data indicating that the range of human MAIT cell functional properties is explained by plasticity rather than distinct lineages. This further underscores the necessity to better understand how different signals regulate MAIT cell function. In this review, we highlight what is known in regards to activating and inhibitory signals for MAIT cells with a specific focus on signals relevant to healthy and inflamed tissues. We consider the quantity, quality, and the temporal order of these signals on MAIT cell function and discuss the current limitations of computational tools to extrapolate which signals are received by MAIT cells in human tissues. Using lessons learned from conventional CD8 T cells, we also discuss how TCR signals may integrate with cytokine signals in MAIT cells to elicit distinct functional states.
Collapse
Affiliation(s)
- Andrew J Konecny
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Yin Huang
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Herbold Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Molecular and Cellular Biology Program, University of Washington, Seattle, Washington, USA
| | - Manu Setty
- Basic Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Herbold Computational Biology Program, Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Martin Prlic
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, Washington, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
| |
Collapse
|