1
|
Liu XR, Li M, Hao QQ, Yu YJ, Liao C, Yu R, Kong DL, Wang Y. Unraveling cysteinyl leukotrienes and their receptors in inflammation through the brain-gut-lung axis. Virulence 2025; 16:2502555. [PMID: 40351036 PMCID: PMC12077450 DOI: 10.1080/21505594.2025.2502555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 04/21/2025] [Accepted: 04/23/2025] [Indexed: 05/14/2025] Open
Abstract
Cysteinyl leukotrienes (CysLTs), as potent lipid inflammatory mediators, play a pivotal role in systemic multi-organ inflammation and inter-organ communication through interactions with their receptors (CysLTRs). However, However, the function of CysLT3R is unclear and lacks a network of cross-organ metabolite interactions, and the clinical use of leukotriene receptor antagonists (LTRAs) has certain limitations. This review systematically synthesizes existing evidence and proposes future directions by clarifying receptor subtype specificity, optimizing targeted therapies, exploring CysLTs' applications in neuroimmunology, and elucidating the dual roles of CysLTs in chronic inflammation. It is indicated that CysLTs activate eosinophils, mast cells, and airway tuft cells, driving type 2 immune responses and mucus secretion in the lungs, thereby exacerbating respiratory diseases such as asthma. In the nervous system, CysLTs aggravate neurodegenerative disorders like cerebral ischemia and Alzheimer's disease by disrupting the blood-brain barrier, promoting glial activation, and inducing neuronal damage. In the gut, CysLTs regulate anti-helminth immunity via the tuft cell-ILC2 pathway and collaborate with prostaglandin D2 (PGD2) to modulate bile excretion and mucosal protection. Furthermore, CysLTs mediate communication through the gut-lung and gut-brain axes via metabolites such as succinate, contributing to cross-organ inflammatory regulation. In conclusion, this review highlights the complex roles of CysLTs in chronic inflammation, providing a theoretical foundation for precise intervention in multi-organ inflammatory diseases, which provides a theoretical framework for precision interventions in multi-organ inflammatory diseases and inspires interdisciplinary breakthroughs.
Collapse
Affiliation(s)
- Xiao-Ru Liu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Ming Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Qian-Qian Hao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Ya-Jie Yu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Cai Liao
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - Rui Yu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| | - De-Lei Kong
- Department of Respiratory and Critical Care Medicine, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
2
|
Kreslavsky T. Thymflammation: The Role of a Constitutively Active Inflammatory Network and "Ectopic" Cell Types in the Thymus in the Induction of T Cell Tolerance and Beyond. Immunol Rev 2025; 332:e70037. [PMID: 40433806 PMCID: PMC12117520 DOI: 10.1111/imr.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/28/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
The thymus exhibits constitutive activation of nearly all major inflammatory pathways, including sterile MyD88-dependent signaling and interferon production by mTECs, the presence of cellular and molecular components of type 1, type 2, and type 3 responses, as well as sustained B cell activation. The reasons for the existence of such a complex constitutively active inflammatory network at the site of T cell development-where the initial pathogen encounter is unlikely-have remained enigmatic. We propose that this inflammatory thymic 'ecosystem' has evolved to promote immunological tolerance to 'inflammatory self'-endogenous molecules absent from most peripheral tissues at steady state but upregulated during pathogen invasion. The spatial and temporal overlap with pathogen presence makes the discrimination of the inflammatory self from pathogen-derived molecules a unique challenge for the adaptive immune system. The frequent occurrence of diseases associated with autoantibodies against proinflammatory cytokines underscores the persistent risk of these molecules being misidentified as foreign. Their abundant representation in the thymus, therefore, is likely to be critical for maintaining tolerance. This review explores current insights into the thymic inflammatory network, its cellular and molecular constituents, and their role in the induction of T cell tolerance.
Collapse
Affiliation(s)
- Taras Kreslavsky
- Division of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska InstitutetKarolinska University HospitalStockholmSweden
- Center for Molecular MedicineKarolinska University HospitalStockholmSweden
| |
Collapse
|
3
|
Liu S, Zheng C, Nechanitzky R, Luo P, Ramachandran P, Nguyen D, Elia AJ, Moghadas Jafari S, Law R, Snow BE, Wakeham AC, Berger T, Chen H, Gill KT, Mcwilliam R, Fortin J, Modares NF, Saunders ME, Murakami K, Qiu Y, You Z, Mohtashami M, Qi H, Ohashi PS, Zúñiga-Pflücker JC, Mak TW. Cholinergic regulation of thymocyte negative selection. Nat Immunol 2025:10.1038/s41590-025-02152-4. [PMID: 40399609 DOI: 10.1038/s41590-025-02152-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 04/04/2025] [Indexed: 05/23/2025]
Abstract
The immune and nervous systems use a common chemical language for communication, namely, the cholinergic signaling involving acetylcholine (ACh) and its receptors (AChRs). Whether and how this language also regulates the development of the immune system is poorly understood. Here, we show that mouse CD4+CD8+ double-positive thymocytes express high levels of α9 nicotinic AChR (nAChR) and that this receptor controls thymic negative selection. α9 nAChR-deficient mice show an altered T cell receptor (TCR) repertoire and reduced CD4+ and CD8+ T cells in a mixed bone marrow chimera setting. α9 nAChR-mediated signaling regulates TCR strength and thymocyte survival. Thymic tuft cells, B cells and some T cells express choline acetyltransferase and are potential ACh sources, with ACh derived from T cells having the most important role. Furthermore, α9 nAChR deficiency during thymocyte development contributes to the altered development of autoimmune diseases in mice. Our results thus reveal a mechanism controlling immune cell development that involves cholinergic signaling.
Collapse
Affiliation(s)
- Shaofeng Liu
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Chunxing Zheng
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Robert Nechanitzky
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Microbiology, Immunology, and Infectious Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ping Luo
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | | | - Dat Nguyen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Andrew J Elia
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Soode Moghadas Jafari
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Rhoda Law
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Bryan E Snow
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Andrew C Wakeham
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Thorsten Berger
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Hui Chen
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China
| | - Kyle T Gill
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Ryan Mcwilliam
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Jerome Fortin
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec, Canada
| | | | - Mary E Saunders
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Kiichi Murakami
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Yangmin Qiu
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Zhiwei You
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Mahmood Mohtashami
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Hai Qi
- School of Basic Medical Sciences, Tsinghua Medicine, Tsinghua University, Beijing, China
| | - Pamela S Ohashi
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Juan Carlos Zúñiga-Pflücker
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- Biological Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Tak W Mak
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada.
- Centre for Oncology and Immunology, Hong Kong Science Park, Hong Kong SAR, China.
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada.
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
4
|
Mohammed A, Wang W, Arreola M, Solomon BD, Slepicka PF, Hubka KM, Nguyen HD, Zheng Z, Chavez MG, Yeh CY, Kim DK, Ma MR, Martin E, Li L, Pasca AM, Winn VD, Gifford CA, Kedlian VR, Park JE, Khatri P, Hollander GA, Roncarolo MG, Sebastiano V, Teichmann SA, Gentles AJ, Weinacht KG. Distinct type I and II interferon responses direct cortical and medullary thymic epithelial cell development. Sci Immunol 2025; 10:eado4720. [PMID: 40315299 DOI: 10.1126/sciimmunol.ado4720] [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: 02/12/2024] [Revised: 12/05/2024] [Accepted: 04/10/2025] [Indexed: 05/04/2025]
Abstract
Advances in genomics have redefined our understanding of thymic epithelial heterogeneity and architecture, yet signals driving thymic epithelial differentiation remain incompletely understood. Here, we elucidated pathways instructing human thymic epithelial cell development in the context of other anterior foregut-derived organs. Activation of interferon response gene regulatory networks distinguished epithelial cells of the thymus from those of other anterior foregut-derived organs. Thymic cortex and medulla epithelia displayed distinctive interferon-responsive signatures defined by lineage-specific chromatin accessibility. We explored the effects of type I and II interferons on thymic epithelial progenitor differentiation from induced pluripotent stem cells. Type II interferon was essential for expressing proteasome and antigen-presenting molecules, whereas type I or II interferons were essential for inducing different cytokines in thymic epithelial progenitor cells. Our findings suggest that interferons are critical to cortical and medullary thymic epithelial cell differentiation.
Collapse
Affiliation(s)
- Abdulvasey Mohammed
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Wenqing Wang
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Martin Arreola
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Benjamin D Solomon
- Department of Pediatrics, Division of Allergy and Immunology, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Priscila F Slepicka
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Kelsea M Hubka
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Hanh Dan Nguyen
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Zihao Zheng
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Michael G Chavez
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Christine Y Yeh
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
- Department of Medicine, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Doo Kyung Kim
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Michael R Ma
- Cardiovascular Institute, Stanford University, Stanford, CA 94305, USA
- Department of Cardiothoracic Surgery, Division of Pediatric Cardiac Surgery, Stanford University, Stanford, CA 94304, USA
| | - Elisabeth Martin
- Department of Cardiothoracic Surgery, Division of Pediatric Cardiac Surgery, Stanford University, Stanford, CA 94304, USA
| | - Li Li
- Department of Pediatrics, Division of Neonatology, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Anca M Pasca
- Department of Pediatrics, Division of Neonatology, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Virginia D Winn
- Department of Obstetrics and Gynecology, Stanford School of Medicine, Stanford, CA, USA
| | - Casey A Gifford
- Department of Pediatrics, Division of Cardiology, Stanford School of Medicine, Stanford, CA 94305, USA
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Veronika R Kedlian
- Wellcome Sanger Institute, Cambridge, UK
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
| | | | - Purvesh Khatri
- Institute for Immunity, Transplantation, and Infection, Stanford University, Stanford, CA 94304, USA
- Center for Biomedical Informatics Research, Department of Medicine, School of Medicine, Stanford University, CA 94305, USA
| | - Georg A Hollander
- Department of Pediatrics and Institute of Developmental and Regenerative Medicine, University of Oxford, Oxford, UK
- Department of Biosystems and Engineering, ETH Zurich, Zurich, Switzerland
- Botnar Institute of Immune Engineering, Basel, Switzerland
| | - Maria Grazia Roncarolo
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| | - Vittorio Sebastiano
- Department of Obstetrics and Gynecology, Stanford School of Medicine, Stanford, CA, USA
| | - Sarah A Teichmann
- Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- CIFAR Macmillan Multi-scale Human Programme, CIFAR, Toronto, Canada
| | - Andrew J Gentles
- Department of Biomedical Data Science, Stanford University, Stanford, CA 94305, USA
- Department of Pathology, Stanford School of Medicine, Stanford, CA 94305, USA
| | - Katja G Weinacht
- Department of Pediatrics, Division of Hematology, Oncology, Stem Cell Transplantation, and Regenerative Medicine, Stanford School of Medicine, Stanford, CA 94304, USA
| |
Collapse
|
5
|
Huisman BD, Zuo C, Mathis D. Thymic Mimetic Cells: Evolutionarily Ancient Mirrors of the Periphery. Immunol Rev 2025; 331:e70028. [PMID: 40247628 PMCID: PMC12067923 DOI: 10.1111/imr.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/03/2025] [Accepted: 04/07/2025] [Indexed: 04/19/2025]
Abstract
Thymic mimetic cells are hybrids of medullary thymic epithelial cells and diverse peripheral cell types. They are important for imposition of self-tolerance and perform other functions similar to those of their peripheral counterparts. Following early histological observations of “misplaced” stromal cells in thymi from multiple species, mimetic cells were first molecularly investigated in mice. Recent studies have characterized mimetic cells in humans and zebrafish with high-resolution. Many mimetic cell types are conserved across species, although specialized subtypes as well as variable frequencies and levels of specialization are also apparent. Features of the human mimetic cell repertoire, such as the expanded nature of muscle mimetic cells with potential implications in myasthenia gravis and the similarity of tuft mimetic cells and thymic carcinomas, hint at their relevance in human disease. Here we review what is known about mimetic cells across diverse organisms. We discuss potential pressures shaping the composition of the mimetic cell repertoire within and across species, and highlight potential therapeutic applications in human disease.
Collapse
Affiliation(s)
| | - Chong Zuo
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
6
|
Carrera Silva EA, Puyssegur J, Errasti AE. Coevolutionary interplay: Helminths-trained immunity and its impact on the rise of inflammatory diseases. eLife 2025; 14:e105393. [PMID: 40231720 PMCID: PMC12002795 DOI: 10.7554/elife.105393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Accepted: 04/01/2025] [Indexed: 04/16/2025] Open
Abstract
The gut biome, a complex ecosystem of micro- and macro-organisms, plays a crucial role in human health. A disruption in this evolutive balance, particularly during early life, can lead to immune dysregulation and inflammatory disorders. 'Biome repletion' has emerged as a potential therapeutic approach, introducing live microbes or helminth-derived products to restore immune balance. While helminth therapy has shown some promise, significant challenges remain in optimizing clinical trials. Factors such as patient genetics, disease status, helminth species, and the optimal timing and dosage of their products or metabolites must be carefully considered to train the immune system effectively. We aim to discuss how helminths and their products induce trained immunity as prospective to treat inflammatory and autoimmune diseases. The molecular repertoire of helminth excretory/secretory products (ESPs), which includes proteins, peptides, lipids, and RNA-carrying extracellular vesicles (EVs), underscores their potential to modulate innate immune cells and hematopoietic stem cell precursors. Mimicking natural delivery mechanisms like synthetic exosomes could revolutionize EV-based therapies and optimizing production and delivery of ESP will be crucial for their translation into clinical applications. By deciphering and harnessing helminth-derived products' diverse modes of action, we can unleash their full therapeutic potential and pave the way for innovative treatments.
Collapse
Affiliation(s)
- Eugenio Antonio Carrera Silva
- EACS and JP Institute of Experimental Medicine, National Scientific and Technical Research Council, National Academy of Medicine (IMEX-CONICET-ANM)Buenos AiresArgentina
| | - Juliana Puyssegur
- EACS and JP Institute of Experimental Medicine, National Scientific and Technical Research Council, National Academy of Medicine (IMEX-CONICET-ANM)Buenos AiresArgentina
| | - Andrea Emilse Errasti
- AEE Institute of Pharmacology, School of Medicine, University of Buenos AiresBuenos AiresArgentina
- National Scientific and Technical Research Council (CONICET)Buenos AiresArgentina
| |
Collapse
|
7
|
Martinez RJ, Hogquist KA. Sterile production of interferons in the thymus. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf048. [PMID: 40184034 DOI: 10.1093/jimmun/vkaf048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Accepted: 02/25/2025] [Indexed: 04/05/2025]
Abstract
T-cell central tolerance is controlled by thymocyte TCR recognition of self-peptides presented by thymic APCs. While thymic epithelial cells are essential for T-cell central tolerance, a variety of other traditional APCs also play critical roles in T-cell selection. Similar to how peripheral APCs require activation to become effective, thymic APCs also require activation to become tolerogenic. Recent studies have identified IFNs as an essential factor for the activation and generation of an optimally tolerogenic thymic environment. In this review, we focus on interferon (IFN) production within the thymus and its effects on thymic APCs and developing thymocytes. We also examine the importance of T-cell tolerance to IFN itself as well as to interferon-stimulated proteins generated during peripheral immune responses.
Collapse
Affiliation(s)
- Ryan J Martinez
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| | - Kristin A Hogquist
- Center for Immunology, University of Minnesota Medical School, Minneapolis, MN, United States
- Department of Laboratory Medicine and Pathology, University of Minnesota Medical School, Minneapolis, MN, United States
| |
Collapse
|
8
|
Herik AI, Sinha S, Arora R, Small C, Dufour A, Biernaskie J, Cobo ER, McKay DM. In silico integrative scRNA analysis of human colonic epithelium indicates four tuft cell subtypes. Am J Physiol Gastrointest Liver Physiol 2025; 328:G96-G109. [PMID: 39589317 DOI: 10.1152/ajpgi.00182.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 11/04/2024] [Accepted: 11/17/2024] [Indexed: 11/27/2024]
Abstract
This study integrated and analyzed human single-cell RNA sequencing data from four publicly available datasets to enhance cellular resolution, unveiling a complex landscape of tuft cell heterogeneity within the human colon. Four tuft subtypes (TC1-TC4) emerged, as defined by unique gene expression profiles, indicating potentially novel biological functions. Tuft cell 1 (TC1) was characterized by an antimicrobial peptide signature; TC2 had an increased transcription machinery gene expression profile consistent with a progenitor-like cell; TC3 expressed genes related to ganglion (neuronal) development; and TC4 expressed genes related to tight junctions. Our analysis of subtype-specific gene expression and pathway enrichment showed variances in tuft cell subtypes between healthy individuals and those with inflammatory bowel disease (IBD). The frequency of TC1 and TC2 differed between healthy controls and IBD. Relative to healthy controls, TC1 and TC2 in IBD tissue showed an upregulation of gene expression, favoring increased metabolism and immune function. These findings provide foundational knowledge about the complexity of the human colon tuft cell population and hint at their potential contributions to gut health. They provide a basis for future studies to explore the specific roles these cells may play in gut function during homeostasis and disease. We demonstrate the value of in silico approaches for hypothesis generation in relation to the putative functions of low-frequency gut cells for subsequent physiological analyses.NEW & NOTEWORTHY This study reveals the nuanced and novel landscape of human colonic tuft cells through integrative scRNA-seq analysis. Four distinct tuft cell subtypes were identified, varying markedly between healthy and individuals with IBD. We uncovered human colonic tuft cell subtypes with unexpected antimicrobial and progenitor-like gene expression signatures. These insights into tuft cell diversity offer new avenues for understanding gut health and disease pathophysiology.
Collapse
Affiliation(s)
- Aydin I Herik
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Sarthak Sinha
- Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rohit Arora
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Caleb Small
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Antoine Dufour
- McCaig Institute for Bone and Joint Health, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
- Department of Physiology and Pharmacology, Alberta Children's Hospital Research Institute, Calgary, Alberta, Canada
| | - Eduardo R Cobo
- Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Derek M McKay
- Gastrointestinal Research Group and Inflammation Research Network, Department of Physiology and Pharmacology, Calvin, Phoebe, and Joan Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
9
|
Huisman BD, Michelson DA, Rubin SA, Kohlsaat K, Gomarga W, Fang Y, Lee JM, Del Nido P, Nathan M, Benoist C, Zon L, Mathis D. Cross-species analyses of thymic mimetic cells reveal evolutionarily ancient origins and both conserved and species-specific elements. Immunity 2025; 58:108-123.e7. [PMID: 39731911 PMCID: PMC11735279 DOI: 10.1016/j.immuni.2024.11.025] [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: 05/22/2024] [Revised: 08/19/2024] [Accepted: 11/27/2024] [Indexed: 12/30/2024]
Abstract
Thymic mimetic cells are molecular hybrids between medullary-thymic-epithelial cells (mTECs) and diverse peripheral cell types. They are involved in eliminating autoreactive T cells and can perform supplementary functions reflective of their peripheral-cell counterparts. Current knowledge about mimetic cells derives largely from mouse models. To provide the high resolution that proved revelatory for mice, we performed single-cell RNA sequencing on purified mimetic-cell compartments from human pediatric donors. The single-cell profiles of individual donors were surprisingly similar, with diversification of neuroendocrine subtypes and expansion of the muscle subtype relative to mice. Informatic and imaging studies on the muscle-mTEC population highlighted a maturation trajectory suggestive of skeletal-muscle differentiation, some striated structures, and occasional cellular groupings reminiscent of neuromuscular junctions. We also profiled thymic mimetic cells from zebrafish. Integration of data from the three species identified species-specific adaptations but substantial interspecies conservation, highlighting the evolutionarily ancient nature of mimetic mTECs. Our findings provide a landscape view of human mimetic cells, with anticipated relevance in autoimmunity.
Collapse
Affiliation(s)
- Brooke D Huisman
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Daniel A Michelson
- Department of Immunology, Harvard Medical School, Boston, MA, USA; Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA; PhD Program in Immunology, Harvard Medical School, Boston, MA, USA
| | - Sara A Rubin
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, MA, USA; PhD Program in Immunology, Harvard Medical School, Boston, MA, USA; Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Katherine Kohlsaat
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Wilson Gomarga
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA
| | - Yuan Fang
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Ji Myung Lee
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Pedro Del Nido
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Meena Nathan
- Department of Cardiac Surgery, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, Harvard Medical School, Boston, MA, USA
| | | | - Leonard Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA, USA; Howard Hughes Medical Institute and Boston Children's Hospital, Boston, MA, USA
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Kellogg TD, Ceglia S, Mortzfeld BM, Tanna TM, Zeamer AL, Mancini MR, Foley SE, Ward DV, Bhattarai SK, McCormick BA, Reboldi A, Bucci V. Succinate-producing microbiota drives tuft cell hyperplasia to protect against Clostridioides difficile. J Exp Med 2025; 222:e20232055. [PMID: 39589553 PMCID: PMC11602550 DOI: 10.1084/jem.20232055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 09/26/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
The role of microbes and their metabolites in modulating tuft cell (TC) dynamics in the large intestine and the relevance of this pathway to infections is unknown. Here, we uncover that microbiome-driven colonic TC hyperplasia protects against Clostridioides difficile infection. Using selective antibiotics, we demonstrate increased type 2 cytokines and TC hyperplasia in the colon but not in the ileum. We demonstrate the causal role of the microbiome in modulating this phenotype using fecal matter transplantation and administration of consortia of succinate-producing bacteria. Administration of succinate production-deficient microbes shows a reduced response in a Pou2f3-dependent manner despite similar intestinal colonization. Finally, antibiotic-treated mice prophylactically administered with succinate-producing bacteria show increased protection against C. difficile-induced morbidity and mortality. This effect is nullified in Pou2f3-/- mice, confirming that the protection occurs via the TC pathway. We propose that activation of TCs by the microbiota in the colon is a mechanism evolved by the host to counterbalance microbiome-derived cues that facilitate invasion by pathogens.
Collapse
Affiliation(s)
- Tasia D. Kellogg
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Simona Ceglia
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Benedikt M. Mortzfeld
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Tanvi M. Tanna
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Abigail L. Zeamer
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Matthew R. Mancini
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Sage E. Foley
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
| | - Doyle V. Ward
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Shakti K. Bhattarai
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Beth A. McCormick
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Andrea Reboldi
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Vanni Bucci
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| |
Collapse
|
11
|
Klein L, Petrozziello E. Antigen presentation for central tolerance induction. Nat Rev Immunol 2025; 25:57-72. [PMID: 39294277 DOI: 10.1038/s41577-024-01076-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/20/2024]
Abstract
The extent of central T cell tolerance is determined by the diversity of self-antigens that developing thymocytes 'see' on thymic antigen-presenting cells (APCs). Here, focusing on insights from the past decade, we review the functional adaptations of medullary thymic epithelial cells, thymic dendritic cells and thymic B cells for the purpose of tolerance induction. Their distinct cellular characteristics range from unconventional phenomena, such as promiscuous gene expression or mimicry of peripheral cell types, to strategic positioning in distinct microenvironments and divergent propensities to preferentially access endogenous or exogenous antigen pools. We also discuss how 'tonic' inflammatory signals in the thymic microenvironment may extend the intrathymically visible 'self' to include autoantigens that are otherwise associated with highly immunogenic peripheral environments.
Collapse
Affiliation(s)
- Ludger Klein
- Institute for Immunology, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany.
| | - Elisabetta Petrozziello
- Institute for Immunology, Biomedical Center (BMC), Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| |
Collapse
|
12
|
Matsumoto M, Sobral F, Cardoso JS, Oya T, Tsuneyama K, Matsumoto M, Alves NL. The Ins and Outs of Thymic Epithelial Cell Differentiation and Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1471:51-79. [PMID: 40067584 DOI: 10.1007/978-3-031-77921-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2025]
Abstract
The thymus is an essential component of the immune system responsible for producing T cells. It is anatomically divided into two main regions: the outer cortex and the inner medulla. This chapter summarizes our current understanding of thymic stromal cell functions, with a particular focus on the interactions between these cells and T cells. This exploration aims to shed light on the pathogenesis of immune disorders, including autoimmunity.
Collapse
Affiliation(s)
- Minoru Matsumoto
- Department of Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Francisco Sobral
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Thymus Development and Function Laboratory, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - João S Cardoso
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Thymus Development and Function Laboratory, Instituto de Biologia Molecular e Celular, Porto, Portugal
| | - Takeshi Oya
- Department of Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Koichi Tsuneyama
- Department of Pathology and Laboratory Medicine, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Mitsuru Matsumoto
- Division of Molecular Immunology, Institute for Enzyme Research, Tokushima University, Tokushima, Japan.
| | - Nuno L Alves
- Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- Thymus Development and Function Laboratory, Instituto de Biologia Molecular e Celular, Porto, Portugal.
| |
Collapse
|
13
|
Silverman JB, Krystofiak EE, Caplan LR, Lau KS, Tyska MJ. Organization of a cytoskeletal superstructure in the apical domain of intestinal tuft cells. J Cell Biol 2024; 223:e202404070. [PMID: 39352498 PMCID: PMC11457492 DOI: 10.1083/jcb.202404070] [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: 04/13/2024] [Revised: 08/26/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024] Open
Abstract
Tuft cells are a rare epithelial cell type that play important roles in sensing and responding to luminal antigens. A defining morphological feature of this lineage is the actin-rich apical "tuft," which contains large fingerlike protrusions. However, details of the cytoskeletal ultrastructure underpinning the tuft, the molecules involved in building this structure, or how it supports tuft cell biology remain unclear. In the context of the small intestine, we found that tuft cell protrusions are supported by long-core bundles that consist of F-actin crosslinked in a parallel and polarized configuration; they also contain a tuft cell-specific complement of actin-binding proteins that exhibit regionalized localization along the bundle axis. Remarkably, in the sub-apical cytoplasm, the array of core actin bundles interdigitates and co-aligns with a highly ordered network of microtubules. The resulting cytoskeletal superstructure is well positioned to support subcellular transport and, in turn, the dynamic sensing functions of the tuft cell that are critical for intestinal homeostasis.
Collapse
Affiliation(s)
- Jennifer B. Silverman
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Evan E. Krystofiak
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
- Vanderbilt Cell Imaging Shared Resource, Vanderbilt University, Nashville, TN, USA
| | - Leah R. Caplan
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Ken S. Lau
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Matthew J. Tyska
- Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, TN, USA
| |
Collapse
|
14
|
Feng X, Flüchter P, De Tenorio JC, Schneider C. Tuft cells in the intestine, immunity and beyond. Nat Rev Gastroenterol Hepatol 2024; 21:852-868. [PMID: 39327439 DOI: 10.1038/s41575-024-00978-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/02/2024] [Indexed: 09/28/2024]
Abstract
Tuft cells have gained substantial attention over the past 10 years due to numerous reports linking them with type 2 immunity and microorganism-sensing capacity in many mucosal tissues. This heightened interest is fuelled by their unique ability to produce an array of biological effector molecules, including IL-25, allergy-related eicosanoids, and the neurotransmitter acetylcholine, enabling downstream responses in diverse cell types. Operating through G protein-coupled receptor-mediated signalling pathways reminiscent of type II taste cells in oral taste buds, tuft cells emerge as chemosensory sentinels that integrate luminal conditions, eliciting appropriate responses in immune, epithelial and neuronal populations. How tuft cells promote tissue alterations and adaptation to the variety of stimuli at mucosal surfaces has been explored in multiple studies in the past few years. Since the initial recognition of the role of tuft cells, the discovery of diverse tuft cell effector functions and associated feedback loops have also revealed the complexity of tuft cell biology. Although earlier work largely focused on extraintestinal tissues, novel genetic tools and recent mechanistic studies on intestinal tuft cells established fundamental concepts of tuft cell activation and functions. This Review is an overview of intestinal tuft cells, providing insights into their development, signalling and interaction modules in immunity and other states.
Collapse
Affiliation(s)
- Xiaogang Feng
- Department of Physiology, University of Zurich, Zurich, Switzerland
| | - Pascal Flüchter
- Department of Physiology, University of Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
15
|
Muro R, Nitta T, Nitta S, Tsukasaki M, Asano T, Nakano K, Okamura T, Nakashima T, Okamoto K, Takayanagi H. Transcript splicing optimizes the thymic self-antigen repertoire to suppress autoimmunity. J Clin Invest 2024; 134:e179612. [PMID: 39403924 PMCID: PMC11473167 DOI: 10.1172/jci179612] [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: 01/23/2024] [Accepted: 08/15/2024] [Indexed: 10/19/2024] Open
Abstract
Immunological self-tolerance is established in the thymus by the expression of virtually all self-antigens, including tissue-restricted antigens (TRAs) and cell-type-restricted antigens (CRAs). Despite a wealth of knowledge about the transcriptional regulation of TRA genes, posttranscriptional regulation remains poorly understood. Here, we show that protein arginine methylation plays an essential role in central immune tolerance by maximizing the self-antigen repertoire in medullary thymic epithelial cells (mTECs). Protein arginine methyltransferase-5 (Prmt5) was required for pre-mRNA splicing of certain key genes in tolerance induction, including Aire as well as various genes encoding TRAs. Mice lacking Prmt5 specifically in thymic epithelial cells exhibited an altered thymic T cell selection, leading to the breakdown of immune tolerance accompanied by both autoimmune responses and enhanced antitumor immunity. Thus, arginine methylation and transcript splicing are essential for establishing immune tolerance and may serve as a therapeutic target in autoimmune diseases as well as cancer immunotherapy.
Collapse
Affiliation(s)
- Ryunosuke Muro
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Molecular Pathology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Takeshi Nitta
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Molecular Pathology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba, Japan
| | - Sachiko Nitta
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Masayuki Tsukasaki
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tatsuo Asano
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Tomoki Nakashima
- Department of Cell Signaling, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
- Division of Immune Environment Dynamics, Cancer Research Institute, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
16
|
Baldwin I, Robey EA. Adjusting to self in the thymus: CD4 versus CD8 lineage commitment and regulatory T cell development. J Exp Med 2024; 221:e20230896. [PMID: 38980291 PMCID: PMC11232887 DOI: 10.1084/jem.20230896] [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: 03/22/2024] [Revised: 05/22/2024] [Accepted: 06/27/2024] [Indexed: 07/10/2024] Open
Abstract
During thymic development, thymocytes adjust their TCR response based on the strength of their reactivity to self-peptide MHC complexes. This tuning process allows thymocytes with a range of self-reactivities to survive positive selection and contribute to a diverse T cell pool. In this review, we will discuss recent advances in our understanding of how thymocytes tune their responsiveness during positive selection, and we present a "sequential selection" model to explain how MHC specificity influences lineage choice. We also discuss recent evidence for cell type diversity in the medulla and discuss how this heterogeneity may contribute to medullary niches for negative selection and regulatory T cell development.
Collapse
Affiliation(s)
- Isabel Baldwin
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Ellen A. Robey
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
17
|
Pala F, Notarangelo LD, Bosticardo M. Rediscovering the human thymus through cutting-edge technologies. J Exp Med 2024; 221:e20230892. [PMID: 39167072 PMCID: PMC11338284 DOI: 10.1084/jem.20230892] [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: 03/08/2024] [Revised: 06/24/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024] Open
Abstract
Recent technological advances have transformed our understanding of the human thymus. Innovations such as high-resolution imaging, single-cell omics, and organoid cultures, including thymic epithelial cell (TEC) differentiation and culture, and improvements in biomaterials, have further elucidated the thymus architecture, cellular dynamics, and molecular mechanisms underlying T cell development, and have unraveled previously unrecognized levels of stromal cell heterogeneity. These advancements offer unprecedented insights into thymic biology and hold promise for the development of novel therapeutic strategies for immune-related disorders.
Collapse
Affiliation(s)
- Francesca Pala
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD, USA
| | - Luigi D Notarangelo
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD, USA
| | - Marita Bosticardo
- Immune Deficiency Genetics Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health , Bethesda, MD, USA
| |
Collapse
|
18
|
Anderson G, Cosway EJ, James KD, Ohigashi I, Takahama Y. Generation and repair of thymic epithelial cells. J Exp Med 2024; 221:e20230894. [PMID: 38980292 PMCID: PMC11232892 DOI: 10.1084/jem.20230894] [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: 02/02/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 07/10/2024] Open
Abstract
In the vertebrate immune system, thymus stromal microenvironments support the generation of αβT cells from immature thymocytes. Thymic epithelial cells are of particular importance, and the generation of cortical and medullary epithelial lineages from progenitor stages controls the initiation and maintenance of thymus function. Here, we discuss the developmental pathways that regulate thymic epithelial cell diversity during both the embryonic and postnatal periods. We also examine how thymus microenvironments respond to injury, with particular focus on mechanisms that ensure regeneration of thymic epithelial cells for the restoration of thymus function.
Collapse
Affiliation(s)
- Graham Anderson
- Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Emilie J. Cosway
- Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Kieran D. James
- Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Izumi Ohigashi
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, University of Tokushima, Tokushima, Japan
| | - Yousuke Takahama
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
19
|
Michelson DA, Mathis D. Thymic Mimetic Cells: Ontogeny as Immunology. Annu Rev Cell Dev Biol 2024; 40:283-300. [PMID: 38608315 PMCID: PMC11446667 DOI: 10.1146/annurev-cellbio-112122-023316] [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: 04/14/2024]
Abstract
Medullary thymic epithelial cells (mTECs) generate immunological self-tolerance by ectopically expressing peripheral-tissue antigens (PTAs) within the thymus to preview the peripheral self to maturing T cells. Recent work, drawing inspiration from old histological observations, has shown that subtypes of mTECs, collectively termed mimetic cells, co-opt developmental programs from throughout the organism to express biologically coherent groups of PTAs. Here, we review key aspects of mimetic cells, especially as they relate to the larger contexts of molecular, cellular, developmental, and evolutionary biology. We highlight lineage-defining transcription factors as key regulators of mimetic cells and speculate as to what other factors, including Aire and the chromatin potential of mTECs, permit mimetic cell differentiation and function. Last, we consider what mimetic cells can teach us about not only the thymus but also other tissues.
Collapse
Affiliation(s)
- Daniel A Michelson
- Harvard-MIT Program in Health Sciences and Technology, Harvard Medical School, Boston, Massachusetts, USA;
| | - Diane Mathis
- Department of Immunology, Harvard Medical School, Boston, Massachusetts, USA;
| |
Collapse
|
20
|
Artusa P, Nguyen Yamamoto L, Barbier C, Valbon SF, Aghazadeh Habashi Y, Djambazian H, Ismailova A, Lebel MÈ, Salehi-Tabar R, Sarmadi F, Ragoussis J, Goltzman D, Melichar HJ, White JH. Skewed epithelial cell differentiation and premature aging of the thymus in the absence of vitamin D signaling. SCIENCE ADVANCES 2024; 10:eadm9582. [PMID: 39321290 PMCID: PMC11423877 DOI: 10.1126/sciadv.adm9582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 08/19/2024] [Indexed: 09/27/2024]
Abstract
Central tolerance of thymocytes to self-antigen depends on the medullary thymic epithelial cell (mTEC) transcription factor autoimmune regulator (Aire), which drives tissue-restricted antigen (TRA) gene expression. Vitamin D signaling regulates Aire and TRA expression in mTECs, providing a basis for links between vitamin D deficiency and autoimmunity. We find that mice lacking Cyp27b1, which cannot produce hormonally active vitamin D, display profoundly reduced thymic cellularity, with a reduced proportion of Aire+ mTECs, attenuated TRA expression, and poorly defined cortical-medullary boundaries. Markers of T cell negative selection are diminished, and organ-specific autoantibodies are present in knockout (KO) mice. Single-cell RNA sequencing revealed that loss of Cyp27b1 skews mTEC differentiation toward Ccl21+ intertypical TECs and generates a gene expression profile consistent with premature aging. KO thymi display accelerated involution and reduced expression of thymic longevity factors. Thus, loss of thymic vitamin D signaling disrupts normal mTEC differentiation and function and accelerates thymic aging.
Collapse
Affiliation(s)
- Patricio Artusa
- Department of Physiology, McGill University, Montreal QC, Canada
| | - Loan Nguyen Yamamoto
- Department of Medicine, McGill University, Montreal QC, Canada
- Calcium Research Laboratory, McGill University Health Centre, Montreal QC, Canada
| | - Camille Barbier
- Department of Physiology, McGill University, Montreal QC, Canada
| | - Stefanie F Valbon
- Department of Microbiology, Immunology and Infectious Disease, Université de Montréal, Montreal, QC, Canada
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
| | | | - Haig Djambazian
- McGill University Genome Centre, McGill University, Montreal, QC, Canada
| | - Aiten Ismailova
- Department of Physiology, McGill University, Montreal QC, Canada
| | - Marie-Ève Lebel
- Maisonneuve-Rosemont Hospital Research Center, McGill University, Montreal, QC, Canada
| | | | - Fatemeh Sarmadi
- Department of Physiology, McGill University, Montreal QC, Canada
| | - Jiannis Ragoussis
- McGill University Genome Centre, McGill University, Montreal, QC, Canada
| | - David Goltzman
- Department of Medicine, McGill University, Montreal QC, Canada
- Calcium Research Laboratory, McGill University Health Centre, Montreal QC, Canada
| | - Heather J Melichar
- Rosalind and Morris Goodman Cancer Institute, McGill University, Montreal, QC, Canada
- Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - John H White
- Department of Physiology, McGill University, Montreal QC, Canada
- Department of Medicine, McGill University, Montreal QC, Canada
| |
Collapse
|
21
|
Yamada Y, Küffer S, Sauer C, Hoki M, Shibuya S, Tsujii H, Ono K, Moriyoshi K, Date H, Yoshizawa A, Szolkowska M, Haga H, Ströbel P, Marx A. Immunohistochemistry for YAP1 N-terminus and C-terminus highlights metaplastic thymoma and high-grade thymic epithelial tumors by different staining patterns. Virchows Arch 2024; 485:461-469. [PMID: 39096416 DOI: 10.1007/s00428-024-03888-4] [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: 06/22/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 08/05/2024]
Abstract
Metaplastic thymoma (MT), a rare subtype of thymic epithelial tumors (TETs), harbors YAP1::MAML2 fusions. Poroma, a skin tumor, also carries these fusions and exhibits a unique staining pattern for YAP1 immunohistochemistry (IHC), namely, a YAP1 N-terminus (YAP1[N])-positive but YAP1 C-terminus (YAP1[C])-negative pattern. In this context, MT was recently reported to lack YAP1(C) expression exclusively among TET subtypes. However, a lack of information about YAP1(N) expression in that study and another report that wild-type YAP1 expression was diminished in type B3 thymoma and thymic carcinoma warrants further studies for YAP1 expression in TETs. Thus, we immunohistochemically examined YAP1(N) and YAP1(C) staining patterns in our TET samples, including 14 cases of MT. In addition, 11 of the 14 MT cases were genetically analyzed with the formalin-fixed paraffin-embedded tissues if they harbored YAP1::MAML2 fusions. MT consistently exhibited YAP1(N)-positive and YAP(C)-negative staining, whereas type B3 thymoma and thymic carcinoma showed relatively heterogeneous staining patterns for YAP1(N) and YAP1(C) and were sometimes negative for both antibodies. Furthermore, a lower expression of YAP1 was found in type B3 compared to B2 thymomas. Among genetically analyzed 11 MT cases, 6 cases showed YAP1::MAML2 fusions, whereas the analysis failed in 5 very old cases due to poor RNA quality. These results indicate that IHC of both YAP1(N) and YAP1(C) is recommended to obtain staining patterns almost unique to MT. The biological significance of YAP1 in high-grade TETs warrants further investigation.
Collapse
Affiliation(s)
- Yosuke Yamada
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan.
- Department of Molecular Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.
| | - Stefan Küffer
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Christian Sauer
- Institute of Pathology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| | - Masahito Hoki
- Department of Diagnostic Pathology, Kyoto Katsura Hospital, Kyoto, Japan
| | - Shinsuke Shibuya
- Department of Diagnostic Pathology, Kyoto Katsura Hospital, Kyoto, Japan
| | - Hideaki Tsujii
- Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | - Kazuo Ono
- Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | - Koki Moriyoshi
- Department of Diagnostic Pathology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Nara Medical University, Kashihara, Japan
| | - Malgorzata Szolkowska
- Department of Pathology, The Institute of Tuberculosis and Lung Diseases, Warsaw, Poland
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-Cho, Sakyo-Ku, Kyoto, 606-8507, Japan
| | - Phillip Ströbel
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Alexander Marx
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| |
Collapse
|
22
|
Huoh YS, Zhang Q, Törner R, Baca SC, Arthanari H, Hur S. Mechanism for controlled assembly of transcriptional condensates by Aire. Nat Immunol 2024; 25:1580-1592. [PMID: 39169234 PMCID: PMC11362013 DOI: 10.1038/s41590-024-01922-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 07/10/2024] [Indexed: 08/23/2024]
Abstract
Transcriptional condensates play a crucial role in gene expression and regulation, yet their assembly mechanisms remain poorly understood. Here, we report a multi-layered mechanism for condensate assembly by autoimmune regulator (Aire), an essential transcriptional regulator that orchestrates gene expression reprogramming for central T cell tolerance. Aire condensates assemble on enhancers, stimulating local transcriptional activities and connecting disparate inter-chromosomal loci. This functional condensate formation hinges upon the coordination between three Aire domains: polymerization domain caspase activation recruitment domain (CARD), histone-binding domain (first plant homeodomain (PHD1)), and C-terminal tail (CTT). Specifically, CTT binds coactivators CBP/p300, recruiting Aire to CBP/p300-rich enhancers and promoting CARD-mediated condensate assembly. Conversely, PHD1 binds to the ubiquitous histone mark H3K4me0, keeping Aire dispersed throughout the genome until Aire nucleates on enhancers. Our findings showed that the balance between PHD1-mediated suppression and CTT-mediated stimulation of Aire polymerization is crucial to form transcriptionally active condensates at target sites, providing new insights into controlled polymerization of transcriptional regulators.
Collapse
Affiliation(s)
- Yu-San Huoh
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Qianxia Zhang
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Ricarda Törner
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sylvan C Baca
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Haribabu Arthanari
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sun Hur
- Howard Hughes Medical Institute and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
You Y, Dunst J, Ye K, Sandoz PA, Reinhardt A, Sandrock I, Comet NR, Sarkar RD, Yang E, Duprez E, Agudo J, Brown BD, Utz PJ, Kastenmüller W, Gerlach C, Prinz I, Önfelt B, Kreslavsky T. Direct presentation of inflammation-associated self-antigens by thymic innate-like T cells induces elimination of autoreactive CD8 + thymocytes. Nat Immunol 2024; 25:1367-1382. [PMID: 38992254 PMCID: PMC11291280 DOI: 10.1038/s41590-024-01899-6] [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: 06/19/2023] [Accepted: 06/17/2024] [Indexed: 07/13/2024]
Abstract
Upregulation of diverse self-antigens that constitute components of the inflammatory response overlaps spatially and temporally with the emergence of pathogen-derived foreign antigens. Therefore, discrimination between these inflammation-associated self-antigens and pathogen-derived molecules represents a unique challenge for the adaptive immune system. Here, we demonstrate that CD8+ T cell tolerance to T cell-derived inflammation-associated self-antigens is efficiently induced in the thymus and supported by redundancy in cell types expressing these molecules. In addition to thymic epithelial cells, this included thymic eosinophils and innate-like T cells, a population that expressed molecules characteristic for all major activated T cell subsets. We show that direct T cell-to-T cell antigen presentation by minute numbers of innate-like T cells was sufficient to eliminate autoreactive CD8+ thymocytes. Tolerance to such effector molecules was of critical importance, as its breach caused by decreased thymic abundance of a single model inflammation-associated self-antigen resulted in autoimmune elimination of an entire class of effector T cells.
Collapse
Affiliation(s)
- Yuanyuan You
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Josefine Dunst
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Kewei Ye
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Patrick A Sandoz
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Annika Reinhardt
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Natalia R Comet
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Rupak Dey Sarkar
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Emily Yang
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA, USA
| | - Estelle Duprez
- Epigenetic Factors in Normal and Malignant Hematopoiesis Lab, CRCM, CNRS, INSERM, Institut Paoli Calmettes, Aix Marseille University, Marseille, France
- Equipe Labellisée Ligue Nationale Contre le Cancer, Paris, France
| | - Judith Agudo
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA, USA
- Parker Institute for Cancer Immunotherapy, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Ludwig Center at Harvard, Boston, MA, USA
| | - Brian D Brown
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paul J Utz
- Department of Medicine, Division of Immunology and Rheumatology, Stanford University, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Wolfgang Kastenmüller
- Max Planck Research Group, Würzburg Institute of Systems Immunology, Julius-Maximilians-Universität Würzburg, Würzburg, Germany
| | - Carmen Gerlach
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Rheumatology, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Institute of Systems Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Taras Kreslavsky
- Division of Immunology and Allergy, Department of Medicine Solna, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden.
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Chiu H, Weinstein KN, Spath S, Hu A, Varela S, Obata-Ninomiya K, Ziegler SF. SKI Regulates Medullary Thymic Epithelial Cell Differentiation to Control Peripheral T Cell Responses in Mice. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:52-62. [PMID: 38767415 PMCID: PMC11182718 DOI: 10.4049/jimmunol.2300262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 04/15/2024] [Indexed: 05/22/2024]
Abstract
The thymus is an important site for the establishment of an appropriate immune response through positive and negative selection of developing T cells. During selection, developing T cells interact with cortical and medullary thymic epithelial cells (TECs), termed cTECs and mTECs, respectively. Using a Foxn1Cre+/-SKIfl/fl mouse model, we found that TEC-specific deletion of SKI reduced the mTEC compartment in the thymus and that tissue-restricted Ag expression in mTECs was altered. This decrease in the medullary area led to a decrease in CD4 thymocyte cellularity; however, mature CD4 cellularity in the spleen remained normal. Interestingly, naive CD4 T cells purified from SKI-deleted mice showed a defect in proliferation in vitro after global TCR stimulation, and these mice were significantly protected from developing experimental autoimmune encephalomyelitis compared with the control mice. Overall, our findings suggest that SKI signaling in the thymus regulates mTEC differentiation and function as well as downstream peripheral T cell responses and provide evidence for targeting SKI in T cell-driven autoimmune diseases such as multiple sclerosis.
Collapse
|
25
|
Li YH, Yang YS, Xue YB, Lei H, Zhang SS, Qian J, Yao Y, Zhou R, Huang L. G protein subunit G γ13-mediated signaling pathway is critical to the inflammation resolution and functional recovery of severely injured lungs. eLife 2024; 12:RP92956. [PMID: 38836551 DOI: 10.7554/elife.92956] [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: 06/06/2024] Open
Abstract
Tuft cells are a group of rare epithelial cells that can detect pathogenic microbes and parasites. Many of these cells express signaling proteins initially found in taste buds. It is, however, not well understood how these taste signaling proteins contribute to the response to the invading pathogens or to the recovery of injured tissues. In this study, we conditionally nullified the signaling G protein subunit Gγ13 and found that the number of ectopic tuft cells in the injured lung was reduced following the infection of the influenza virus H1N1. Furthermore, the infected mutant mice exhibited significantly larger areas of lung injury, increased macrophage infiltration, severer pulmonary epithelial leakage, augmented pyroptosis and cell death, greater bodyweight loss, slower recovery, worsened fibrosis and increased fatality. Our data demonstrate that the Gγ13-mediated signal transduction pathway is critical to tuft cells-mediated inflammation resolution and functional repair of the damaged lungs.To our best knowledge, it is the first report indicating subtype-specific contributions of tuft cells to the resolution and recovery.
Collapse
Affiliation(s)
- Yi-Hong Li
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yi-Sen Yang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Yan-Bo Xue
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Hao Lei
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Sai-Sai Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, China
| | - Junbin Qian
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Genetics, Zhejiang University School of Medicine, Hangzhou, China
- Cancer Center, Zhejiang University, Hangzhou, China
| | - Yushi Yao
- Institute of Immunology and Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ruhong Zhou
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
| | - Liquan Huang
- College of Life Sciences, Zhejiang University, Hangzhou, China
- Zhejiang University Shanghai Institute for Advanced Study, Shanghai, Shanghai, China
- Monell Chemical Senses Center, Philadelphia, United States
| |
Collapse
|
26
|
Miller CN, Waterfield MR, Gardner JM, Anderson MS. Aire in Autoimmunity. Annu Rev Immunol 2024; 42:427-53. [PMID: 38360547 PMCID: PMC11774315 DOI: 10.1146/annurev-immunol-090222-101050] [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: 02/17/2024]
Abstract
The role of the autoimmune regulator (Aire) in central immune tolerance and thymic self-representation was first described more than 20 years ago, but fascinating new insights into its biology continue to emerge, particularly in the era of advanced single-cell genomics. We briefly describe the role of human genetics in the discovery of Aire, as well as insights into its function gained from genotype-phenotype correlations and the spectrum of Aire-associated autoimmunity-including insights from patients with Aire mutations with broad and diverse implications for human health. We then highlight emerging trends in Aire biology, focusing on three topic areas. First, we discuss medullary thymic epithelial diversity and the role of Aire in thymic epithelial development. Second, we highlight recent developments regarding the molecular mechanisms of Aire and its binding partners. Finally, we describe the rapidly evolving biology of the identity and function of extrathymic Aire-expressing cells (eTACs), and a novel eTAC subset called Janus cells, as well as their potential roles in immune homeostasis.
Collapse
Affiliation(s)
- Corey N Miller
- Diabetes Center, University of California, San Francisco, California, USA; ,
- Department of Medicine, University of California, San Francisco, California, USA
| | - Michael R Waterfield
- Department of Pediatrics, University of California, San Francisco, California, USA
| | - James M Gardner
- Diabetes Center, University of California, San Francisco, California, USA; ,
- Department of Surgery, University of California, San Francisco, California, USA
| | - Mark S Anderson
- Diabetes Center, University of California, San Francisco, California, USA; ,
- Department of Medicine, University of California, San Francisco, California, USA
| |
Collapse
|
27
|
Ribeiro C, Ferreirinha P, Landry JJM, Macedo F, Sousa LG, Pinto R, Benes V, Alves NL. Foxo3 regulates cortical and medullary thymic epithelial cell homeostasis with implications in T cell development. Cell Death Dis 2024; 15:352. [PMID: 38773063 PMCID: PMC11109193 DOI: 10.1038/s41419-024-06728-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/23/2024]
Abstract
Within the thymus, thymic epithelial cells (TECs) create dedicated microenvironments for T cell development and selection. Considering that TECs are sensitive to distinct pathophysiological conditions, uncovering the molecular elements that coordinate their thymopoietic role has important fundamental and clinical implications. Particularly, medullary thymic epithelial cells (mTECs) play a crucial role in central tolerance. Our previous studies, along with others, suggest that mTECs depend on molecular factors linked to genome-protecting pathways, but the precise mechanisms underlying their function remain unknown. These observations led us to examine the role of Foxo3, as it is expressed in TECs and involved in DNA damage response. Our findings show that mice with TEC-specific deletion of Foxo3 (Foxo3cKO) displayed a disrupted mTEC compartment, with a more profound impact on the numbers of CCL21+ and thymic tuft mTEClo subsets. At the molecular level, Foxo3 controls distinct functional modules in the transcriptome of cTECs and mTECs under normal conditions, which includes the regulation of ribosomal biogenesis and DNA damage response, respectively. These changes in the TEC compartment resulted in a reduced total thymocyte cellularity and specific changes in regulatory T cell and iNKT cell development in the Foxo3cKO thymus. Lastly, the thymic defects observed in adulthood correlated with mild signs of altered peripheral immunotolerance in aged Foxo3cKO mice. Moreover, the deficiency in Foxo3 moderately aggravated the autoimmune predisposition observed in Aire-deficient mice. Our findings highlight the importance of Foxo3 in preserving the homeostasis of TECs and in supporting their role in T cell development and tolerance.
Collapse
Affiliation(s)
- Camila Ribeiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Pedro Ferreirinha
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Jonathan J M Landry
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Fátima Macedo
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- Departamento de Ciências Médicas, Universidade de Aveiro, Aveiro, Portugal
| | - Laura G Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Rute Pinto
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - Vladimir Benes
- Genomics Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Nuno L Alves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
28
|
Zhu S, Wei W. Progress in research on the role of fluoride in immune damage. Front Immunol 2024; 15:1394161. [PMID: 38807586 PMCID: PMC11130356 DOI: 10.3389/fimmu.2024.1394161] [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: 03/04/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
Excessive fluoride intake from residential environments may affect multiple tissues and organs; however, the specific pathogenic mechanisms are unclear. Researchers have recently focused on the damaging effects of fluoride on the immune system. Damage to immune function seriously affects the quality of life of fluoride-exposed populations and increases the incidence of infections and malignant tumors. Probing the mechanism of damage to immune function caused by fluoride helps identify effective drugs and methods to prevent and treat fluorosis and improve people's living standards in fluorosis-affected areas. Here, the recent literature on the effects of fluoride on the immune system is reviewed, and research on fluoride damage to the immune system is summarized in terms of three perspectives: immune organs, immune cells, and immune-active substances. We reviewed that excessive fluoride can damage immune organs, lead to immune cells dysfunction and interfere with the expression of immune-active substances. This review aimed to provide a potential direction for future fluorosis research from the perspective of fluoride-induced immune function impairment. In order to seek the key regulatory indicators of fluoride on immune homeostasis in the future.
Collapse
Affiliation(s)
- Siqi Zhu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Wei
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Lab of Trace Elements and Human Health Harbin Medical University, Harbin, China
| |
Collapse
|
29
|
Ushio A, Matsuda-Lennikov M, Kalle-Youngoue F, Shimizu A, Abdelmaksoud A, Kelly MC, Ishimaru N, Takahama Y. Functionally diverse thymic medullary epithelial cells interplay to direct central tolerance. Cell Rep 2024; 43:114072. [PMID: 38581680 PMCID: PMC11079940 DOI: 10.1016/j.celrep.2024.114072] [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: 10/13/2023] [Revised: 02/24/2024] [Accepted: 03/21/2024] [Indexed: 04/08/2024] Open
Abstract
Medullary thymic epithelial cells (mTECs) are essential for the establishment of self-tolerance in T cells. Promiscuous gene expression by a subpopulation of mTECs regulated by the nuclear protein Aire contributes to the display of self-genomic products to newly generated T cells. Recent reports have highlighted additional self-antigen-displaying mTEC subpopulations, namely Fezf2-expressing mTECs and a mosaic of self-mimetic mTECs including thymic tuft cells. In addition, a functionally different subset of mTECs produces chemokine CCL21, which attracts developing thymocytes to the medullary region. Here, we report that CCL21+ mTECs and Aire+ mTECs non-redundantly cooperate to direct self-tolerance to prevent autoimmune pathology by optimizing the deletion of self-reactive T cells and the generation of regulatory T cells. We also detect cooperation for self-tolerance between Aire and Fezf2, the latter of which unexpectedly regulates thymic tuft cells. Our results indicate an indispensable interplay among functionally diverse mTECs for the establishment of central self-tolerance.
Collapse
Affiliation(s)
- Aya Ushio
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Department of Oral Molecular Pathology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto, Tokushima 770-8504, Japan
| | - Mami Matsuda-Lennikov
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Felix Kalle-Youngoue
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD 21701, USA
| | - Akihide Shimizu
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Abdalla Abdelmaksoud
- Center for Cancer Research Collaborative Bioinformatics Resource, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Michael C Kelly
- Single Cell Analysis Facility, Cancer Research Technology Program, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Naozumi Ishimaru
- Department of Oral Molecular Pathology, Graduate School of Biomedical Sciences, Tokushima University, Kuramoto, Tokushima 770-8504, Japan
| | - Yousuke Takahama
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
30
|
Shirafkan F, Hensel L, Rattay K. Immune tolerance and the prevention of autoimmune diseases essentially depend on thymic tissue homeostasis. Front Immunol 2024; 15:1339714. [PMID: 38571951 PMCID: PMC10987875 DOI: 10.3389/fimmu.2024.1339714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 03/11/2024] [Indexed: 04/05/2024] Open
Abstract
The intricate balance of immune reactions towards invading pathogens and immune tolerance towards self is pivotal in preventing autoimmune diseases, with the thymus playing a central role in establishing and maintaining this equilibrium. The induction of central immune tolerance in the thymus involves the elimination of self-reactive T cells, a mechanism essential for averting autoimmunity. Disruption of the thymic T cell selection mechanisms can lead to the development of autoimmune diseases. In the dynamic microenvironment of the thymus, T cell migration and interactions with thymic stromal cells are critical for the selection processes that ensure self-tolerance. Thymic epithelial cells are particularly significant in this context, presenting self-antigens and inducing the negative selection of autoreactive T cells. Further, the synergistic roles of thymic fibroblasts, B cells, and dendritic cells in antigen presentation, selection and the development of regulatory T cells are pivotal in maintaining immune responses tightly regulated. This review article collates these insights, offering a comprehensive examination of the multifaceted role of thymic tissue homeostasis in the establishment of immune tolerance and its implications in the prevention of autoimmune diseases. Additionally, the developmental pathways of the thymus are explored, highlighting how genetic aberrations can disrupt thymic architecture and function, leading to autoimmune conditions. The impact of infections on immune tolerance is another critical area, with pathogens potentially triggering autoimmunity by altering thymic homeostasis. Overall, this review underscores the integral role of thymic tissue homeostasis in the prevention of autoimmune diseases, discussing insights into potential therapeutic strategies and examining putative avenues for future research on developing thymic-based therapies in treating and preventing autoimmune conditions.
Collapse
|
31
|
Silverman JB, Krystofiak EE, Caplan LR, Lau KS, Tyska MJ. Intestinal tuft cells assemble a cytoskeletal superstructure composed of co-aligned actin bundles and microtubules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.19.585757. [PMID: 38562898 PMCID: PMC10983963 DOI: 10.1101/2024.03.19.585757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background & Aims All tissues consist of a distinct set of cell types, which collectively support organ function and homeostasis. Tuft cells are a rare epithelial cell type found in diverse epithelia, where they play important roles in sensing antigens and stimulating downstream immune responses. Exhibiting a unique polarized morphology, tuft cells are defined by an array of giant actin filament bundles that support ∼2 μm of apical membrane protrusion and extend over 7 μm towards the cell's perinuclear region. Despite their established roles in maintaining intestinal epithelial homeostasis, tuft cells remain understudied due to their rarity (e.g. ∼ 1% in the small intestinal epithelium). Details regarding the ultrastructural organization of the tuft cell cytoskeleton, the molecular components involved in building the array of giant actin bundles, and how these cytoskeletal structures support tuft cell biology remain unclear. Methods To begin to answer these questions, we used advanced light and electron microscopy to perform quantitative morphometry of the small intestinal tuft cell cytoskeleton. Results We found that tuft cell core bundles consist of actin filaments that are crosslinked in a parallel "barbed-end out" configuration. These polarized structures are also supported by a unique group of tuft cell enriched actin-binding proteins that are differentially localized along the giant core bundles. Furthermore, we found that tuft cell actin bundles are co-aligned with a highly ordered network of microtubules. Conclusions Tuft cells assemble a cytoskeletal superstructure that is well positioned to serve as a track for subcellular transport along the apical-basolateral axis and in turn, support the dynamic sensing functions that are critical for intestinal epithelial homeostasis. SYNOPSIS This research leveraged advanced light and electron microscopy to perform quantitative morphometry of the intestinal tuft cell cytoskeleton. Three-dimensional reconstructions of segmented image data revealed a co-aligned actin-microtubule superstructure that may play a fundamental role in tuft cell function.
Collapse
|
32
|
Lagou MK, Argyris DG, Vodopyanov S, Gunther-Cummins L, Hardas A, Poutahidis T, Panorias C, DesMarais S, Entenberg C, Carpenter RS, Guzik H, Nishku X, Churaman J, Maryanovich M, DesMarais V, Macaluso FP, Karagiannis GS. Morphometric Analysis of the Thymic Epithelial Cell (TEC) Network Using Integrated and Orthogonal Digital Pathology Approaches. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.11.584509. [PMID: 38559037 PMCID: PMC10979902 DOI: 10.1101/2024.03.11.584509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
The thymus, a central primary lymphoid organ of the immune system, plays a key role in T cell development. Surprisingly, the thymus is quite neglected with regards to standardized pathology approaches and practices for assessing structure and function. Most studies use multispectral flow cytometry to define the dynamic composition of the thymus at the cell population level, but they are limited by lack of contextual insight. This knowledge gap hinders our understanding of various thymic conditions and pathologies, particularly how they affect thymic architecture, and subsequently, immune competence. Here, we introduce a digital pathology pipeline to address these challenges. Our approach can be coupled to analytical algorithms and utilizes rationalized morphometric assessments of thymic tissue, ranging from tissue-wide down to microanatomical and ultrastructural levels. This pipeline enables the quantitative assessment of putative changes and adaptations of thymic structure to stimuli, offering valuable insights into the pathophysiology of thymic disorders. This versatile pipeline can be applied to a wide range of conditions that may directly or indirectly affect thymic structure, ranging from various cytotoxic stimuli inducing acute thymic involution to autoimmune diseases, such as myasthenia gravis. Here, we demonstrate applicability of the method in a mouse model of age-dependent thymic involution, both by confirming established knowledge, and by providing novel insights on intrathymic remodeling in the aged thymus. Our orthogonal pipeline, with its high versatility and depth of analysis, promises to be a valuable and practical toolset for both basic and translational immunology laboratories investigating thymic function and disease.
Collapse
Affiliation(s)
- Maria K Lagou
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - Dimitrios G Argyris
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
- Integrated Imaging Program for Cancer Research, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - Stepan Vodopyanov
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
| | - Leslie Gunther-Cummins
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Alexandros Hardas
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, North Mymms, Hatfield, United Kingdom
| | - Theofilos Poutahidis
- Laboratory of Pathology, School of Veterinary Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Christos Panorias
- Division of Statistics and Operational Research, Department of Mathematics, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Sophia DesMarais
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Conner Entenberg
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Randall S Carpenter
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Hillary Guzik
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Xheni Nishku
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Joseph Churaman
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Maria Maryanovich
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Ruth L. and David S. Gottesman Institute for Stem Cell and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Cancer Dormancy and Tumor Microenvironment Institute, Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Vera DesMarais
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - Frank P Macaluso
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Analytical Imaging Facility, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| | - George S Karagiannis
- Department of Microbiology and Immunology, Albert Einstein College of Medicine, Bronx, NY, USA
- Tumor Microenvironment and Metastasis Program, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
- Integrated Imaging Program for Cancer Research, Montefiore-Einstein Comprehensive Cancer Center, Bronx, NY, USA
- Gruss-Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
- Cancer Dormancy and Tumor Microenvironment Institute, Montefiore-Einstein Comprehensive Cancer, Center, Bronx, NY, USA
| |
Collapse
|
33
|
Ohigashi I, White AJ, Yang MT, Fujimori S, Tanaka Y, Jacques A, Kiyonari H, Matsushita Y, Turan S, Kelly MC, Anderson G, Takahama Y. Developmental conversion of thymocyte-attracting cells into self-antigen-displaying cells in embryonic thymus medulla epithelium. eLife 2024; 12:RP92552. [PMID: 38466627 PMCID: PMC10928509 DOI: 10.7554/elife.92552] [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: 03/13/2024] Open
Abstract
Thymus medulla epithelium establishes immune self-tolerance and comprises diverse cellular subsets. Functionally relevant medullary thymic epithelial cells (mTECs) include a self-antigen-displaying subset that exhibits genome-wide promiscuous gene expression promoted by the nuclear protein Aire and that resembles a mosaic of extrathymic cells including mucosal tuft cells. An additional mTEC subset produces the chemokine CCL21, thereby attracting positively selected thymocytes from the cortex to the medulla. Both self-antigen-displaying and thymocyte-attracting mTEC subsets are essential for self-tolerance. Here, we identify a developmental pathway by which mTECs gain their diversity in functionally distinct subsets. We show that CCL21-expressing mTECs arise early during thymus ontogeny in mice. Fate-mapping analysis reveals that self-antigen-displaying mTECs, including Aire-expressing mTECs and thymic tuft cells, are derived from CCL21-expressing cells. The differentiation capability of CCL21-expressing embryonic mTECs is verified in reaggregate thymus experiments. These results indicate that CCL21-expressing embryonic mTECs carry a developmental potential to give rise to self-antigen-displaying mTECs, revealing that the sequential conversion of thymocyte-attracting subset into self-antigen-displaying subset serves to assemble functional diversity in the thymus medulla epithelium.
Collapse
Affiliation(s)
- Izumi Ohigashi
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, University of TokushimaTokushimaJapan
| | - Andrea J White
- Institute for Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Mei-Ting Yang
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Sayumi Fujimori
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, University of TokushimaTokushimaJapan
| | - Yu Tanaka
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Alison Jacques
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics ResearchHyogoJapan
| | - Yosuke Matsushita
- Division of Genome Medicine, Institute of Advanced Medical Sciences, University of TokushimaTokushimaJapan
| | - Sevilay Turan
- Sequencing Facility, Frederick National Laboratory for Cancer Research, National Cancer InstituteFrederickUnited States
| | - Michael C Kelly
- Single Cell Analysis Facility, Cancer Research Technology Program, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Graham Anderson
- Institute for Immunology and Immunotherapy, University of BirminghamBirminghamUnited Kingdom
| | - Yousuke Takahama
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| |
Collapse
|
34
|
Oftedal BE, Sjøgren T, Wolff ASB. Interferon autoantibodies as signals of a sick thymus. Front Immunol 2024; 15:1327784. [PMID: 38455040 PMCID: PMC10917889 DOI: 10.3389/fimmu.2024.1327784] [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: 10/25/2023] [Accepted: 02/07/2024] [Indexed: 03/09/2024] Open
Abstract
Type I interferons (IFN-I) are key immune messenger molecules that play an important role in viral defense. They act as a bridge between microbe sensing, immune function magnitude, and adaptive immunity to fight infections, and they must therefore be tightly regulated. It has become increasingly evident that thymic irregularities and mutations in immune genes affecting thymic tolerance can lead to the production of IFN-I autoantibodies (autoAbs). Whether these biomarkers affect the immune system or tissue integrity of the host is still controversial, but new data show that IFN-I autoAbs may increase susceptibility to severe disease caused by certain viruses, including SARS-CoV-2, herpes zoster, and varicella pneumonia. In this article, we will elaborate on disorders that have been identified with IFN-I autoAbs, discuss models of how tolerance to IFN-Is is lost, and explain the consequences for the host.
Collapse
Affiliation(s)
- Bergithe E. Oftedal
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Thea Sjøgren
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| | - Anette S. B. Wolff
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Medicine, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
35
|
Cui G, Abe S, Kato R, Ikuta K. Insights into the heterogeneity of iNKT cells: tissue-resident and circulating subsets shaped by local microenvironmental cues. Front Immunol 2024; 15:1349184. [PMID: 38440725 PMCID: PMC10910067 DOI: 10.3389/fimmu.2024.1349184] [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: 12/04/2023] [Accepted: 02/06/2024] [Indexed: 03/06/2024] Open
Abstract
Invariant natural killer T (iNKT) cells are a distinct subpopulation of innate-like T lymphocytes. They are characterized by semi-invariant T cell receptors (TCRs) that recognize both self and foreign lipid antigens presented by CD1d, a non-polymorphic MHC class I-like molecule. iNKT cells play a critical role in stimulating innate and adaptive immune responses, providing an effective defense against infections and cancers, while also contributing to chronic inflammation. The functions of iNKT cells are specific to their location, ranging from lymphoid to non-lymphoid tissues, such as the thymus, lung, liver, intestine, and adipose tissue. This review aims to provide insights into the heterogeneity of development and function in iNKT cells. First, we will review the expression of master transcription factors that define subsets of iNKT cells and their production of effector molecules such as cytokines and granzymes. In this article, we describe the gene expression profiles contributing to the kinetics, distribution, and cytotoxicity of iNKT cells across different tissue types. We also review the impact of cytokine production in distinct immune microenvironments on iNKT cell heterogeneity, highlighting a recently identified circulating iNKT cell subset. Additionally, we explore the potential of exploiting iNKT cell heterogeneity to create potent immunotherapies for human cancers in the future.
Collapse
Affiliation(s)
- Guangwei Cui
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Shinya Abe
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Ryoma Kato
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
- Faculty of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | - Koichi Ikuta
- Laboratory of Immune Regulation, Department of Virus Research, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|
36
|
Hoki M, Yamada Y, Hiratomo E, Hirata M, Takeuchi Y, Yoshimatsu M, Kikuchi M, Kishimoto Y, Marx A, Haga H. Expression of FOXI1 and POU2F3 varies among different salivary gland neoplasms and is higher in Warthin tumor. Discov Oncol 2024; 15:36. [PMID: 38358561 PMCID: PMC10869675 DOI: 10.1007/s12672-024-00892-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 02/11/2024] [Indexed: 02/16/2024] Open
Abstract
PURPOSE Salivary gland tumors are histologically diverse. Ionocytes and tuft cells, rare epithelial cells found in normal salivary glands, might be associated with salivary tumors. Here, we explored the expression of FOXI1 and POU2F3, master regulators of ionocytes and tuft cells, respectively, for common salivary neoplasms using immunohistochemistry. METHODS We analyzed normal salivary tissues and nine salivary gland tumors; Warthin tumors (WT), pleomorphic adenomas (PA), basal cell adenomas, and oncocytomas were benign, whereas mucoepidermoid, adenoid cystic, acinic cell, salivary duct carcinomas, and polymorphous adenocarcinomas were malignant. RESULTS Normal salivary glands contained a few FOXI1- and POU2F3-positive cells in the ducts instead of the acini, consistent with ionocytes and tuft cells, respectively. Among the benign tumors, only WTs and PAs consistently expressed FOXI1 (10/10 and 9/10, respectively). The median H-score of WTs was significantly higher than that of PAs (17.5 vs. 4, P = 0.01). While WTs and PAs harbored POU2F3-positive cells (10/10 and 9/10, respectively), the median H-score was higher in WTs than in PAs (10.5 vs 4, respectively). Furthermore, WTs exhibited a unique staining pattern of FOXI1- and POU2F3-positive cells, which were present in luminal and abluminal locations, respectively. Whereas none of the malignant tumors expressed FOXI1, only adenoid cystic carcinoma consistently expressed POU2F3 (5/5), with a median H-score of 4. CONCLUSION The expression patterns of the characteristic transcription factors found in ionocytes and tuft cells vary among salivary gland tumor types and are higher in WT, which might be relevant for understanding and diagnosing salivary gland neoplasms.
Collapse
Affiliation(s)
- Masahito Hoki
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yosuke Yamada
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Emi Hiratomo
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masahiro Hirata
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yasuhide Takeuchi
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masayoshi Yoshimatsu
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Masahiro Kikuchi
- Department of Otolaryngology-Head & Neck Surgery, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Yo Kishimoto
- Department of Otolaryngology, Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Alexander Marx
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, 54 Shogoin Kawahara-cho, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|
37
|
Lammers S, Barrera V, Brennecke P, Miller C, Yoon J, Balolong J, Anderson MS, Ho Sui S, Steinmetz LM, von Andrian UH, Rattay K. Ehf and Fezf2 regulate late medullary thymic epithelial cell and thymic tuft cell development. Front Immunol 2024; 14:1277365. [PMID: 38420512 PMCID: PMC10901246 DOI: 10.3389/fimmu.2023.1277365] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/29/2023] [Indexed: 03/02/2024] Open
Abstract
Thymic epithelial cells are indispensable for T cell maturation and selection and the induction of central immune tolerance. The self-peptide repertoire expressed by medullary thymic epithelial cells is in part regulated by the transcriptional regulator Aire (Autoimmune regulator) and the transcription factor Fezf2. Due to the high complexity of mTEC maturation stages (i.e., post-Aire, Krt10+ mTECs, and Dclk1+ Tuft mTECs) and the heterogeneity in their gene expression profiles (i.e., mosaic expression patterns), it has been challenging to identify the additional factors complementing the transcriptional regulation. We aimed to identify the transcriptional regulators involved in the regulation of mTEC development and self-peptide expression in an unbiased and genome-wide manner. We used ATAC footprinting analysis as an indirect approach to identify transcription factors involved in the gene expression regulation in mTECs, which we validated by ChIP sequencing. This study identifies Fezf2 as a regulator of the recently described thymic Tuft cells (i.e., Tuft mTECs). Furthermore, we identify that transcriptional regulators of the ELF, ESE, ERF, and PEA3 subfamily of the ETS transcription factor family and members of the Krüppel-like family of transcription factors play a role in the transcriptional regulation of genes involved in late mTEC development and promiscuous gene expression.
Collapse
Affiliation(s)
- Sören Lammers
- Institute for Theoretical Physics, Heidelberg University, Heidelberg, Germany
| | - Victor Barrera
- Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Philip Brennecke
- Department of Genetics, Stanford University, School of Medicine, Stanford, CA, United States
- Stanford Genome Technology Center, Stanford University, Stanford, CA, United States
| | - Corey Miller
- Diabetes Center, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Joon Yoon
- Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Jared Balolong
- Diabetes Center, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Mark S. Anderson
- Diabetes Center, University of California, San Francisco (UCSF), San Francisco, CA, United States
| | - Shannan Ho Sui
- Bioinformatics Core, Harvard T.H. Chan School of Public Health, Boston, MA, United States
| | - Lars M. Steinmetz
- Department of Genetics, Stanford University, School of Medicine, Stanford, CA, United States
- Stanford Genome Technology Center, Stanford University, Stanford, CA, United States
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Ulrich H. von Andrian
- Department of Immunology & HMS Center for Immune Imaging, Harvard Medical School, Boston, MA, United States
- The Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States
| | - Kristin Rattay
- Department of Immunology & HMS Center for Immune Imaging, Harvard Medical School, Boston, MA, United States
- Pharmacological Institute, Biochemical Pharmacological Center, University of Marburg, Marburg, Germany
| |
Collapse
|
38
|
Eshleman EM, Rice T, Potter C, Waddell A, Hashimoto-Hill S, Woo V, Field S, Engleman L, Lim HW, Schumacher MA, Frey MR, Denson LA, Finkelman FD, Alenghat T. Microbiota-derived butyrate restricts tuft cell differentiation via histone deacetylase 3 to modulate intestinal type 2 immunity. Immunity 2024; 57:319-332.e6. [PMID: 38295798 PMCID: PMC10901458 DOI: 10.1016/j.immuni.2024.01.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 10/14/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024]
Abstract
Tuft cells in mucosal tissues are key regulators of type 2 immunity. Here, we examined the impact of the microbiota on tuft cell biology in the intestine. Succinate induction of tuft cells and type 2 innate lymphoid cells was elevated with loss of gut microbiota. Colonization with butyrate-producing bacteria or treatment with butyrate suppressed this effect and reduced intestinal histone deacetylase activity. Epithelial-intrinsic deletion of the epigenetic-modifying enzyme histone deacetylase 3 (HDAC3) inhibited tuft cell expansion in vivo and impaired type 2 immune responses during helminth infection. Butyrate restricted stem cell differentiation into tuft cells, and inhibition of HDAC3 in adult mice and human intestinal organoids blocked tuft cell expansion. Collectively, these data define a HDAC3 mechanism in stem cells for tuft cell differentiation that is dampened by a commensal metabolite, revealing a pathway whereby the microbiota calibrate intestinal type 2 immunity.
Collapse
Affiliation(s)
- Emily M Eshleman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Taylor Rice
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Crystal Potter
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Immunology, Allergy and Rheumatology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Amanda Waddell
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Seika Hashimoto-Hill
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Vivienne Woo
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Sydney Field
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Laura Engleman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Hee-Woong Lim
- Division of Biomedical Informatics, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Michael A Schumacher
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA; Department of Pediatrics and Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Mark R Frey
- The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA; Department of Pediatrics and Department of Biochemistry and Molecular Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Lee A Denson
- Division of Gastroenterology, Hepatology, and Nutrition, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Fred D Finkelman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Division of Immunology, Allergy and Rheumatology, Department of Internal Medicine, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Theresa Alenghat
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center and Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA; Center for Inflammation and Tolerance, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA.
| |
Collapse
|
39
|
Silverman JB, Vega PN, Tyska MJ, Lau KS. Intestinal Tuft Cells: Morphology, Function, and Implications for Human Health. Annu Rev Physiol 2024; 86:479-504. [PMID: 37863104 PMCID: PMC11193883 DOI: 10.1146/annurev-physiol-042022-030310] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2023]
Abstract
Tuft cells are a rare and morphologically distinct chemosensory cell type found throughout many organs, including the gastrointestinal tract. These cells were identified by their unique morphologies distinguished by large apical protrusions. Ultrastructural data have begun to describe the molecular underpinnings of their cytoskeletal features, and tuft cell-enriched cytoskeletal proteins have been identified, although the connection of tuft cell morphology to tuft cell functionality has not yet been established. Furthermore, tuft cells display variations in function and identity between and within tissues, leading to the delineation of distinct tuft cell populations. As a chemosensory cell type, they display receptors that are responsive to ligands specific for their environment. While many studies have demonstrated the tuft cell response to protists and helminths in the intestine, recent research has highlighted other roles of tuft cells as well as implicated tuft cells in other disease processes including inflammation, cancer, and viral infections. Here, we review the literature on the cytoskeletal structure of tuft cells. Additionally, we focus on new research discussing tuft cell lineage, ligand-receptor interactions, tuft cell tropism, and the role of tuft cells in intestinal disease. Finally, we discuss the implication of tuft cell-targeted therapies in human health and how the morphology of tuft cells may contribute to their functionality.
Collapse
Affiliation(s)
- Jennifer B Silverman
- Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; ,
| | - Paige N Vega
- Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; ,
| | - Matthew J Tyska
- Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; ,
| | - Ken S Lau
- Epithelial Biology Center and Department of Cell and Developmental Biology, Vanderbilt University School of Medicine, Nashville, Tennessee, USA; ,
| |
Collapse
|
40
|
Miccoli A, Pianese V, Bidoli C, Fausto AM, Scapigliati G, Picchietti S. Transcriptome profiling of microdissected cortex and medulla unravels functional regionalization in the European sea bass Dicentrarchus labrax thymus. FISH & SHELLFISH IMMUNOLOGY 2024; 145:109319. [PMID: 38145782 DOI: 10.1016/j.fsi.2023.109319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 12/15/2023] [Accepted: 12/18/2023] [Indexed: 12/27/2023]
Abstract
The thymus is a sophisticated primary lymphoid organ in jawed vertebrates, but knowledge on teleost thymus remains scarce. In this study, for the first time in the European sea bass, laser capture microdissection was leveraged to collect two thymic regions based on histological features, namely the cortex and the medulla. The two regions were then processed by RNAseq and in-depth functional transcriptome analyses with the aim of revealing differential gene expression patterns and gene sets enrichments, ultimately unraveling unique microenvironments imperative for the development of functional T cells. The sea bass cortex emerged as a hub of T cell commitment, somatic recombination, chromatin remodeling, cell cycle regulation, and presentation of self antigens from autophagy-, proteasome- or proteases-processed proteins. The cortex therefore accommodated extensive thymocyte proliferation and differentiation up to the checkpoint of positive selection. The medulla instead appeared as the center stage in autoimmune regulation by negative selection and deletion of autoreactive T cells, central tolerance mechanisms and extracellular matrix organization. Region-specific canonical markers of T and non-T lineage cells as well as signals for migration to/from, and trafficking within, the thymus were identified, shedding light on the highly coordinated and exquisitely complex bi-directional interactions among thymocytes and stromal components. Markers ascribable to thymic nurse cells and poorly characterized post-aire mTEC populations were found in the cortex and medulla, respectively. An in-depth data mining also exposed previously un-annotated genomic resources with differential signatures. Overall, our findings contribute to a broader understanding of the relationship between regional organization and function in the European sea bass thymus, and provide essential insights into the molecular mechanisms underlying T-cell mediated adaptive immune responses in teleosts.
Collapse
Affiliation(s)
- A Miccoli
- National Research Council, Institute for Marine Biological Resources and Biotechnology (IRBIM), 60125, Ancona, Italy
| | - V Pianese
- Dept. for Innovation in Biological, Agro-food and Forest Systems (DIBAF), University of Tuscia, Largo Dell'Università Snc, 01100, Viterbo, Italy
| | - C Bidoli
- Dept. of Life Sciences, University of Trieste, 34127, Trieste, Italy
| | - A M Fausto
- Dept. for Innovation in Biological, Agro-food and Forest Systems (DIBAF), University of Tuscia, Largo Dell'Università Snc, 01100, Viterbo, Italy
| | - G Scapigliati
- Dept. for Innovation in Biological, Agro-food and Forest Systems (DIBAF), University of Tuscia, Largo Dell'Università Snc, 01100, Viterbo, Italy
| | - S Picchietti
- Dept. for Innovation in Biological, Agro-food and Forest Systems (DIBAF), University of Tuscia, Largo Dell'Università Snc, 01100, Viterbo, Italy.
| |
Collapse
|
41
|
Kellogg TD, Ceglia S, Mortzfeld BM, Zeamer AL, Foley SE, Ward DV, Bhattarai SK, McCormick BA, Reboldi A, Bucci V. Microbiota encoded fatty-acid metabolism expands tuft cells to protect tissues homeostasis during Clostridioides difficile infection in the large intestine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.29.574039. [PMID: 38352546 PMCID: PMC10862725 DOI: 10.1101/2024.01.29.574039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/22/2024]
Abstract
Metabolic byproducts of the intestinal microbiota are crucial in maintaining host immune tone and shaping inter-species ecological dynamics. Among these metabolites, succinate is a driver of tuft cell (TC) differentiation and consequent type 2 immunity-dependent protection against invading parasites in the small intestine. Succinate is also a growth enhancer of the nosocomial pathogen Clostridioides difficile in the large intestine. To date, no research has shown the role of succinate in modulating TC dynamics in the large intestine, or the relevance of this immune pathway to C. difficile pathophysiology. Here we reveal the existence of a three-way circuit between commensal microbes, C. difficile and host epithelial cells which centers around succinate. Through selective microbiota depletion experiments we demonstrate higher levels of type 2 cytokines leading to expansion of TCs in the colon. We then demonstrate the causal role of the microbiome in modulating colonic TC abundance and subsequent type 2 cytokine induction using rational supplementation experiments with fecal transplants and microbial consortia of succinate-producing bacteria. We show that administration of a succinate-deficient Bacteroides thetaiotaomicron knockout (Δfrd) significantly reduces the enhanced type 2 immunity in mono-colonized mice. Finally, we demonstrate that mice prophylactically administered with the consortium of succinate-producing bacteria show reduced C. difficile-induced morbidity and mortality compared to mice administered with heat-killed bacteria or the vehicle. This effect is reduced in a partial tuft cell knockout mouse, Pou2f3+/-, and nullified in the tuft cell knockout mouse, Pou2f3-/-, confirming that the observed protection occurs via the TC pathway. Succinate is an intermediary metabolite of the production of short-chain fatty acids, and its concentration often increases during dysbiosis. The first barrier to enteric pathogens alike is the intestinal epithelial barrier, and host maintenance and strengthening of barrier integrity is vital to homeostasis. Considering our data, we propose that activation of TC by the microbiota-produced succinate in the colon is a mechanism evolved by the host to counterbalance microbiome-derived cues that facilitate invasion by intestinal pathogens.
Collapse
Affiliation(s)
- Tasia D. Kellogg
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Simona Ceglia
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Benedikt M. Mortzfeld
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Abigail L. Zeamer
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Sage E. Foley
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Current address: Transformational and Translational Immunology Discovery Department, AbbVie, Cambridge, MA, USA
| | - Doyle V. Ward
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Shakti K. Bhattarai
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Beth A. McCormick
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Andrea Reboldi
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Vanni Bucci
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| |
Collapse
|
42
|
Jang B, Kim H, Lee S, Won Y, Kaji I, Coffey RJ, Choi E, Goldenring JR. Dynamic tuft cell expansion during gastric metaplasia and dysplasia. J Pathol Clin Res 2024; 10:e352. [PMID: 38117182 PMCID: PMC10766036 DOI: 10.1002/cjp2.352] [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: 06/10/2023] [Revised: 11/14/2023] [Accepted: 11/17/2023] [Indexed: 12/21/2023]
Abstract
Tuft cells are chemosensory cells associated with luminal homeostasis, immune response, and tumorigenesis in the gastrointestinal tract. We aimed to elucidate alterations in tuft cell populations during gastric atrophy and tumorigenesis in humans with correlative comparison to relevant mouse models. Tuft cell distribution was determined in human stomachs from organ donors and in gastric pathologies including Ménétrier's disease, Helicobacter pylori gastritis, intestinal metaplasia (IM), and gastric tumors. Tuft cell populations were examined in Lrig1-KrasG12D , Mist1-KrasG12D , and MT-TGFα mice. Tuft cells were evenly distributed throughout the entire normal human stomach, primarily concentrated in the isthmal region in the fundus. Ménétrier's disease stomach showed increased tuft cells. Similarly, Lrig1-Kras mice and mice overexpressing TGFα showed marked foveolar hyperplasia and expanded tuft cell populations. Human stomach with IM or dysplasia also showed increased tuft cell numbers. Similarly, Mist1-Kras mice had increased numbers of tuft cells during metaplasia and dysplasia development. In human gastric cancers, tuft cells were rarely observed, but showed positive associations with well-differentiated lesions. In mouse gastric cancer xenografts, tuft cells were restricted to dysplastic well-differentiated mucinous cysts and were lost in less differentiated cancers. Taken together, tuft cell populations increased in atrophic human gastric pathologies, metaplasia, and dysplasia, but were decreased in gastric cancers. Similar findings were observed in mouse models, suggesting that, while tuft cells are associated with precancerous pathologies, their loss is most associated with the progression to invasive cancer.
Collapse
Affiliation(s)
- Bogun Jang
- Section of Surgical SciencesVanderbilt University Medical CenterNashvilleTNUSA
- Jeju National University College of MedicineJejuRepublic of Korea
- Department of PathologyJeju National University HospitalJejuRepublic of Korea
| | - Hyesung Kim
- Section of Surgical SciencesVanderbilt University Medical CenterNashvilleTNUSA
- Jeju National University College of MedicineJejuRepublic of Korea
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTNUSA
| | - Su‐Hyung Lee
- Section of Surgical SciencesVanderbilt University Medical CenterNashvilleTNUSA
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Yoonkyung Won
- Section of Surgical SciencesVanderbilt University Medical CenterNashvilleTNUSA
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Izumi Kaji
- Section of Surgical SciencesVanderbilt University Medical CenterNashvilleTNUSA
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTNUSA
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - Robert J Coffey
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Department of Internal MedicineVanderbilt University Medical CenterNashvilleTNUSA
| | - Eunyoung Choi
- Section of Surgical SciencesVanderbilt University Medical CenterNashvilleTNUSA
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTNUSA
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
| | - James R Goldenring
- Section of Surgical SciencesVanderbilt University Medical CenterNashvilleTNUSA
- Department of Cell and Developmental BiologyVanderbilt UniversityNashvilleTNUSA
- Epithelial Biology CenterVanderbilt University Medical CenterNashvilleTNUSA
- Nashville VA Medical CenterNashvilleTNUSA
| |
Collapse
|
43
|
Matsumoto M, Matsumoto M. Learning the Autoimmune Pathogenesis Through the Study of Aire. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1444:19-32. [PMID: 38467970 DOI: 10.1007/978-981-99-9781-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/13/2024]
Abstract
One of the difficulties in studying the pathogenesis of autoimmune diseases is that the disease is multifactorial involving sex, age, MHC, environment, and some genetic factors. Because deficiency of Aire, a transcriptional regulator, is an autoimmune disease caused by a single gene abnormality, Aire is an ideal research target for approaching the enigma of autoimmunity, e.g., the mechanisms underlying Aire deficiency can be studied using genetically modified animals. Nevertheless, the exact mechanisms of the breakdown of self-tolerance due to Aire's dysfunction have not yet been fully clarified. This is due, at least in part, to the lack of information on the exact target genes controlled by Aire. State-of-the-art research infrastructures such as single-cell analysis are now in place to elucidate the essential function of Aire. The knowledge gained through the study of Aire-mediated tolerance should help our understanding of the pathogenesis of autoimmune disease in general.
Collapse
Affiliation(s)
| | - Minoru Matsumoto
- Department of Molecular Pathology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| |
Collapse
|
44
|
Fujimori S, Ohigashi I. The role of thymic epithelium in thymus development and age-related thymic involution. THE JOURNAL OF MEDICAL INVESTIGATION 2024; 71:29-39. [PMID: 38735722 DOI: 10.2152/jmi.71.29] [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: 05/14/2024]
Abstract
The establishment of an adaptive immune system is critical for protecting our bodies from neoplastic cancers and invading pathogens such as viruses and bacteria. As a primary lymphoid organ, the thymus generates lymphoid T cells that play a major role in the adaptive immune system. T cell generation in the thymus is controlled by interactions between thymocytes and other thymic cells, primarily thymic epithelial cells. Thus, the normal development and function of thymic epithelial cells are important for the generation of immunocompetent and self-tolerant T cells. On the other hand, the degeneration of the thymic epithelium due to thymic aging causes thymic involution, which is associated with the decline of adaptive immune function. Herein we summarize basic and current knowledge of the development and function of thymic epithelial cells and the mechanism of thymic involution. J. Med. Invest. 71 : 29-39, February, 2024.
Collapse
Affiliation(s)
- Sayumi Fujimori
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| | - Izumi Ohigashi
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, Japan
| |
Collapse
|
45
|
Barachini S, Pardini E, Burzi IS, Sardo Infirri G, Montali M, Petrini I. Molecular and Functional Key Features and Oncogenic Drivers in Thymic Carcinomas. Cancers (Basel) 2023; 16:166. [PMID: 38201593 PMCID: PMC10778094 DOI: 10.3390/cancers16010166] [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: 11/07/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Thymic epithelial tumors, comprising thymic carcinomas and thymomas, are rare neoplasms. They differ in histology, prognosis, and association with autoimmune diseases such as myasthenia gravis. Thymomas, but not thymic carcinomas, often harbor GTF2I mutations. Mutations of CDKN2A, TP53, and CDKN2B are the most common thymic carcinomas. The acquisition of mutations in genes that control chromatin modifications and epigenetic regulation occurs in the advanced stages of thymic carcinomas. Anti-angiogenic drugs and immune checkpoint inhibitors targeting the PD-1/PD-L1 axis have shown promising results for the treatment of unresectable tumors. Since thymic carcinomas are frankly aggressive tumors, this report presents insights into their oncogenic drivers, categorized under the established hallmarks of cancer.
Collapse
Affiliation(s)
- Serena Barachini
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Eleonora Pardini
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Irene Sofia Burzi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Gisella Sardo Infirri
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Marina Montali
- Department of Translational Research and of New Surgical and Medical Technologies, University of Pisa, 56126 Pisa, Italy
| | - Iacopo Petrini
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
46
|
Yamada Y, Iwane K, Nakanishi Y, Haga H. Thymic Carcinoma: Unraveling Neuroendocrine Differentiation and Epithelial Cell Identity Loss. Cancers (Basel) 2023; 16:115. [PMID: 38201543 PMCID: PMC10778300 DOI: 10.3390/cancers16010115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/10/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND The histogenesis of thymic epithelial tumors (TETs) has been a subject of debate. Recent technological advancements have revealed that thymic carcinomas often exhibit a phenotype akin to tuft cells, which is a subset of medullary TECs. Here, we further explored the gene expression signatures of thymic carcinomas in relation to tuft cells and their kinships-ionocytes and neuroendocrine cells (neuroendocrine group). METHODS We analyzed a single-cell RNA sequencing dataset from the normal human thymus. Concurrently, we examined publicly available datasets on the mRNA expression and methylation status of TECs and lung cancers. Real-time quantitative PCR was also conducted with our tissue samples. RESULTS Thymic carcinomas displayed a neuroendocrine phenotype biased toward tuft cells and ionocytes. When exploring the possible regulators of this phenotype, we discovered that HDAC9 and NFATC1 were characteristically expressed in the neuroendocrine group in adult TECs and thymic carcinomas. Additionally, the pan-thymic epithelium markers, exemplified by PAX9 and SIX1, were significantly suppressed in thymic carcinomas. CONCLUSIONS Thymic carcinomas might be characterized by unique neuroendocrine differentiation and loss of identity as thymic epithelial cells. Future studies investigating the role of HDAC9 and NFATC1 in thymic epithelium are warranted to explore their potential as therapeutic targets in TETs.
Collapse
Affiliation(s)
- Yosuke Yamada
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto 606-8507, Japan;
| | - Kosuke Iwane
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan; (K.I.); (Y.N.)
| | - Yuki Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan; (K.I.); (Y.N.)
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto 606-8507, Japan;
| |
Collapse
|
47
|
Ohigashi I, White AJ, Yang MT, Fujimori S, Tanaka Y, Jacques A, Kiyonari H, Matsushita Y, Turan S, Kelly MC, Anderson G, Takahama Y. Developmental conversion of thymocyte-attracting cells into self-antigen-displaying cells in embryonic thymus medulla epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.560657. [PMID: 37873155 PMCID: PMC10592888 DOI: 10.1101/2023.10.03.560657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Thymus medulla epithelium establishes immune self-tolerance and comprises diverse cellular subsets. Functionally relevant medullary thymic epithelial cells (mTECs) include a self-antigen-displaying subset that exhibits genome-wide promiscuous gene expression promoted by the nuclear protein Aire and that resembles a mosaic of extrathymic cells including mucosal tuft cells. An additional mTEC subset produces the chemokine CCL21, thereby attracting positively selected thymocytes from the cortex to the medulla. Both self-antigen-displaying and thymocyte-attracting mTEC subsets are essential for self-tolerance. Here we identify a developmental pathway by which mTECs gain their diversity in functionally distinct subsets. We show that CCL21-expressing mTECs arise early during thymus ontogeny. Fate-mapping analysis reveals that self-antigen-displaying mTECs, including Aire-expressing mTECs and thymic tuft cells, are derived from CCL21-expressing cells. The differentiation capability of CCL21-expressing embryonic mTECs is verified in reaggregate thymus experiments. These results indicate that CCL21-expressing embryonic mTECs carry a developmental potential to give rise to self-antigen-displaying mTECs, revealing that the sequential conversion of thymocyte-attracting subset into self-antigen-displaying subset serves to assemble functional diversity in the thymus medulla epithelium.
Collapse
Affiliation(s)
- Izumi Ohigashi
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, University of Tokushima, Tokushima 770-8503, Japan
| | - Andrea J. White
- Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Mei-Ting Yang
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Sayumi Fujimori
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, University of Tokushima, Tokushima 770-8503, Japan
| | - Yu Tanaka
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Alison Jacques
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, 2-2-3 Minatojima-minamimachi, Chuo, Kobe, Hyogo 650-0047, Japan
| | - Yosuke Matsushita
- Division of Genome Medicine, Institute of Advanced Medical Sciences, University of Tokushima, Tokushima 770-8503, Japan
| | - Sevilay Turan
- Sequencing Facility, Frederick National Laboratory for Cancer Research, National Cancer Institute, Frederick, MD 21701, USA
| | - Michael C. Kelly
- Single Cell Analysis Facility, Cancer Research Technology Program, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Graham Anderson
- Institute for Immunology and Immunotherapy, University of Birmingham, Birmingham B15 2TT, UK
| | - Yousuke Takahama
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
48
|
Tao W, Ye Z, Wei Y, Wang J, Yang W, Yu G, Xiong J, Jia S. Insm1 regulates mTEC development and immune tolerance. Cell Mol Immunol 2023; 20:1472-1486. [PMID: 37990032 PMCID: PMC10687002 DOI: 10.1038/s41423-023-01102-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 10/30/2023] [Indexed: 11/23/2023] Open
Abstract
The expression of self-antigens in medullary thymic epithelial cells (mTECs) is essential for the establishment of immune tolerance, but the regulatory network that controls the generation and maintenance of the multitude of cell populations expressing self-antigens is poorly understood. Here, we show that Insm1, a zinc finger protein with known functions in neuroendocrine and neuronal cells, is broadly coexpressed with an autoimmune regulator (Aire) in mTECs. Insm1 expression is undetectable in most mimetic cell populations derived from mTECs but persists in neuroendocrine mimetic cells. Mutation of Insm1 in mice downregulated Aire expression, dysregulated the gene expression program of mTECs, and altered mTEC subpopulations and the expression of tissue-restricted antigens. Consistent with these findings, loss of Insm1 resulted in autoimmune responses in multiple peripheral tissues. We found that Insm1 regulates gene expression in mTECs by binding to chromatin. Interestingly, the majority of the Insm1 binding sites are co-occupied by Aire and enriched in superenhancer regions. Together, our data demonstrate the important role of Insm1 in the regulation of the repertoire of self-antigens needed to establish immune tolerance.
Collapse
Affiliation(s)
- Weihua Tao
- The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, Jinan University, Guangzhou, China
- The Institute of Clinical Medicine, Jinan University, Guangzhou, China
- Key Lab of Guangzhou Basic and Translational Research of Pan-Vascular Diseases, Guangzhou, China
| | - Zhihuan Ye
- The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, Jinan University, Guangzhou, China
- The Institute of Clinical Medicine, Jinan University, Guangzhou, China
| | - Yiqiu Wei
- The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, Jinan University, Guangzhou, China
- The Institute of Clinical Medicine, Jinan University, Guangzhou, China
| | - Jianxue Wang
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Weixin Yang
- The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Guoxing Yu
- The First Affiliated Hospital of Jinan University, Guangzhou, China
- The Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, Jinan University, Guangzhou, China
- The Institute of Clinical Medicine, Jinan University, Guangzhou, China
| | - Jieyi Xiong
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.
| | - Shiqi Jia
- The First Affiliated Hospital of Jinan University, Guangzhou, China.
- The Guangdong-Hong Kong-Macao Joint University Laboratory of Metabolic and Molecular Medicine, Jinan University, Guangzhou, China.
- The Institute of Clinical Medicine, Jinan University, Guangzhou, China.
- Key Lab of Guangzhou Basic and Translational Research of Pan-Vascular Diseases, Guangzhou, China.
| |
Collapse
|
49
|
Malin J, Martinez-Ruiz GU, Zhao Y, Shissler SC, Cowan JE, Ding Y, Morales-Sanchez A, Ishikawa M, Lavaert M, Das A, Butcher D, Warner AC, Kallarakal M, Chen J, Kedei N, Kelly M, Brinster LR, Allman D, Bhandoola A. Expression of the transcription factor Klf6 by thymic epithelial cells is required for thymus development. SCIENCE ADVANCES 2023; 9:eadg8126. [PMID: 37967174 PMCID: PMC10651122 DOI: 10.1126/sciadv.adg8126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 10/16/2023] [Indexed: 11/17/2023]
Abstract
Thymic epithelial cells (TEC) control T cell development and play essential roles in establishing self-tolerance. By using Foxn1-Cre-driven ablation of Klf6 gene in TEC, we identified Klf6 as a critical factor in TEC development. Klf6 deficiency resulted in a hypoplastic thymus-evident from fetal stages into adulthood-in which a dramatic increase in the frequency of apoptotic TEC was observed. Among cortical TEC (cTEC), a previously unreported cTEC population expressing the transcription factor Sox10 was relatively expanded. Within medullary TEC (mTEC), mTEC I and Tuft-like mTEC IV were disproportionately decreased. Klf6 deficiency altered chromatin accessibility and affected TEC chromatin configuration. Consistent with these defects, naïve conventional T cells and invariant natural killer T cells were reduced in the spleen. Late stages of T cell receptor-dependent selection of thymocytes were affected, and mice exhibited autoimmunity. Thus, Klf6 has a prosurvival role and affects the development of specific TEC subsets contributing to thymic function.
Collapse
Affiliation(s)
- Justin Malin
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gustavo Ulises Martinez-Ruiz
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Research Division, Faculty of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
- Children’s Hospital Federico Gomez, Mexico City, Mexico
| | - Yongge Zhao
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susannah C. Shissler
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jennifer E. Cowan
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Institute of Immunity and Transplantation, University College London, London, UK
| | - Yi Ding
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Abigail Morales-Sanchez
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Children’s Hospital Federico Gomez, Mexico City, Mexico
| | - Masaki Ishikawa
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Marieke Lavaert
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arundhoti Das
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Donna Butcher
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Andrew C. Warner
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Melissa Kallarakal
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jingqiu Chen
- Office of Science and Technology Resources, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- ACROBiosystems, Newark, DE, USA
| | - Noemi Kedei
- Office of Science and Technology Resources, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Michael Kelly
- Single Cell Analysis Facility, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Lauren R. Brinster
- Division of Veterinary Resources, Office of Research Services, National Institutes of Health, Bethesda, MD, USA
| | - David Allman
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Avinash Bhandoola
- Laboratory of Genome Integrity, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
50
|
Horie K, Namiki K, Kinoshita K, Miyauchi M, Ishikawa T, Hayama M, Maruyama Y, Hagiwara N, Miyao T, Murata S, Kobayashi TJ, Akiyama N, Akiyama T. Acute irradiation causes a long-term disturbance in the heterogeneity and gene expression profile of medullary thymic epithelial cells. Front Immunol 2023; 14:1186154. [PMID: 38022666 PMCID: PMC10652284 DOI: 10.3389/fimmu.2023.1186154] [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: 03/14/2023] [Accepted: 10/20/2023] [Indexed: 12/01/2023] Open
Abstract
The thymus has the ability to regenerate from acute injury caused by radiation, infection, and stressors. In addition to thymocytes, thymic epithelial cells in the medulla (mTECs), which are crucial for T cell self-tolerance by ectopically expressing and presenting thousands of tissue-specific antigens (TSAs), are damaged by these insults and recover thereafter. However, given recent discoveries on the high heterogeneity of mTECs, it remains to be determined whether the frequency and properties of mTEC subsets are restored during thymic recovery from radiation damage. Here we demonstrate that acute total body irradiation with a sublethal dose induces aftereffects on heterogeneity and gene expression of mTECs. Single-cell RNA-sequencing (scRNA-seq) analysis showed that irradiation reduces the frequency of mTECs expressing AIRE, which is a critical regulator of TSA expression, 15 days after irradiation. In contrast, transit-amplifying mTECs (TA-mTECs), which are progenitors of AIRE-expressing mTECs, and Ccl21a-expressing mTECs, were less affected. Interestingly, a detailed analysis of scRNA-seq data suggested that the proportion of a unique mTEC cluster expressing Ccl25 and a high level of TSAs was severely decreased by irradiation. In sum, we propose that the effects of acute irradiation disrupt the heterogeneity and properties of mTECs over an extended period, which potentially leads to an impairment of thymic T cell selection.
Collapse
Affiliation(s)
- Kenta Horie
- Laboratory for Immune Homeostasis, RIKEN Center of Integrative Medical Sciences, Yokohama, Japan
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | - Kano Namiki
- Laboratory for Immune Homeostasis, RIKEN Center of Integrative Medical Sciences, Yokohama, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Kyouhei Kinoshita
- Graduate School of Engineering, The University of Tokyo, Tokyo, Japan
| | - Maki Miyauchi
- Laboratory for Immune Homeostasis, RIKEN Center of Integrative Medical Sciences, Yokohama, Japan
| | - Tatsuya Ishikawa
- Laboratory for Immune Homeostasis, RIKEN Center of Integrative Medical Sciences, Yokohama, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Mio Hayama
- Laboratory for Immune Homeostasis, RIKEN Center of Integrative Medical Sciences, Yokohama, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Yuya Maruyama
- Laboratory for Immune Homeostasis, RIKEN Center of Integrative Medical Sciences, Yokohama, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Naho Hagiwara
- Laboratory for Immune Homeostasis, RIKEN Center of Integrative Medical Sciences, Yokohama, Japan
| | - Takahisa Miyao
- YCI Laboratory for Immunological Transcriptomics, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| | - Shigeo Murata
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan
| | | | - Nobuko Akiyama
- Laboratory for Immune Homeostasis, RIKEN Center of Integrative Medical Sciences, Yokohama, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| | - Taishin Akiyama
- Laboratory for Immune Homeostasis, RIKEN Center of Integrative Medical Sciences, Yokohama, Japan
- Immunobiology, Graduate School of Medical Life Science, Yokohama City University, Yokohama, Japan
| |
Collapse
|