1
|
Verstichel G, Cheroutre H. T cells with a taste for tissue remodeling. Immunity 2025; 58:781-783. [PMID: 40203807 DOI: 10.1016/j.immuni.2025.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Revised: 03/12/2025] [Accepted: 03/12/2025] [Indexed: 04/11/2025]
Abstract
Specialized T cells can support tissue remodeling, but how T cells contribute to mammary gland remodeling during pregnancy is not fully understood. In a recent Cell issue, Corral et al. demonstrate that self-sensing T cells migrate to the mammary gland where they optimize milk production.
Collapse
Affiliation(s)
- Greet Verstichel
- Center for Autoimmune Disease and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA
| | - Hilde Cheroutre
- Center for Autoimmune Disease and Inflammation, La Jolla Institute for Immunology, La Jolla, CA 92037, USA; Center for Cancer Immunotherapy, La Jolla Institute for Immunology, La Jolla, CA 92037, USA.
| |
Collapse
|
2
|
Corral D, Ansaldo E, Delaleu J, Pichler AC, Kabat J, Oguz C, Teijeiro A, Yong D, Abid M, Rivera CA, Link VM, Yang K, Chi L, Nie J, Kamenyeva O, Fan Y, Chan JKY, Ginhoux F, Bosselut R, Belkaid Y. Mammary intraepithelial lymphocytes promote lactogenesis and offspring fitness. Cell 2025; 188:1662-1680.e24. [PMID: 39954680 DOI: 10.1016/j.cell.2025.01.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 10/24/2024] [Accepted: 01/17/2025] [Indexed: 02/17/2025]
Abstract
Breastfeeding is an obligatory requirement of mammalian survival. This fundamental process is associated with the adaptation of maternal physiology, including the transformation of the mammary gland into a milk-secreting organ. How maternal immunity contributes to mammary gland remodeling and function remains largely unknown. Here, we show that maternal adaptive immunity plays a critical role in shaping lactogenesis. Specifically, physiological adaptation during pregnancy is associated with thymic involution and a paradoxical enrichment in intraepithelial lymphocyte (IEL) precursors that no longer migrate to the gut but instead preferentially accumulate within the mammary gland. IEL precursors differentiate into T-bet-expressing unconventional CD8αα lymphocytes in an IL-15-dependent manner. Mammary IELs control milk production by favoring the differentiation and maturation of contractile and milk-secreting cells, thereby promoting offspring fitness. Altogether, this work uncovers a contribution of the maternal adaptive immune system in organismal remodeling during pregnancy that is associated with mammary gland development and function.
Collapse
Affiliation(s)
- Dan Corral
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| | - Eduard Ansaldo
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jérémie Delaleu
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrea C Pichler
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Juraj Kabat
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Cihan Oguz
- Integrated Data Sciences Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ana Teijeiro
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniel Yong
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mahnoor Abid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Claudia A Rivera
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Verena M Link
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Katharine Yang
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore, Singapore
| | - Liang Chi
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Jia Nie
- Laboratory of Immune Cell Biology and Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Olena Kamenyeva
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yiping Fan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore; Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University Health System, Singapore 117597, Singapore; Obstetrics and Gynecology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Jerry Kok Yen Chan
- Department of Reproductive Medicine, KK Women's and Children's Hospital, Singapore 229899, Singapore; Experimental Fetal Medicine Group, Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University Health System, Singapore 117597, Singapore; Obstetrics and Gynecology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore, Singapore; Gustave Roussy, INSERM U1015, Villejuif, France
| | - Rémy Bosselut
- Laboratory of Immune Cell Biology and Laboratory of Integrative Cancer Immunology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunity Section, Laboratory of Host Immunity and Microbiome, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
3
|
Billiet L, Jansen H, Pille M, Boehme L, Sanchez Sanchez G, De Cock L, Goetgeluk G, Pascal E, De Munter S, Deseins L, Ingels J, Michiels T, De Vos R, Zolfaghari A, Vandamme N, Roels J, Kerre T, Dmitriev RI, Taghon T, Vermijlen D, Vandekerckhove B. ThymoSpheres culture: A model to study human polyclonal unconventional T cells. Eur J Immunol 2024; 54:e2451265. [PMID: 39246170 PMCID: PMC11628907 DOI: 10.1002/eji.202451265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024]
Abstract
In vitro cultures remain crucial for studying the fundamental mechanisms of human T-cell development. Here, we introduce a novel in vitro cultivation system based on ThymoSpheres (TS): dense spheroids consisting of DLL4-expressing stromal cells and human hematopoietic precursor cells, in the absence of thymic epithelial cells. These spheroids are subsequently cultured at the air-liquid interphase. TS generate large numbers of mature T cells, are easy to manipulate, scalable, and can be repeatably sampled to monitor T-cell differentiation. The mature T cells generated from primary human hematopoietic precursor cells were extensively characterized using single-cell RNA and combined T-cell receptor (TCR) sequencing. These predominantly CD8α T cells exhibit transcriptional and TCR CDR3 characteristics similar to the recently described human polyclonal αβ unconventional T cell (UTC) lineage. This includes the expression of hallmark genes associated with agonist selection, such as IKZF2 (Helios), and the expression of various natural killer receptors. The TCR repertoire of these UTCs is polyclonal and enriched for CDR3-associated autoreactive features and early rearrangements of the TCR-α chain. In conclusion, TS cultures offer an intriguing platform to study the development of this human polyclonal UTC lineage and its inducing selection mechanisms.
Collapse
MESH Headings
- Humans
- Cell Differentiation/immunology
- Cells, Cultured
- Cell Culture Techniques/methods
- Thymus Gland/cytology
- Thymus Gland/immunology
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Spheroids, Cellular/immunology
- Hematopoietic Stem Cells/cytology
- Hematopoietic Stem Cells/immunology
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
Collapse
|
4
|
Abstract
The intestinal epithelium, which segregates the highly stimulatory lumen from the underlying tissue, harbors one of the largest lymphocyte populations in the body, intestinal intraepithelial lymphocytes (IELs). IELs must balance tolerance, resistance, and tissue protection to maintain epithelial homeostasis and barrier integrity. This review discusses the ontogeny, environmental imprinting, T cell receptor (TCR) repertoire, and function of intestinal IELs. Despite distinct developmental pathways, IEL subsets share core traits including an epithelium-adapted profile, innate-like properties, cytotoxic potential, and limited TCR diversity. IELs also receive important developmental and functional cues through interactions with epithelial cells, microbiota, and dietary components. The restricted TCR diversity of IELs suggests that a limited set of intestinal antigens drives IEL responses, with potential functional consequences. Finally, IELs play a key role in promoting homeostatic immunity and epithelial barrier integrity but can become pathogenic upon dysregulation. Therefore, IELs represent intriguing but underexamined therapeutic targets for inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Ainsley Lockhart
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA; ,
- Current affiliation: Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA; ,
- Howard Hughes Medical Institute, The Rockefeller University, New York, NY, USA
| | - Angelina M Bilate
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY, USA; ,
| |
Collapse
|
5
|
Guan J, Peske JD, Manoharan Valerio M, Park C, Robey EA, Sadegh-Nasseri S. Commensal bacteria maintain a Qa-1 b-restricted unconventional CD8 + T population in gut epithelium. eLife 2023; 12:RP90466. [PMID: 38127067 PMCID: PMC10735220 DOI: 10.7554/elife.90466] [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: 12/23/2023] Open
Abstract
Intestinal intraepithelial lymphocytes (IELs) are characterized by an unusual phenotype and developmental pathway, yet their specific ligands and functions remain largely unknown. Here by analysis of QFL T cells, a population of CD8+ T cells critical for monitoring the MHC I antigen processing pathway, we established that unconventional Qa-1b-restricted CD8+ T cells are abundant in intestinal epithelium. We found that QFL T cells showed a Qa-1b-dependent unconventional phenotype in the spleen and small intestine of naïve wild-type mice. The splenic QFL T cells showed innate-like functionality exemplified by rapid response to cytokines or antigens, while the gut population was refractory to stimuli. Microbiota was required for the maintenance, but not the initial gut homing of QFL T cells. Moreover, monocolonization with Pediococcus pentosaceus, which expresses a peptide that cross-activated QFL T cells, was sufficient to maintain QFL T cells in the intestine. Thus, microbiota is critical for shaping the Qa-1b-restricted IEL landscape.
Collapse
Affiliation(s)
- Jian Guan
- Department of Pathology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Institute of Cell Engineering, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - J David Peske
- Department of Pathology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Institute of Cell Engineering, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Michael Manoharan Valerio
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Chansu Park
- Department of Pathology, Johns Hopkins University School of MedicineBaltimoreUnited States
- Institute of Cell Engineering, Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Ellen A Robey
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | | |
Collapse
|
6
|
Manoharan Valerio M, Arana K, Guan J, Chan SW, Yang X, Kurd N, Lee A, Shastri N, Coscoy L, Robey EA. The promiscuous development of an unconventional Qa1b-restricted T cell population. Front Immunol 2023; 14:1250316. [PMID: 38022509 PMCID: PMC10644506 DOI: 10.3389/fimmu.2023.1250316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 10/06/2023] [Indexed: 12/01/2023] Open
Abstract
MHC-E restricted CD8 T cells show promise in vaccine settings, but their development and specificity remain poorly understood. Here we focus on a CD8 T cell population reactive to a self-peptide (FL9) bound to mouse MHC-E (Qa-1b) that is presented in response to loss of the MHC I processing enzyme ERAAP, termed QFL T cells. We find that mature QFL thymocytes are predominantly CD8αβ+CD4-, show signs of agonist selection, and give rise to both CD8αα and CD8αβ intraepithelial lymphocytes (IEL), as well as memory phenotype CD8αβ T cells. QFL T cells require the MHC I subunit β-2 microglobulin (β2m), but do not require Qa1b or classical MHC I for positive selection. However, QFL thymocytes do require Qa1b for agonist selection and full functionality. Our data highlight the relaxed requirements for positive selection of an MHC-E restricted T cell population and suggest a CD8αβ+CD4- pathway for development of CD8αα IELs.
Collapse
Affiliation(s)
- Michael Manoharan Valerio
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Kathya Arana
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Jian Guan
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Shiao Wei Chan
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Xiaokun Yang
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Nadia Kurd
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Angus Lee
- Gene Targeting Facility Cancer Research Laboratory, University of California Berkeley, Berkeley, CA, United States
| | - Nilabh Shastri
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Laurent Coscoy
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| | - Ellen A. Robey
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, United States
| |
Collapse
|
7
|
Guan J, Peske JD, Valerio MM, Park C, Robey EA, Sadegh-Nasseri S. Commensal Bacteria Maintain a Qa-1 b -restricted Unconventional CD8 + T Population in Gut Epithelium. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.01.530600. [PMID: 36909616 PMCID: PMC10002720 DOI: 10.1101/2023.03.01.530600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Intestinal intraepithelial lymphocytes (IELs) are characterized by an unusual phenotype and developmental pathway, yet their specific ligands and functions remain largely unknown. Here by analysis of QFL T cells, a population of CD8 + T cells critical for monitoring the MHC I antigen processing pathway, we established that unconventional Qa-1 b -restricted CD8 + T cells are abundant in intestinal epithelium. We found that QFL T cells showed a Qa-1 b -dependent unconventional phenotype in the spleen and small intestine of naïve wild-type mice. The splenic QFL T cells showed innate-like functionality exemplified by rapid response to cytokines or antigen, while the gut population was refractory to stimuli. Microbiota was required for the maintenance, but not the initial gut homing of QFL T cells. Moreover, monocolonization with Pediococcus pentosaceus, which expresses a peptide that cross-activated QFL T cells, was sufficient to maintain QFL T cells in the intestine. Thus, microbiota is critical for shaping the Qa-1 b -restricted IEL landscape.
Collapse
|
8
|
Seo GY, Takahashi D, Wang Q, Mikulski Z, Chen A, Chou TF, Marcovecchio P, McArdle S, Sethi A, Shui JW, Takahashi M, Surh CD, Cheroutre H, Kronenberg M. Epithelial HVEM maintains intraepithelial T cell survival and contributes to host protection. Sci Immunol 2022; 7:eabm6931. [PMID: 35905286 PMCID: PMC9422995 DOI: 10.1126/sciimmunol.abm6931] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Intraepithelial T cells (IETs) are in close contact with intestinal epithelial cells and the underlying basement membrane, and they detect invasive pathogens. How intestinal epithelial cells and basement membrane influence IET survival and function, at steady state or after infection, is unclear. The herpes virus entry mediator (HVEM), a member of the TNF receptor superfamily, is constitutively expressed by intestinal epithelial cells and is important for protection from pathogenic bacteria. Here, we showed that at steady-state LIGHT, an HVEM ligand, binding to epithelial HVEM promoted the survival of small intestine IETs. RNA-seq and addition of HVEM ligands to epithelial organoids indicated that HVEM increased epithelial synthesis of basement membrane proteins, including collagen IV, which bound to β1 integrins expressed by IETs. Therefore, we proposed that IET survival depended on β1 integrin binding to collagen IV and showed that β1 integrin-collagen IV interactions supported IET survival in vitro. Moreover, the absence of β1 integrin expression by T lymphocytes decreased TCR αβ+ IETs in vivo. Intravital microscopy showed that the patrolling movement of IETs was reduced without epithelial HVEM. As likely consequences of decreased number and movement, protective responses to Salmonella enterica were reduced in mice lacking either epithelial HVEM, HVEM ligands, or β1 integrins. Therefore, IETs, at steady state and after infection, depended on HVEM expressed by epithelial cells for the synthesis of collagen IV by epithelial cells. Collagen IV engaged β1 integrins on IETs that were important for their maintenance and for their protective function in mucosal immunity.
Collapse
Affiliation(s)
- Goo-Young Seo
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Qingyang Wang
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Angeline Chen
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | | | - Sara McArdle
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Ashu Sethi
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Jr-Wen Shui
- La Jolla Institute for Immunology, La Jolla, CA, USA
| | | | - Charles D Surh
- La Jolla Institute for Immunology, La Jolla, CA, USA.,Institute for Basic Science (IBS), Academy of Immunology and Microbiology, Pohang, South Korea
| | | | - Mitchell Kronenberg
- La Jolla Institute for Immunology, La Jolla, CA, USA.,Division of Biology, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
9
|
Huseby ES, Teixeiro E. The perception and response of T cells to a changing environment are based on the law of initial value. Sci Signal 2022; 15:eabj9842. [PMID: 35639856 PMCID: PMC9290192 DOI: 10.1126/scisignal.abj9842] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
αβ T cells are critical components of the adaptive immune system and are capable of inducing sterilizing immunity after pathogen infection and eliminating transformed tumor cells. The development and function of T cells are controlled through the T cell antigen receptor, which recognizes peptides displayed on major histocompatibility complex (MHC) molecules. Here, we review how T cells generate the ability to recognize self-peptide-bound MHC molecules and use signals derived from these interactions to instruct cellular development, activation thresholds, and functional specialization in the steady state and during immune responses. We argue that the basic tenants of T cell development and function follow Weber-Fetcher's law of just noticeable differences and Wilder's law of initial value. Together, these laws argue that the ability of a system to respond and the quality of that response are scalable to the basal state of that system. Manifestation of these laws in T cells generates clone-specific activation thresholds that are based on perceivable differences between homeostasis and pathogen encounter (self versus nonself discrimination), as well as poised states for subsequent differentiation into specific effector cell lineages.
Collapse
Affiliation(s)
- Eric S. Huseby
- Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Emma Teixeiro
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
10
|
Programme of self-reactive innate-like T cell-mediated cancer immunity. Nature 2022; 605:139-145. [PMID: 35444279 DOI: 10.1038/s41586-022-04632-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 03/10/2022] [Indexed: 12/12/2022]
Abstract
Cellular transformation induces phenotypically diverse populations of tumour-infiltrating T cells1-5, and immune checkpoint blockade therapies preferentially target T cells that recognize cancer cell neoantigens6,7. Yet, how other classes of tumour-infiltrating T cells contribute to cancer immunosurveillance remains elusive. Here, in a survey of T cells in mouse and human malignancies, we identified a population of αβ T cell receptor (TCR)-positive FCER1G-expressing innate-like T cells with high cytotoxic potential8 (ILTCKs). These cells were broadly reactive to unmutated self-antigens, arose from distinct thymic progenitors following early encounter with cognate antigens, and were continuously replenished by thymic progenitors during tumour progression. Notably, expansion and effector differentiation of intratumoural ILTCKs depended on interleukin-15 (IL-15) expression in cancer cells, and inducible activation of IL-15 signalling in adoptively transferred ILTCK progenitors suppressed tumour growth. Thus, the antigen receptor self-reactivity, unique ontogeny, and distinct cancer cell-sensing mechanism distinguish ILTCKs from conventional cytotoxic T cells, and define a new class of tumour-elicited immune response.
Collapse
|
11
|
Nie J, Carpenter AC, Chopp LB, Chen T, Balmaceno-Criss M, Ciucci T, Xiao Q, Kelly MC, McGavern DB, Belkaid Y, Bosselut R. The transcription factor LRF promotes integrin β7 expression by and gut homing of CD8αα + intraepithelial lymphocyte precursors. Nat Immunol 2022; 23:594-604. [PMID: 35354951 PMCID: PMC9290758 DOI: 10.1038/s41590-022-01161-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 02/10/2022] [Indexed: 11/08/2022]
Abstract
While T cell receptor (TCR) αβ+CD8α+CD8β- intraepithelial lymphocytes (CD8αα+ IELs) differentiate from thymic IEL precursors (IELps) and contribute to gut homeostasis, the transcriptional control of their development remains poorly understood. In the present study we showed that mouse thymocytes deficient for the transcription factor leukemia/lymphoma-related factor (LRF) failed to generate TCRαβ+CD8αα+ IELs and their CD8β-expressing counterparts, despite giving rise to thymus and spleen CD8αβ+ T cells. LRF-deficient IELps failed to migrate to the intestine and to protect against T cell-induced colitis, and had impaired expression of the gut-homing integrin α4β7. Single-cell RNA-sequencing found that LRF was necessary for the expression of genes characteristic of the most mature IELps, including Itgb7, encoding the β7 subunit of α4β7. Chromatin immunoprecipitation and gene-regulatory network analyses both defined Itgb7 as an LRF target. Our study identifies LRF as an essential transcriptional regulator of IELp maturation in the thymus and subsequent migration to the intestinal epithelium.
Collapse
Affiliation(s)
- Jia Nie
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Andrea C Carpenter
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD, USA
| | - Laura B Chopp
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
- Immunology Graduate Group, University of Pennsylvania Medical School, Philadelphia, PA, USA
| | - Ting Chen
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Mariah Balmaceno-Criss
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Thomas Ciucci
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
- David H. Smith Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Qi Xiao
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA
| | - Michael C Kelly
- CCR Single Analysis Facility, Cancer Research Technology Program, Frederick National Laboratory for Cancer Research, Bethesda, MD, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, NIH, Bethesda, MD, USA
| | - Yasmine Belkaid
- Metaorganism Immunology Section, Laboratory of Immune System Biology, Bethesda, MD, USA
- Microbiome core, National Institute of Allergy and Infectious Disease, NIH, Bethesda, MD, USA
| | - Rémy Bosselut
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health (NIH), Bethesda, MD, USA.
| |
Collapse
|
12
|
Mayassi T, Barreiro LB, Rossjohn J, Jabri B. A multilayered immune system through the lens of unconventional T cells. Nature 2021; 595:501-510. [PMID: 34290426 PMCID: PMC8514118 DOI: 10.1038/s41586-021-03578-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/23/2021] [Indexed: 02/07/2023]
Abstract
The unconventional T cell compartment encompasses a variety of cell subsets that straddle the line between innate and adaptive immunity, often reside at mucosal surfaces and can recognize a wide range of non-polymorphic ligands. Recent advances have highlighted the role of unconventional T cells in tissue homeostasis and disease. In this Review, we recast unconventional T cell subsets according to the class of ligand that they recognize; their expression of semi-invariant or diverse T cell receptors; the structural features that underlie ligand recognition; their acquisition of effector functions in the thymus or periphery; and their distinct functional properties. Unconventional T cells follow specific selection rules and are poised to recognize self or evolutionarily conserved microbial antigens. We discuss these features from an evolutionary perspective to provide insights into the development and function of unconventional T cells. Finally, we elaborate on the functional redundancy of unconventional T cells and their relationship to subsets of innate and adaptive lymphoid cells, and propose that the unconventional T cell compartment has a critical role in our survival by expanding and complementing the role of the conventional T cell compartment in protective immunity, tissue healing and barrier function.
Collapse
Affiliation(s)
- Toufic Mayassi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Committee on Immunology, University of Chicago, Chicago, IL, USA
| | - Luis B. Barreiro
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Committee on Genetics, Genomics, and Systems Biology, University of Chicago, Chicago, IL, USA.,Section of Genetic Medicine, Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Jamie Rossjohn
- Infection and Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Institute of Infection and Immunity, Cardiff University, School of Medicine, Heath Park, Cardiff, UK
| | - Bana Jabri
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Medicine, University of Chicago, Chicago, IL, USA.,Department of Pathology, University of Chicago, Chicago, IL, USA.,Department of Pediatrics, University of Chicago, Chicago, IL, USA.,Correspondence and requests for materials should be addressed to B.J.,
| |
Collapse
|
13
|
Lee ST, Georgiev H, Breed ER, Ruscher R, Hogquist KA. MHC Class I on murine hematopoietic APC selects Type A IEL precursors in the thymus. Eur J Immunol 2021; 51:1080-1088. [PMID: 33521937 PMCID: PMC9846822 DOI: 10.1002/eji.202048996] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/06/2020] [Accepted: 01/29/2021] [Indexed: 01/21/2023]
Abstract
TCRαβ+ CD8α+ CD8β- intestinal intraepithelial lymphocytes (CD8αα IEL) are gut T cells that maintain barrier surface homeostasis. Most CD8αα IEL are derived from thymic precursors (IELp) through a mechanism referred to as clonal diversion. In this model, self-reactive thymocytes undergo deletion in the presence of CD28 costimulation, but in its absence undergo diversion to the IEL fate. While previous reports showed that IELp were largely β2m dependent, the APC that drive the development of these cells are poorly defined. We found that both CD80 and CD86 restrain IELp development, and conventional DCs play a prominent role. We sought to define a CD80/86 negative, MHCI positive APC that supports the development to the IEL lineage. Chimera studies showed that MHCI needs to be expressed on hematopoietic APC for selection. As thymic hematopoietic APC are heterogeneous in their expression of MHCI and costimulatory molecules, we identified four thymic APC types that were CD80/86neg/low and MHCI+ . However, selective depletion of β2m in individual APC suggested functional redundancy. Thus, while hematopoietic APC play a critical role in clonal diversion, no single APC subset is specialized to promote the CD8αα IEL fate.
Collapse
Affiliation(s)
| | | | | | - Roland Ruscher
- Corresponding authors: Kristin Hogquist, , Roland Ruscher,
| | | |
Collapse
|
14
|
Ruscher R, Lee ST, Salgado OC, Breed ER, Osum SH, Hogquist KA. Intestinal CD8αα IELs derived from two distinct thymic precursors have staggered ontogeny. J Exp Med 2021; 217:151959. [PMID: 32687575 PMCID: PMC7398160 DOI: 10.1084/jem.20192336] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 04/27/2020] [Accepted: 06/12/2020] [Indexed: 12/13/2022] Open
Abstract
CD8αα intraepithelial lymphocytes (IELs) are abundant T cells that protect the gut epithelium. Their thymic precursors (IELps) include PD-1+ type A and Tbet+ type B populations, which differ in their antigen-receptor specificities. To better understand CD8αα IEL ontogeny, we performed "time-stamp" fate mapping experiments and observed that it seeds the intestine predominantly during a narrow time window in early life. Adoptively transferred IELps parked better in the intestines of young mice than in adults. In young mice, both type A and type B IELps had an S1PR1+ and α4β7+ emigration- and mucosal-homing competent phenotype, while this was restricted to type A IELps in adults. Only CD8αα IELs established in early life were enriched in cells bearing type B IELp TCR usage. Together, our results suggest that the young intestine facilitates CD8αα IEL establishment and that early IELs are distinct from IELs established after this initial wave. These data provide novel insight into the ontogeny of CD8αα IELs.
Collapse
Affiliation(s)
- Roland Ruscher
- Center for Immunology and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - S Thera Lee
- Center for Immunology and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Oscar C Salgado
- Center for Immunology and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Elise R Breed
- Center for Immunology and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Sara H Osum
- Center for Immunology and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| | - Kristin A Hogquist
- Center for Immunology and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN
| |
Collapse
|
15
|
Kurd NS, Hoover A, Yoon J, Weist BM, Lutes L, Chan SW, Robey EA. Factors that influence the thymic selection of CD8αα intraepithelial lymphocytes. Mucosal Immunol 2021; 14:68-79. [PMID: 32483197 PMCID: PMC10443950 DOI: 10.1038/s41385-020-0295-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 03/19/2020] [Accepted: 04/06/2020] [Indexed: 02/04/2023]
Abstract
Thymocytes bearing αβ T cell receptors (TCRαβ) with high affinity for self-peptide-MHC complexes undergo negative selection or are diverted to alternate T cell lineages, a process termed agonist selection. Among thymocytes bearing TCRs restricted to MHC class I, agonist selection can lead to the development of precursors that can home to the gut and give rise to CD8αα-expressing intraepithelial lymphocytes (CD8αα IELs). The factors that influence the choice between negative selection versus CD8αα IEL development remain largely unknown. Using a synchronized thymic tissue slice model that supports both negative selection and CD8αα IEL development, we show that the affinity threshold for CD8αα IEL development is higher than for negative selection. We also investigate the impact of peptide presenting cells and cytokines, and the migration patterns associated with these alternative cell fates. Our data highlight the roles of TCR affinity and the thymic microenvironments on T cell fate.
Collapse
Affiliation(s)
- Nadia S Kurd
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Department of Medicine, University of California San Diego, San Diego, CA, 92093, USA
| | - Ashley Hoover
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Jaewon Yoon
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Brian M Weist
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
- Gilead Sciences, Foster City, CA, 94404, USA
| | - Lydia Lutes
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Shiao Wei Chan
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA
| | - Ellen A Robey
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA, 94720, USA.
| |
Collapse
|
16
|
Watanabe M, Lu Y, Breen M, Hodes RJ. B7-CD28 co-stimulation modulates central tolerance via thymic clonal deletion and Treg generation through distinct mechanisms. Nat Commun 2020; 11:6264. [PMID: 33293517 PMCID: PMC7722925 DOI: 10.1038/s41467-020-20070-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Accepted: 11/09/2020] [Indexed: 12/22/2022] Open
Abstract
The molecular and cellular mechanisms mediating thymic central tolerance and prevention of autoimmunity are not fully understood. Here we show that B7-CD28 co-stimulation and B7 expression by specific antigen-presenting cell (APC) types are required for clonal deletion and for regulatory T (Treg) cell generation from endogenous tissue-restricted antigen (TRA)-specific thymocytes. While B7-CD28 interaction is required for both clonal deletion and Treg induction, these two processes differ in their CD28 signaling requirements and in their dependence on B7-expressing dendritic cells, B cells, and thymic epithelial cells. Meanwhile, defective thymic clonal deletion due to altered B7-CD28 signaling results in the accumulation of mature, peripheral TRA-specific T cells capable of mediating destructive autoimmunity. Our findings thus reveal a function of B7-CD28 co-stimulation in shaping the T cell repertoire and limiting autoimmunity through both thymic clonal deletion and Treg cell generation.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/metabolism
- Autoimmunity/physiology
- B7-1 Antigen/metabolism
- CD28 Antigens/genetics
- CD28 Antigens/metabolism
- Cell Differentiation/immunology
- Central Tolerance
- Clonal Deletion
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Flow Cytometry
- Gene Knock-In Techniques
- Mice
- Mice, Knockout
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Signal Transduction/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Thymocytes/physiology
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
Collapse
Affiliation(s)
- Masashi Watanabe
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Ying Lu
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Michael Breen
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Richard J Hodes
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD, 20892, USA.
| |
Collapse
|
17
|
Human Thymic CD10 + PD-1 + Intraepithelial Lymphocyte Precursors Acquire Interleukin-15 Responsiveness at the CD1a - CD95 + CD28 - CCR7 - Developmental Stage. Int J Mol Sci 2020; 21:ijms21228785. [PMID: 33233766 PMCID: PMC7699974 DOI: 10.3390/ijms21228785] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/15/2020] [Accepted: 11/17/2020] [Indexed: 11/24/2022] Open
Abstract
Human thymic CD8αα+ CD10+ PD-1+ αβ T cells selected through early agonist selection have been proposed as the putative thymic precursors of the human CD8αα+ intestinal intraepithelial lymphocytes (IELs). However, the progeny of these thymic precursor cells in human blood or tissues has not yet been characterized. Here, we studied the phenotypical and transcriptional differentiation of the thymic IEL precursor (IELp) lineage upon in vitro exposure to cytokines prominent in the peripheral tissues such as interleukin-15 (IL-15) and the inflammatory cytokines interleukin-12 (IL-12) and interleukin-18 (IL-18). We showed that only the CD1a− fraction of the CD10+ PD-1+ IELp population was able to proliferate with IL-15, suggesting that this subset had acquired functionality. These cells downregulated PD-1 expression and completely lost CD10 expression, whereas other surface markers such as CD95 and CXCR3 remained highly expressed. RNA-seq analysis of the IL-15-cultured cells clearly showed induction of innate-like and effector genes. Induction of the cytotoxic machinery by the CD10+ PD-1+ population was acquired in the presence of IL-15 and was further augmented by inflammatory cytokines. Our data suggest that only the CD1a− CD10+ PD-1+ population exits the thymus and survives in the periphery. Furthermore, PD-1 and CD10 expression is not an intrinsic property of this lineage, but rather characterizes a transient stage in differentiation. CD95 and CXCR3 expression combined with the absence of CD28, CCR7, and CD6 expression might be more powerful markers to define this lineage in the periphery.
Collapse
|
18
|
Bilate AM, London M, Castro TBR, Mesin L, Bortolatto J, Kongthong S, Harnagel A, Victora GD, Mucida D. T Cell Receptor Is Required for Differentiation, but Not Maintenance, of Intestinal CD4 + Intraepithelial Lymphocytes. Immunity 2020; 53:1001-1014.e20. [PMID: 33022229 PMCID: PMC7677182 DOI: 10.1016/j.immuni.2020.09.003] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 07/02/2020] [Accepted: 09/09/2020] [Indexed: 01/28/2023]
Abstract
The gut epithelium is populated by intraepithelial lymphocytes (IELs), a heterogeneous T cell population with cytotoxic and regulatory properties, which can be acquired at the epithelial layer. However, the role of T cell receptor (TCR) in this process remains unclear. Single-cell transcriptomic analyses revealed distinct clonal expansions between cell states, with CD4+CD8αα+ IELs being one of the least diverse populations. Conditional deletion of TCR on differentiating CD4+ T cells or of major histocompatibility complex (MHC) class II on intestinal epithelial cells prevented CD4+CD8αα+ IEL differentiation. However, TCR ablation on differentiated CD4+CD8αα+ IELs or long-term cognate antigen withdraw did not affect their maintenance. TCR re-engagement of antigen-specific CD4+CD8αα+ IELs by Listeria monocytogenes did not alter their state but correlated with reduced bacterial invasion. Thus, local antigen recognition is an essential signal for differentiation of CD4+ T cells at the epithelium, yet differentiated IELs are able to preserve an effector program in the absence of TCR signaling.
Collapse
Affiliation(s)
- Angelina M Bilate
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA.
| | - Mariya London
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Tiago B R Castro
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA; Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY 10065, USA
| | - Luka Mesin
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY 10065, USA
| | - Juliana Bortolatto
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA; Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY 10065, USA
| | - Suppawat Kongthong
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Audrey Harnagel
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA
| | - Gabriel D Victora
- Laboratory of Lymphocyte Dynamics, The Rockefeller University, New York, NY 10065, USA
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|
19
|
Taming the Sentinels: Microbiome-Derived Metabolites and Polarization of T Cells. Int J Mol Sci 2020; 21:ijms21207740. [PMID: 33086747 PMCID: PMC7589579 DOI: 10.3390/ijms21207740] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/27/2020] [Accepted: 10/11/2020] [Indexed: 02/07/2023] Open
Abstract
A global increase in the prevalence of metabolic syndromes and digestive tract disorders, like food allergy or inflammatory bowel disease (IBD), has become a severe problem in the modern world. Recent decades have brought a growing body of evidence that links the gut microbiome’s complexity with host physiology. Hence, understanding the mechanistic aspects underlying the synergy between the host and its associated gut microbiome are among the most crucial questions. The functionally diversified adaptive immune system plays a central role in maintaining gut and systemic immune homeostasis. The character of the reciprocal interactions between immune components and host-dwelling microbes or microbial consortia determines the outcome of the organisms’ coexistence within the holobiont structure. It has become apparent that metabolic by-products of the microbiome constitute crucial multimodal transmitters within the host–microbiome interactome and, as such, contribute to immune homeostasis by fine-tuning of the adaptive arm of immune system. In this review, we will present recent insights and discoveries regarding the broad landscape of microbiome-derived metabolites, highlighting the role of these small compounds in the context of the balance between pro- and anti-inflammatory mechanisms orchestrated by the host T cell compartment.
Collapse
|
20
|
Vandereyken M, James OJ, Swamy M. Mechanisms of activation of innate-like intraepithelial T lymphocytes. Mucosal Immunol 2020; 13:721-731. [PMID: 32415229 PMCID: PMC7434593 DOI: 10.1038/s41385-020-0294-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 04/09/2020] [Accepted: 04/11/2020] [Indexed: 02/04/2023]
Abstract
Intraepithelial T lymphocytes (T-IEL) contain subsets of innate-like T cells that evoke innate and adaptive immune responses to provide rapid protection at epithelial barrier sites. In the intestine, T-IEL express variable T cell antigen receptors (TCR), with unknown antigen specificities. Intriguingly, they also express multiple inhibitory receptors, many of which are normally found on exhausted or antigen-experienced T cells. This pattern suggests that T-IEL are antigen-experienced, yet it is not clear where, and in what context, T-IEL encounter TCR ligands. We review recent evidence indicating TCR antigens for intestinal innate-like T-IEL are found on thymic or intestinal epithelium, driving agonist selection of T-IEL. We explore the contributions of the TCR and various co-stimulatory and co-inhibitory receptors in activating T-IEL effector functions. The balance between inhibitory and activating signals may be key to keeping these highly cytotoxic, rapidly activated cells in check, and key to harnessing their immune surveillance potential.
Collapse
Affiliation(s)
- Maud Vandereyken
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Olivia J James
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Mahima Swamy
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK.
| |
Collapse
|
21
|
Wu R, Zhang D, Zanvit P, Jin W, Wang H, Chen W. Identification and Regulation of TCRαβ +CD8αα + Intraepithelial Lymphocytes in Murine Oral Mucosa. Front Immunol 2020; 11:1702. [PMID: 32849598 PMCID: PMC7417446 DOI: 10.3389/fimmu.2020.01702] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 06/25/2020] [Indexed: 11/13/2022] Open
Abstract
TCRαβ+CD8αα+ intraepithelial lymphocytes (IELs) are abundant in gastrointestinal (GI) tract and play an important role in regulation of mucosal immunity and tolerance in the gut. However, it is unknown whether TCRαβ+CD8αα+ IELs exist in the oral mucosa and if yes, what controls their development. We here identified and characterized TCRαβ+CD8αα+ IELs from the murine oral mucosa. We showed that the number and function of TCRαβ+CD8αα+ IELs were regulated by TGF-β. We further revealed that oral TCRαβ+CD8αα+ IELs could be altered under systemic inflammatory conditions and by antibiotic treatment at the neonatal age of the mice. Our findings have revealed a previously unrecognized population of oral IELs that may regulate oral mucosal immune responses.
Collapse
Affiliation(s)
- Ruiqing Wu
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States.,Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Dunfang Zhang
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Peter Zanvit
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Wenwen Jin
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| | - Hao Wang
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States.,Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - WanJun Chen
- Mucosal Immunology Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
22
|
Collin R, Lombard-Vadnais F, Hillhouse EE, Lebel MÈ, Chabot-Roy G, Melichar HJ, Lesage S. MHC-Independent Thymic Selection of CD4 and CD8 Coreceptor Negative αβ T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 205:133-142. [PMID: 32434937 DOI: 10.4049/jimmunol.2000156] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/23/2020] [Indexed: 12/12/2022]
Abstract
It is becoming increasingly clear that unconventional T cell subsets, such as NKT, γδ T, mucosal-associated invariant T, and CD8αα T cells, each play distinct roles in the immune response. Subsets of these cell types can lack both CD4 and CD8 coreceptor expression. Beyond these known subsets, we identify CD4-CD8-TCRαβ+, double-negative (DN) T cells, in mouse secondary lymphoid organs. DN T cells are a unique unconventional thymic-derived T cell subset. In contrast to CD5high DN thymocytes that preferentially yield TCRαβ+ CD8αα intestinal lymphocytes, we find that mature CD5low DN thymocytes are precursors to peripheral DN T cells. Using reporter mouse strains, we show that DN T cells transit through the immature CD4+CD8+ (double-positive) thymocyte stage. Moreover, we provide evidence that DN T cells can differentiate in MHC-deficient mice. Our study demonstrates that MHC-independent thymic selection can yield DN T cells that are distinct from NKT, γδ T, mucosal-associated invariant T, and CD8αα T cells.
Collapse
Affiliation(s)
- Roxanne Collin
- Immunology-Oncology Section, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| | - Félix Lombard-Vadnais
- Immunology-Oncology Section, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, Quebec H3A 0G4, Canada; and
| | - Erin E Hillhouse
- Immunology-Oncology Section, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada
| | - Marie-Ève Lebel
- Immunology-Oncology Section, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada
| | - Geneviève Chabot-Roy
- Immunology-Oncology Section, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada
| | - Heather J Melichar
- Immunology-Oncology Section, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada.,Département de Médecine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Sylvie Lesage
- Immunology-Oncology Section, Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec H1T 2M4, Canada; .,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
23
|
Rodríguez-Rodríguez N, Flores-Mendoza G, Apostolidis SA, Rosetti F, Tsokos GC, Crispín JC. TCR-α/β CD4 - CD8 - double negative T cells arise from CD8 + T cells. J Leukoc Biol 2020; 108:851-857. [PMID: 32052478 DOI: 10.1002/jlb.1ab0120-548r] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/30/2020] [Accepted: 02/01/2020] [Indexed: 01/02/2023] Open
Abstract
The cellular origin of CD4- CD8- (double negative, DNT) TCR-α/β+ T cells remains unknown. Available evidence indicates that they may derive from CD8+ T cells, but most published data have been obtained using cells that bear an invariant transgenic T cell receptor that recognizes an Ag that is not present in normal mice. Here, we have used complementary fate mapping and adoptive transfer experiments to identify the cellular lineage of origin of DNT cells in wild-type mice with a polyclonal T cell repertoire. We show that TCR-α/β+ DNT cells can be traced back to CD8+ and CD4+ CD8+ double positive cells in the thymus. We also demonstrate that polyclonal DNT cells generated in secondary lymphoid organs proliferate upon adoptive transfer and can regain CD8 expression in lymphopenic environment. These results demonstrate the cellular origin of DNT cells and provide a conceptual framework to understand their presence in pathological circumstances.
Collapse
Affiliation(s)
- Noé Rodríguez-Rodríguez
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Current address: Medical Research Council Laboratory of Molecular Biology, Cambridge, UK
| | - Giovanna Flores-Mendoza
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sokratis A Apostolidis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Current address: Division of Rheumatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Florencia Rosetti
- Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - George C Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - José C Crispín
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA.,Department of Immunology and Rheumatology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Escuela de Medicina y Ciencias de la Salud, Tecnologico de Monterrey, Mexico City, Mexico
| |
Collapse
|
24
|
Single-cell RNA-sequencing identifies the developmental trajectory of C-Myc-dependent NK1.1 - T-bet + intraepithelial lymphocyte precursors. Mucosal Immunol 2020; 13:257-270. [PMID: 31712600 PMCID: PMC7039806 DOI: 10.1038/s41385-019-0220-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 10/22/2019] [Indexed: 02/04/2023]
Abstract
Natural intraepithelial lymphocytes (IELs) are thymus-derived adaptive immune cells, which are important contributors to intestinal immune homeostasis. Similar to other innate-like T cells, they are induced in the thymus through high-avidity interaction that would otherwise lead to clonal deletion in conventional CD4 and CD8 T cells. By applying single-cell RNA-sequencing (scRNA-seq) on a heterogeneous population of thymic CD4-CD8αβ-TCRαβ+NK1.1- IEL precursors (NK1.1- IELPs), we define a developmental trajectory that can be tracked based on the sequential expression of CD122 and T-bet. Moreover, we identify the Id proteins Id2 and Id3 as a novel regulator of IELP development and show that all NK1.1- IELPs progress through a PD-1 stage that precedes the induction of T-bet. The transition from PD-1 to T-bet is regulated by the transcription factor C-Myc, which has far reaching effects on cell cycle, energy metabolism, and the translational machinery during IELP development. In summary, our results provide a high-resolution molecular framework for thymic IEL development of NK1.1- IELPs and deepen our understanding of this still elusive cell type.
Collapse
|
25
|
Kurd NS, Lutes LK, Yoon J, Chan SW, Dzhagalov IL, Hoover AR, Robey EA. A role for phagocytosis in inducing cell death during thymocyte negative selection. eLife 2019; 8:48097. [PMID: 31868579 PMCID: PMC6957271 DOI: 10.7554/elife.48097] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 12/21/2019] [Indexed: 12/11/2022] Open
Abstract
Autoreactive thymocytes are eliminated during negative selection in the thymus, a process important for establishing self-tolerance. Thymic phagocytes serve to remove dead thymocytes, but whether they play additional roles during negative selection remains unclear. Here, using a murine thymic slice model in which thymocytes undergo negative selection in situ, we demonstrate that phagocytosis promotes negative selection, and provide evidence for the escape of autoreactive CD8 T cells to the periphery when phagocytosis in the thymus is impaired. We also show that negative selection is more efficient when the phagocyte also presents the negative selecting peptide. Our findings support a model for negative selection in which the death process initiated following strong TCR signaling is facilitated by phagocytosis. Thus, the phagocytic capability of cells that present self-peptides is a key determinant of thymocyte fate.
Collapse
Affiliation(s)
- Nadia S Kurd
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Lydia K Lutes
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Jaewon Yoon
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Shiao Wei Chan
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Ivan L Dzhagalov
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Ashley R Hoover
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| | - Ellen A Robey
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, United States
| |
Collapse
|
26
|
McDonald BD, Jabri B, Bendelac A. Diverse developmental pathways of intestinal intraepithelial lymphocytes. Nat Rev Immunol 2019; 18:514-525. [PMID: 29717233 DOI: 10.1038/s41577-018-0013-7] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The intestinal epithelial barrier is patrolled by resident intraepithelial lymphocytes (IELs) that are involved in host defence against pathogens, wound repair and homeostatic interactions with the epithelium, microbiota and nutrients. Intestinal IELs are one of the largest populations of lymphocytes in the body and comprise several distinct subsets, the identity and lineage relationships of which have long remained elusive. Here, we review advances in unravelling the complexity of intestinal IEL populations, which comprise conventional αβ T cell receptor (TCRαβ)+ subsets, unconventional TCRαβ+ and TCRγδ+ subsets, group 1 innate lymphoid cells (ILC1s) and ILC1-like cells. Although these intestinal IEL lineages have partially overlapping effector programmes and recognition properties, they have strikingly different developmental pathways. We suggest that evolutionary pressure has driven the recurrent generation of cytolytic effector lymphocytes to protect the intestinal epithelial layer, but they may also precipitate intestinal inflammatory disorders, such as coeliac disease.
Collapse
Affiliation(s)
- Benjamin D McDonald
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Pathology, University of Chicago, Chicago, IL, USA.,Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Bana Jabri
- Committee on Immunology, University of Chicago, Chicago, IL, USA.,Department of Pathology, University of Chicago, Chicago, IL, USA.,Department of Medicine, University of Chicago, Chicago, IL, USA
| | - Albert Bendelac
- Committee on Immunology, University of Chicago, Chicago, IL, USA. .,Department of Pathology, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
27
|
Ruscher R, Hogquist KA. Development, ontogeny, and maintenance of TCRαβ + CD8αα IEL. Curr Opin Immunol 2019; 58:83-88. [PMID: 31146182 DOI: 10.1016/j.coi.2019.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 04/19/2019] [Indexed: 02/07/2023]
Abstract
The intestinal epithelium is the outermost cellular layer that separates the body from the gut lumen. The integrity of this protective mucosal barrier is crucial and maintained by specialized cells-intraepithelial lymphocytes (IEL). Much research has been conducted on these cells and our overall understanding of them is increasing rapidly. In this review we focus on the TCRαβ+ subset of CD8αα IEL. We discuss recent studies that shed light on the development, ontogeny, maintenance, and functional characteristics of CD8αα IEL, and highlight yet unanswered questions for future studies.
Collapse
Affiliation(s)
- Roland Ruscher
- Center for Immunology and Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Kristin A Hogquist
- Center for Immunology and Department of Laboratory Medicine & Pathology, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
28
|
Deletion of self-reactive CCR7- thymocytes in the absence of MHC expression on thymic epithelial cells. Cell Death Differ 2019; 26:2727-2739. [PMID: 31019259 DOI: 10.1038/s41418-019-0331-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 03/18/2019] [Accepted: 04/05/2019] [Indexed: 12/11/2022] Open
Abstract
The selection of αβ T cells in the thymus is punctuated by checkpoints at which thymocytes differentiate or undergo apoptosis. Wave 1 deletion is defined as apoptosis within nascent αβ T-cell antigen receptor (TCR)-signalled thymocytes that lack CCR7 expression. The antigen-presenting cell (APC) types that mediate wave 1 deletion are unclear. To measure wave 1 deletion, we compared the frequencies of TCRβ + CD5 + Helios + CCR7- cells in nascent thymocyte cohorts in mice with normal or defective apoptosis. This thymocyte population is small in mice lacking major histocompatibility complex (MHC) expression. The scale of wave 1 deletion was increased by transgenic expression of the self-reactive Yae62 TCRβ chain, was almost halved when haemopoietic APCs lacked MHC expression and, surprisingly, was unchanged when epithelial cells lacked MHC expression. These findings demonstrate efficiency, and some redundancy, in the APC types that mediate wave 1 deletion in the normal mouse thymus.
Collapse
|
29
|
Cippà PE, Liu J, Sun B, Kumar S, Naesens M, McMahon AP. A late B lymphocyte action in dysfunctional tissue repair following kidney injury and transplantation. Nat Commun 2019; 10:1157. [PMID: 30858375 PMCID: PMC6411919 DOI: 10.1038/s41467-019-09092-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 02/20/2019] [Indexed: 12/24/2022] Open
Abstract
The mechanisms initiating late immune responses to an allograft are poorly understood. Here we show, via transcriptome analysis of serial protocol biopsies from kidney transplants, that the initial responses to kidney injury correlate with a late B lymphocyte signature relating to renal dysfunction and fibrosis. With a potential link between dysfunctional repair and immunoreactivity, we investigate the immunological consequences of dysfunctional repair examining chronic disease in mouse kidneys 18 months after a bilateral ischemia/reperfusion injury event. In the absence of foreign antigens, a sustained immune response involving both innate and adaptive immune systems accompanies a transition to chronic kidney damage. At late stages, B lymphocytes exhibite an antigen-driven proliferation, selection and maturation into broadly-reacting antibody-secreting cells. These findings reveal a previously unappreciated role for dysfunctional tissue repair in local immunomodulation that may have particular relevance to transplant-associated immunobiology.
Collapse
Affiliation(s)
- Pietro E Cippà
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, 90033-9080, CA, USA.
- Division of Nephrology, Regional Hospital Lugano, Lugano, 6900, Switzerland.
| | - Jing Liu
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, 90033-9080, CA, USA
| | - Bo Sun
- Molecular and Computational Biology, University of Southern California, Los Angeles, 90089-2910, CA, USA
| | - Sanjeev Kumar
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, 90033-9080, CA, USA
| | - Maarten Naesens
- Department of Microbiology and Immunology, KU Leuven, Leuven, 3000, Belgium
- Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, 3000, Belgium
| | - Andrew P McMahon
- Department of Stem Cell Biology and Regenerative Medicine, Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research, University of Southern California, Los Angeles, 90033-9080, CA, USA.
| |
Collapse
|
30
|
Van Kaer L, Olivares-Villagómez D. Development, Homeostasis, and Functions of Intestinal Intraepithelial Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2019; 200:2235-2244. [PMID: 29555677 PMCID: PMC5863587 DOI: 10.4049/jimmunol.1701704] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/25/2018] [Indexed: 12/13/2022]
Abstract
The intestine is continuously exposed to commensal microorganisms, food, and environmental agents and also serves as a major portal of entry for many pathogens. A critical defense mechanism against microbial invasion in the intestine is the single layer of epithelial cells that separates the gut lumen from the underlying tissues. The barrier function of the intestinal epithelium is supported by cells and soluble factors of the intestinal immune system. Chief among them are intestinal intraepithelial lymphocytes (iIELs), which are embedded in the intestinal epithelium and represent one of the single largest populations of lymphocytes in the body. Compared with lymphocytes in other parts of the body, iIELs exhibit unique phenotypic, developmental, and functional properties that reflect their key roles in maintaining the intestinal epithelial barrier. In this article, we review the biology of iIELs in supporting normal health and how their dysregulation can contribute to disease.
Collapse
Affiliation(s)
- Luc Van Kaer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| | - Danyvid Olivares-Villagómez
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232
| |
Collapse
|
31
|
Chou C, Li MO. Tissue-Resident Lymphocytes Across Innate and Adaptive Lineages. Front Immunol 2018; 9:2104. [PMID: 30298068 PMCID: PMC6160555 DOI: 10.3389/fimmu.2018.02104] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 08/28/2018] [Indexed: 01/08/2023] Open
Abstract
Lymphocytes are an integral component of the immune system. Classically, all lymphocytes were thought to perpetually recirculate between secondary lymphoid organs and only traffic to non-lymphoid tissues upon activation. In recent years, a diverse family of non-circulating lymphocytes have been identified. These include innate lymphocytes, innate-like T cells and a subset of conventional T cells. Spanning the innate-adaptive spectrum, these tissue-resident lymphocytes carry out specialized functions and cross-talk with other immune cell types to maintain tissue integrity and homeostasis both at the steady state and during pathological conditions. In this review, we provide an overview of the heterogeneous tissue-resident lymphocyte populations, discuss their development, and highlight their functions both in the context of microbial infection and cancer.
Collapse
Affiliation(s)
- Chun Chou
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Ming O Li
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| |
Collapse
|
32
|
Mayassi T, Jabri B. Human intraepithelial lymphocytes. Mucosal Immunol 2018; 11:1281-1289. [PMID: 29674648 PMCID: PMC6178824 DOI: 10.1038/s41385-018-0016-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 02/02/2018] [Accepted: 02/03/2018] [Indexed: 02/04/2023]
Abstract
The location of intraepithelial lymphocytes (IEL) between epithelial cells, their effector memory, cytolytic and inflammatory phenotype positions them to kill infected epithelial cells and protect the intestine against pathogens. Human TCRαβ+CD8αβ+ IEL have the dual capacity to recognize modified self via natural killer (NK) receptors (autoreactivity) as well as foreign antigen via the T cell receptor (TCR), which is accomplished in mouse by two cell subsets, the naturally occurring TCRαβ+CD8αα+ and adaptively induced TCRαβ+CD8αβ+ IEL subsets, respectively. The private/oligoclonal nature of the TCR repertoire of both human and mouse IEL suggests local environmental factors dictate the specificity of IEL responses. The line between sensing of foreign antigens and autoreactivity is blurred for IEL in celiac disease, where recognition of stress ligands by induced activating NK receptors in conjunction with inflammatory signals such as IL-15 can result in low-affinity TCR/non-cognate antigen and NK receptor/stress ligand interactions triggering destruction of intestinal epithelial cells.
Collapse
Affiliation(s)
- Toufic Mayassi
- Department of Medicine, University of Chicago, Chicago, USA
- Committee on Immunology, University of Chicago, Chicago, USA
| | - Bana Jabri
- Department of Medicine, University of Chicago, Chicago, USA.
- Committee on Immunology, University of Chicago, Chicago, USA.
- Department of Pathology, University of Chicago, Chicago, USA.
- Department of Pediatrics, University of Chicago, Chicago, USA.
| |
Collapse
|
33
|
Non-canonicaly recruited TCRαβCD8αα IELs recognize microbial antigens. Sci Rep 2018; 8:10848. [PMID: 30022086 PMCID: PMC6052027 DOI: 10.1038/s41598-018-29073-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 06/29/2018] [Indexed: 11/10/2022] Open
Abstract
In the gut, various subsets of intraepithelial T cells (IELs) respond to self or non-self-antigens derived from the body, diet, commensal and pathogenic microbiota. Dominant subset of IELs in the small intestine are TCRαβCD8αα+ cells, which are derived from immature thymocytes that express self-reactive TCRs. Although most of TCRαβCD8αα+ IELs are thymus-derived, their repertoire adapts to microbial flora. Here, using high throughput TCR sequencing we examined how clonal diversity of TCRαβCD8αα+ IELs changes upon exposure to commensal-derived antigens. We found that fraction of CD8αα+ IELs and CD4+ T cells express identical αβTCRs and this overlap raised parallel to a surge in the diversity of microbial flora. We also found that an opportunistic pathogen (Staphylococcus aureus) isolated from mouse small intestine specifically activated CD8αα+ IELs and CD4+ derived T cell hybridomas suggesting that some of TCRαβCD8αα+ clones with microbial specificities have extrathymic origin. We also report that CD8ααCD4+ IELs and Foxp3CD4+ T cells from the small intestine shared many αβTCRs, regardless whether the later subset was isolated from Foxp3CNS1 sufficient or Foxp3CNS1 deficient mice that lacks peripherally-derived Tregs. Overall, our results imply that repertoire of TCRαβCD8αα+ in small intestine expends in situ in response to changes in microbial flora.
Collapse
|
34
|
Schattgen SA, Thomas PG. Bohemian T cell receptors: sketching the repertoires of unconventional lymphocytes. Immunol Rev 2018; 284:79-90. [PMID: 29944761 PMCID: PMC6128411 DOI: 10.1111/imr.12668] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Over the last several decades, novel populations of unconventional T cells have been identified; defined by an invariant (or nearly invariant) T cell receptor (TCR) with a fixed specificity to non-canonical antigens and major histocompatibility (MHC) molecules, they form large, functionally monoclonal populations tasked with surveying for their specific antigens. With residence in both lymphoid and non-lymphoid tissues coupled with their ability to rapidly produce a spectrum of cytokines and effector molecules, the unconventional T cells are poised as some of the first responders to infection/damage and are thought to provide critical coverage before more focused, conventional T cell responses are mobilized. However, new technologies for the measurement and characterization of TCR repertoires have identified an underappreciated amount of TCR diversity in the unconventional T cells. In many cases, the specificities of these diverse TCRs converge on the same or similar antigens as their invariant counterparts, while others have yet to be defined. Here, we will review the current knowledge of the TCR repertoires of unconventional T cells and discuss how repertoires might be used as a framework for their organization, and further our understanding of their role not only during an immune response, but also their contribution in maintaining homeostasis.
Collapse
Affiliation(s)
| | - Paul G Thomas
- St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
35
|
Wirasinha RC, Singh M, Archer SK, Chan A, Harrison PF, Goodnow CC, Daley SR. αβ T-cell receptors with a central CDR3 cysteine are enriched in CD8αα intraepithelial lymphocytes and their thymic precursors. Immunol Cell Biol 2018; 96:553-561. [PMID: 29726044 DOI: 10.1111/imcb.12047] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 03/09/2018] [Accepted: 03/23/2018] [Indexed: 01/04/2023]
Abstract
The thymus plays a crucial role in immune tolerance by exposing developing T cells (thymocytes) to a myriad of self-antigens. Strong T-cell receptor (TCR) engagement induces tolerance in self-reactive thymocytes by stimulating apoptosis or selection into specialized T-cell lineages, including intestinal TCRαβ+ CD8αα+ intraepithelial lymphocytes (IEL). TCR-intrinsic amino acid motifs that can be used to predict whether a TCR will be strongly self-reactive remain elusive. Here, a novel TCR sequence alignment approach revealed that T-cell lineages in C57BL/6 mice had divergent usage of cysteine within two positions of the amino acid at the apex of the complementarity-determining region 3 (CDR3) of the TCRα or TCRβ chain. Compared to pre-selection thymocytes, central CDR3 cysteine usage was increased in IEL and Type A IEL precursors (IELp) and markedly decreased in Foxp3+ regulatory T cells (T-reg) and naïve T cells. These findings reveal a TCR-intrinsic motif that distinguishes Type A IELp and IEL from T-reg and naïve T cells.
Collapse
Affiliation(s)
- Rushika C Wirasinha
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Mandeep Singh
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia
| | - Stuart K Archer
- Monash Bioinformatics Platform, Monash University, Melbourne, VIC, 3800, Australia
| | - Anna Chan
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| | - Paul F Harrison
- Monash Bioinformatics Platform, Monash University, Melbourne, VIC, 3800, Australia
| | - Christopher C Goodnow
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, 2010, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Stephen R Daley
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, 3800, Australia
| |
Collapse
|
36
|
Kasler HG, Lee IS, Lim HW, Verdin E. Histone Deacetylase 7 mediates tissue-specific autoimmunity via control of innate effector function in invariant Natural Killer T Cells. eLife 2018; 7:e32109. [PMID: 29664401 PMCID: PMC5943034 DOI: 10.7554/elife.32109] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 04/05/2018] [Indexed: 12/13/2022] Open
Abstract
We report that Histone Deacetylase 7 (HDAC7) controls the thymic effector programming of Natural Killer T (NKT) cells, and that interference with this function contributes to tissue-specific autoimmunity. Gain of HDAC7 function in thymocytes blocks both negative selection and NKT development, and diverts Vα14/Jα18 TCR transgenic thymocytes into a Tconv-like lineage. Conversely, HDAC7 deletion promotes thymocyte apoptosis and causes expansion of innate-effector cells. Investigating the mechanisms involved, we found that HDAC7 binds PLZF and modulates PLZF-dependent transcription. Moreover, HDAC7 and many of its transcriptional targets are human risk loci for IBD and PSC, autoimmune diseases that strikingly resemble the disease we observe in HDAC7 gain-of-function in mice. Importantly, reconstitution of iNKT cells in these mice mitigated their disease, suggesting that the combined defects in negative selection and iNKT cells due to altered HDAC7 function can cause tissue-restricted autoimmunity, a finding that may explain the association between HDAC7 and hepatobiliary autoimmunity.
Collapse
Affiliation(s)
- Herbert G Kasler
- Gladstone Institute of Virology and ImmunologySan FranciscoUnited States
- Department of MedicineUniversity of California, San FranciscoSan FranciscoUnited States
- Buck Institute for Research on AgingNovatoUnited States
| | - Intelly S Lee
- Gladstone Institute of Virology and ImmunologySan FranciscoUnited States
- Department of MedicineUniversity of California, San FranciscoSan FranciscoUnited States
| | - Hyung W Lim
- Gladstone Institute of Virology and ImmunologySan FranciscoUnited States
- Department of MedicineUniversity of California, San FranciscoSan FranciscoUnited States
| | - Eric Verdin
- Gladstone Institute of Virology and ImmunologySan FranciscoUnited States
- Department of MedicineUniversity of California, San FranciscoSan FranciscoUnited States
- Buck Institute for Research on AgingNovatoUnited States
| |
Collapse
|
37
|
Olivares-Villagómez D, Van Kaer L. Intestinal Intraepithelial Lymphocytes: Sentinels of the Mucosal Barrier. Trends Immunol 2018; 39:264-275. [PMID: 29221933 PMCID: PMC8056148 DOI: 10.1016/j.it.2017.11.003] [Citation(s) in RCA: 180] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/14/2017] [Accepted: 11/15/2017] [Indexed: 02/06/2023]
Abstract
Intestinal intraepithelial lymphocytes (IELs) are a large and diverse population of lymphoid cells that reside between the intestinal epithelial cells (IECs) that form the intestinal mucosal barrier. Although IEL biology has traditionally focused on T cells, recent studies have identified several subsets of T cell receptor (TCR)-negative IELs with intriguing properties. New insight into the development, homeostasis, and functions of distinct IEL subsets has recently been provided. Additional studies have revealed intricate interactions between different IEL subsets, reciprocal interactions between IELs and IECs, and communication of IELs with immune cells that reside outside the intestinal epithelium. We review here sentinel functions of IELs in the maintenance of the mucosal barrier integrity, as well as how dysregulated IEL responses can contribute to pathology.
Collapse
Affiliation(s)
- Danyvid Olivares-Villagómez
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| | - Luc Van Kaer
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN, USA.
| |
Collapse
|
38
|
A committed postselection precursor to natural TCRαβ + intraepithelial lymphocytes. Mucosal Immunol 2018; 11:333-344. [PMID: 28745324 DOI: 10.1038/mi.2017.54] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2016] [Accepted: 05/08/2017] [Indexed: 02/04/2023]
Abstract
The intestine is a major immune organ with several specialized lymphoid structures and immune cells. Among these are thymus-derived natural intraepithelial lymphocytes (IELs) that lack expression of the classical co-receptors CD4 or CD8αβ (double negative (DN)). Natural IELs are both αβ+ and γδ+ T cells that play important roles in the maintenance of the epithelial barrier at steady state and during inflammation. The transcription factor T-bet is essential for the peripheral development of natural IELs, but its role during thymic development has remained less clear. Here we show that a T-bet gradient in DN TCRαβ+NK1.1- thymocytes (IEL precursors (IELPs)) determines IEL fate in natural TCRαβ+ IELs. Employing T-bet ZsGreen reporter mice in in vitro cultures and in vivo transfer experiments, we demonstrate that with increasing expression of T-bet, DN TCRαβ+NK1.1- thymocytes are gradually restricted to a DN IEL fate. Furthermore, we show that the natural TCRαβ+ IELs seed the intestine within the first month of life. This in turn is preceded by the appearance of T-bet- and T-bet+ IELPs that egress from the thymus in a sphingosine-1-phosphate (S1P)-dependent manner. In summary, the use of T-bet reporter mice has enabled us to identify and refine an immediate and clearly committed postselection precursor of natural TCRαβ+ IELs.
Collapse
|
39
|
Brandt D, Hedrich CM. TCRαβ +CD3 +CD4 -CD8 - (double negative) T cells in autoimmunity. Autoimmun Rev 2018; 17:422-430. [PMID: 29428806 DOI: 10.1016/j.autrev.2018.02.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Accepted: 12/07/2017] [Indexed: 12/14/2022]
Abstract
TCRαβ+CD3+CD4-CD8- "double negative" (DN) T cells comprise a small subset of mature peripheral T cells. The origin and function of DN T cells are somewhat unclear and discussed controversially. While DN T cells resemble a rare and heterogeneous T cell subpopulation in healthy individuals, numbers of TCRαβ+ DN T cells are expanded in several inflammatory conditions, where they also exhibit distinct effector phenotypes and infiltrate inflamed tissues. Thus, DN T cells may be involved in systemic inflammation and tissue damage in autoimmune/inflammatory conditions, including SLE, Sjögren's syndrome, and psoriasis. Here, the current understanding of the origin and phenotype of DN T cells, and their role in the instruction of immune responses, autoimmunity and inflammation will be discussed in health and disease.
Collapse
Affiliation(s)
- D Brandt
- Division of Pediatric Rheumatology and Immunology, Children's Hospital Dresden, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - C M Hedrich
- Division of Pediatric Rheumatology and Immunology, Children's Hospital Dresden, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany; Department of Women's & Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK; Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust Hospital, Liverpool, UK.
| |
Collapse
|
40
|
Abstract
Thymocyte selection involves the positive and negative selection of the repertoire of T cell receptors (TCRs) such that the organism does not suffer autoimmunity, yet has the benefit of the ability to recognize any invading pathogen. The signal transduced through the TCR is translated into a number of different signaling cascades that result in transcription factor activity in the nucleus and changes to the cytoskeleton and motility. Negative selection involves inducing apoptosis in thymocytes that express strongly self-reactive TCRs, whereas positive selection must induce survival and differentiation programs in cells that are more weakly self-reactive. The TCR recognition event is analog by nature, but the outcome of signaling is not. A large number of molecules regulate the strength of the TCR-derived signal at various points in the cascades. This review discusses the various factors that can regulate the strength of the TCR signal during thymocyte development.
Collapse
Affiliation(s)
- Nicholas R J Gascoigne
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, and Immunology Program, National University of Singapore, Singapore 11759;
| | - Vasily Rybakin
- Laboratory of Immunobiology, REGA Institute, Department of Microbiology and Immunology, KU Leuven, Leuven 3000, Belgium
| | - Oreste Acuto
- Sir William Dunn School of Pathology, University of Oxford, Oxford OX1 3RE, United Kingdom
| | - Joanna Brzostek
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, and Immunology Program, National University of Singapore, Singapore 11759;
| |
Collapse
|
41
|
Generating CD8αα IELs from two sources of thymic precursors. Cell Mol Immunol 2017; 15:640-641. [PMID: 28890546 DOI: 10.1038/cmi.2017.93] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 08/01/2017] [Indexed: 12/25/2022] Open
|
42
|
Increased TCR signal strength in DN thymocytes promotes development of gut TCRαβ (+)CD8αα (+) intraepithelial lymphocytes. Sci Rep 2017; 7:10659. [PMID: 28878277 PMCID: PMC5587556 DOI: 10.1038/s41598-017-09368-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Accepted: 07/24/2017] [Indexed: 01/15/2023] Open
Abstract
CD4(+)CD8(+) “double positive” (DP) thymocytes differentiate into diverse αβ T cell sub-types using mechanistically distinct programs. For example, conventional αβ T cells develop from DP cells after partial-agonist T cell receptor (TCR) interactions with self-peptide/MHC, whereas unconventional αβ T cells, such as TCRαβ(+)CD8αα(+) intraepithelial lymphocytes (IELs), require full-agonist TCR interactions. Despite this, DP cells appear homogeneous, and it remains unclear how distinct TCR signalling instructs distinct developmental outcomes. Moreover, whether TCR signals at earlier stages of development, for example in CD4(−)CD8(−) double negative (DN) cells, impact on later fate decisions is presently unknown. Here, we assess four strains of mice that display altered TCR signal strength in DN cells, which correlates with altered generation of unconventional TCRαβ(+)CD8αα(+) IELs. FVB/n mice (compared to C57BL/6 animals) and mice with altered preTCRα (pTα) expression, both displayed weaker TCR signalling in DN cells, an inefficient DN-to-DP transition, and reduced contribution of TCRαβ(+)CD8αα(+) IELs to gut epithelium. Conversely, TCRαβ(+)CD8αα(+) IEL development was favoured in mice with increased TCR signal strength in DN cells. Collectively, these data suggest TCR signal strength in DN cells directly impacts on subsequent DP cell differentiation, fundamentally altering the potential of thymocyte progenitors to adopt conventional versus unconventional T cell fates.
Collapse
|
43
|
Ruscher R, Kummer RL, Lee YJ, Jameson SC, Hogquist KA. CD8αα intraepithelial lymphocytes arise from two main thymic precursors. Nat Immunol 2017; 18:771-779. [PMID: 28530714 PMCID: PMC5505317 DOI: 10.1038/ni.3751] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Accepted: 04/24/2017] [Indexed: 12/15/2022]
Abstract
TCRαβ+CD4-CD8α+CD8β- intestinal intraepithelial lymphocytes (CD8αα IELs) are an abundant population of thymus-derived T cells that protect the gut barrier surface. We sought to better define the thymic IEL precursor (IELp) through analysis of its maturation, localization and emigration. We defined two precursor populations among TCRβ+CD4-CD8- thymocytes by dependence on the kinase TAK1 and rigorous lineage-exclusion criteria. Those IELp populations included a nascent PD-1+ population and a T-bet+ population that accumulated with age. Both gave rise to intestinal CD8αα IELs after adoptive transfer. The PD-1+ IELp population included more strongly self-reactive clones and was largely restricted by classical major histocompatibility complex (MHC) molecules. Those cells localized to the cortex and efficiently emigrated in a manner dependent on the receptor S1PR1. The T-bet+ IELp population localized to the medulla, included cells restricted by non-classical MHC molecules and expressed the receptor NK1.1, the integrin CD103 and the chemokine receptor CXCR3. The two IELp populations further differed in their use of the T cell antigen receptor (TCR) α-chain variable region (Vα) and β-chain variable region (Vβ). These data provide a foundation for understanding the biology of CD8αα IELs.
Collapse
Affiliation(s)
- Roland Ruscher
- The Department of Laboratory Medicine and Pathology and Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Rebecca L Kummer
- The Department of Laboratory Medicine and Pathology and Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - You Jeong Lee
- Academy of Immunology and Microbiology, Institute for Basic Science, and Division of Integrative Biosciences and Biotechnology, Pohang University of Science and Technology, Pohang, Republic of Korea
| | - Stephen C Jameson
- The Department of Laboratory Medicine and Pathology and Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kristin A Hogquist
- The Department of Laboratory Medicine and Pathology and Center for Immunology, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
44
|
Daley SR, Teh C, Hu DY, Strasser A, Gray DH. Cell death and thymic tolerance. Immunol Rev 2017; 277:9-20. [DOI: 10.1111/imr.12532] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Stephen R. Daley
- Infection and Immunity Program; Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology; Monash University; Melbourne VIC Australia
| | - Charis Teh
- The Walter and Eliza Hall Institute of Medical Research; Melbourne VIC Australia
- Department of Medical Biology; The University of Melbourne; Parkville VIC Australia
| | | | - Andreas Strasser
- The Walter and Eliza Hall Institute of Medical Research; Melbourne VIC Australia
- Department of Medical Biology; The University of Melbourne; Parkville VIC Australia
| | - Daniel H.D. Gray
- The Walter and Eliza Hall Institute of Medical Research; Melbourne VIC Australia
- Department of Medical Biology; The University of Melbourne; Parkville VIC Australia
| |
Collapse
|
45
|
Faria AMC, Reis BS, Mucida D. Tissue adaptation: Implications for gut immunity and tolerance. J Exp Med 2017; 214:1211-1226. [PMID: 28432200 PMCID: PMC5413340 DOI: 10.1084/jem.20162014] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/31/2017] [Accepted: 04/04/2017] [Indexed: 12/22/2022] Open
Abstract
Faria et al. discuss the concept that immune cells undergo specialized adaptation to tissue-specific conditions and its potential implications for tolerance and immunity. Tissue adaptation is an intrinsic component of immune cell development, influencing both resistance to pathogens and tolerance. Chronically stimulated surfaces of the body, in particular the gut mucosa, are the major sites where immune cells traffic and reside. Their adaptation to these environments requires constant discrimination between natural stimulation coming from harmless microbiota and food, and pathogens that need to be cleared. This review will focus on the adaptation of lymphocytes to the gut mucosa, a highly specialized environment that can help us understand the plasticity of leukocytes arriving at various tissue sites and how tissue-related factors operate to shape immune cell fate and function.
Collapse
Affiliation(s)
- Ana M C Faria
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065 .,Departamento de Bioquímica e Imunologia, Universidade Federal de Minas Gerais, Belo Horizonte, MG 31270901, Brazil
| | - Bernardo S Reis
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065
| | - Daniel Mucida
- Laboratory of Mucosal Immunology, The Rockefeller University, New York, NY 10065
| |
Collapse
|
46
|
Verstichel G, Vermijlen D, Martens L, Goetgeluk G, Brouwer M, Thiault N, Van Caeneghem Y, De Munter S, Weening K, Bonte S, Leclercq G, Taghon T, Kerre T, Saeys Y, Van Dorpe J, Cheroutre H, Vandekerckhove B. The checkpoint for agonist selection precedes conventional selection in human thymus. Sci Immunol 2017; 2:2/8/eaah4232. [PMID: 28783686 DOI: 10.1126/sciimmunol.aah4232] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 11/07/2016] [Accepted: 01/11/2017] [Indexed: 11/02/2022]
Abstract
The thymus plays a central role in self-tolerance, partly by eliminating precursors with a T cell receptor (TCR) that binds strongly to self-antigens. However, the generation of self-agonist-selected lineages also relies on strong TCR signaling. How thymocytes discriminate between these opposite outcomes remains elusive. Here, we identified a human agonist-selected PD-1+ CD8αα+ subset of mature CD8αβ+ T cells that displays an effector phenotype associated with agonist selection. TCR stimulation of immature post-β-selection thymocyte blasts specifically gives rise to this innate subset and fixes early T cell receptor alpha variable (TRAV) and T cell receptor alpha joining (TRAJ) rearrangements in the TCR repertoire. These findings suggest that the checkpoint for agonist selection precedes conventional selection in the human thymus.
Collapse
Affiliation(s)
- Greet Verstichel
- Faculty of Medicine and Health Sciences, Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, University Hospital Ghent, MRB2, De Pintelaan 185, 9000 Ghent, Belgium
| | - David Vermijlen
- Department of Biopharmacy, Université Libre de Bruxelles (ULB), Boulevard du Triomphe, accès 2, 1050 Brussels, Belgium.,Institute for Medical Immunology, ULB, Rue Adrienne Bolland 8, 6041 Gosselies, Belgium
| | - Liesbet Martens
- Data Mining and Modeling for Systems Immunology, Vlaams Instituut voor Biotechnologie Inflammation Research Center, Technologiepark 927, 9052 Ghent, Belgium
| | - Glenn Goetgeluk
- Faculty of Medicine and Health Sciences, Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, University Hospital Ghent, MRB2, De Pintelaan 185, 9000 Ghent, Belgium
| | - Margreet Brouwer
- Institute for Medical Immunology, ULB, Rue Adrienne Bolland 8, 6041 Gosselies, Belgium
| | - Nicolas Thiault
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Yasmine Van Caeneghem
- Faculty of Medicine and Health Sciences, Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, University Hospital Ghent, MRB2, De Pintelaan 185, 9000 Ghent, Belgium
| | - Stijn De Munter
- Faculty of Medicine and Health Sciences, Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, University Hospital Ghent, MRB2, De Pintelaan 185, 9000 Ghent, Belgium
| | - Karin Weening
- Faculty of Medicine and Health Sciences, Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, University Hospital Ghent, MRB2, De Pintelaan 185, 9000 Ghent, Belgium
| | - Sarah Bonte
- Faculty of Medicine and Health Sciences, Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, University Hospital Ghent, MRB2, De Pintelaan 185, 9000 Ghent, Belgium
| | - Georges Leclercq
- Faculty of Medicine and Health Sciences, Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, University Hospital Ghent, MRB2, De Pintelaan 185, 9000 Ghent, Belgium
| | - Tom Taghon
- Faculty of Medicine and Health Sciences, Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, University Hospital Ghent, MRB2, De Pintelaan 185, 9000 Ghent, Belgium
| | - Tessa Kerre
- Faculty of Medicine and Health Sciences, Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, University Hospital Ghent, MRB2, De Pintelaan 185, 9000 Ghent, Belgium
| | - Yvan Saeys
- Data Mining and Modeling for Systems Immunology, Vlaams Instituut voor Biotechnologie Inflammation Research Center, Technologiepark 927, 9052 Ghent, Belgium.,Department of Internal Medicine, Ghent University, De Pintelaan 185, 9000 Ghent, Belgium
| | - Jo Van Dorpe
- Faculty of Medicine and Health Sciences, Department of Medical and Forensic Pathology, Ghent University, University Hospital Ghent, De Pintelaan 185, 9000 Ghent, Belgium
| | - Hilde Cheroutre
- Division of Developmental Immunology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Bart Vandekerckhove
- Faculty of Medicine and Health Sciences, Department of Clinical Chemistry, Microbiology and Immunology, Ghent University, University Hospital Ghent, MRB2, De Pintelaan 185, 9000 Ghent, Belgium.
| |
Collapse
|
47
|
The composition of T cell subtypes in duodenal biopsies are altered in coeliac disease patients. PLoS One 2017; 12:e0170270. [PMID: 28166225 PMCID: PMC5293270 DOI: 10.1371/journal.pone.0170270] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 01/02/2017] [Indexed: 12/11/2022] Open
Abstract
One of the hallmarks of Celiac disease (CD) is intraepithelial lymphocytosis in the small intestine. Until now, investigations to characterize the T cell subpopulations within the epithelial layer have not discriminated between the heterodimeric co-receptor molecule, CD8αβ, and the possibly immunoregulatory CD8αα homodimer molecule. Besides TCRαβ+ CD4+ cells, no other phenotypes have been shown to be gluten-reactive. Using flow cytometry on lymphocytes from duodenal biopsies, we determined that the number of B cells (CD3- CD19+) and the number of CD3+ CD4- CD8- double-negative (DN) T cells were elevated 6–7 fold in children with CD. We next isolated and quantified intraepithelial lymphocytes (IELs) from biopsies obtained from patients (both children and adults) with CD, potential CD and non-CD controls. Flow cytometric analysis of the duodenal T cell subpopulations was performed including the markers TCRαβ, TCRγδ, CD4, CD8α and CD8β. Proportions of γδ T cells and CD8αβ+ cells among IELs were increased in CD patients, whereas proportions of CD4+ CD8αα+ and CD4+ single-positive T cells were decreased. Additionally, two gluten-reactive T cell lines (TCLs) derived from CD biopsies were analyzed for changes in proportions of T cell subsets before and after gluten stimulation. In a proliferation assay, dividing cells were tracked with carboxyfluorescein succinimidyl ester (CFSE), and both αβ and γδ T cells proliferated in response to gluten. Changes in duodenal T cell subpopulations in potential CD patients followed the same pattern as for CD patients, but with less pronounced effect.
Collapse
|
48
|
Abstract
The ability of T cells to respond to a wide array of foreign antigens while avoiding reactivity to self is largely determined by cellular selection of developing T cells in the thymus. While a great deal is known about the cell types and molecules involved in T-cell selection in the thymus, our understanding of the spatial and temporal aspects of this process remain relatively poorly understood. Thymocytes are highly motile within the thymus and travel between specialized microenvironments at different phases of their development while interacting with distinct sets of self-peptides and peptide presenting cells. A knowledge of when, where, and how thymocytes encounter self-peptide MHC ligands at different stages of thymic development is key to understanding T-cell selection. In the past several years, our laboratory has investigated this topic using two-photon time-lapse microscopy to directly visualize thymocyte migration and signaling events, together with a living thymic slice preparation to provide a synchronized experimental model of T-cell selection in situ. Here, we discuss recent advances in our understanding of the temporal and spatial aspects of T-cell selection, highlighting our own work, and placing them in the context of work from other groups.
Collapse
Affiliation(s)
- Nadia Kurd
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| | - Ellen A Robey
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, 94720, USA
| |
Collapse
|
49
|
Li KP, Fähnrich A, Roy E, Cuda CM, Grimes HL, Perlman HR, Kalies K, Hildeman DA. Temporal Expression of Bim Limits the Development of Agonist-Selected Thymocytes and Skews Their TCRβ Repertoire. THE JOURNAL OF IMMUNOLOGY 2016; 198:257-269. [PMID: 27852740 DOI: 10.4049/jimmunol.1601200] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Accepted: 10/25/2016] [Indexed: 12/11/2022]
Abstract
CD8αα TCRαβ+ intestinal intraepithelial lymphocytes play a critical role in promoting intestinal homeostasis, although mechanisms controlling their development and peripheral homeostasis remain unclear. In this study, we examined the spatiotemporal role of Bim in the thymic selection of CD8αα precursors and the fate of these cells in the periphery. We found that T cell-specific expression of Bim during early/cortical, but not late/medullary, thymic development controls the agonist selection of CD8αα precursors and limits their private TCRβ repertoire. During this process, agonist-selected double-positive cells lose CD4/8 coreceptor expression and masquerade as double-negative (DN) TCRαβhi thymocytes. Although these DN thymocytes fail to re-express coreceptors after OP9-DL1 culture, they eventually mature and accumulate in the spleen where TCR and IL-15/STAT5 signaling promotes their conversion to CD8αα cells and their expression of gut-homing receptors. Adoptive transfer of splenic DN cells gives rise to CD8αα cells in the gut, establishing their precursor relationship in vivo. Interestingly, Bim does not restrict the IL-15-driven maturation of CD8αα cells that is critical for intestinal homeostasis. Thus, we found a temporal and tissue-specific role for Bim in limiting thymic agonist selection of CD8αα precursors and their TCRβ repertoire, but not in the maintenance of CD8αα intraepithelial lymphocytes in the intestine.
Collapse
Affiliation(s)
- Kun-Po Li
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229.,Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45257
| | - Anke Fähnrich
- Institute for Anatomy, University of Lübeck, 23538 Lübeck, Germany; and
| | - Eron Roy
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229.,Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45257
| | - Carla M Cuda
- Rheumatology Division, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - H Leighton Grimes
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229.,Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45257
| | - Harris R Perlman
- Rheumatology Division, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Kathrin Kalies
- Institute for Anatomy, University of Lübeck, 23538 Lübeck, Germany; and
| | - David A Hildeman
- Division of Immunobiology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229; .,Department of Pediatrics, University of Cincinnati, Cincinnati, OH 45257
| |
Collapse
|
50
|
Ettersperger J, Montcuquet N, Malamut G, Guegan N, Lopez-Lastra S, Gayraud S, Reimann C, Vidal E, Cagnard N, Villarese P, Andre-Schmutz I, Gomes Domingues R, Godinho-Silva C, Veiga-Fernandes H, Lhermitte L, Asnafi V, Macintyre E, Cellier C, Beldjord K, Di Santo JP, Cerf-Bensussan N, Meresse B. Interleukin-15-Dependent T-Cell-like Innate Intraepithelial Lymphocytes Develop in the Intestine and Transform into Lymphomas in Celiac Disease. Immunity 2016; 45:610-625. [PMID: 27612641 DOI: 10.1016/j.immuni.2016.07.018] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 05/12/2016] [Accepted: 06/02/2016] [Indexed: 01/19/2023]
Abstract
The nature of gut intraepithelial lymphocytes (IELs) lacking antigen receptors remains controversial. Herein we showed that, in humans and in mice, innate intestinal IELs expressing intracellular CD3 (iCD3(+)) differentiate along an Id2 transcription factor (TF)-independent pathway in response to TF NOTCH1, interleukin-15 (IL-15), and Granzyme B signals. In NOTCH1-activated human hematopoietic precursors, IL-15 induced Granzyme B, which cleaved NOTCH1 into a peptide lacking transcriptional activity. As a result, NOTCH1 target genes indispensable for T cell differentiation were silenced and precursors were reprogrammed into innate cells with T cell marks including intracellular CD3 and T cell rearrangements. In the intraepithelial lymphoma complicating celiac disease, iCD3(+) innate IELs acquired gain-of-function mutations in Janus kinase 1 or Signal transducer and activator of transcription 3, which enhanced their response to IL-15. Overall we characterized gut T cell-like innate IELs, deciphered their pathway of differentiation and showed their malignant transformation in celiac disease.
Collapse
Affiliation(s)
- Julien Ettersperger
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Nicolas Montcuquet
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Georgia Malamut
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; AP-HP, Department of Gastroenterology, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Nicolas Guegan
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Silvia Lopez-Lastra
- Innate Immunity Unit, Institut Pasteur, 75015 Paris, France; INSERM U 668, 75015 Paris, France
| | - Ségolène Gayraud
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France
| | - Christian Reimann
- Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, 75015 Paris, France
| | - Elodie Vidal
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | | | - Patrick Villarese
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Isabelle Andre-Schmutz
- Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; INSERM UMR1163, Laboratory of Human Lymphohematopoiesis, 75015 Paris, France
| | - Rita Gomes Domingues
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, 1649-028 Lisboa, Portugal
| | - Cristina Godinho-Silva
- Instituto de Medicina Molecular, Faculdade de Medicina de Lisboa, 1649-028 Lisboa, Portugal
| | | | - Ludovic Lhermitte
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Vahid Asnafi
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Elizabeth Macintyre
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France
| | - Christophe Cellier
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France; AP-HP, Department of Gastroenterology, Hôpital Européen Georges Pompidou, 75015 Paris, France
| | - Kheira Beldjord
- Université Paris Descartes-Sorbonne Paris Cité, Institut Necker-Enfants-Malades, INSERM UMR1151 and, Biological Hematology, AP-HP Necker-Enfants-Malades, 75015 Paris, France; Institut Universitaire d'Hématologie, Hôpital Saint-Louis, 75010 Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, 75015 Paris, France; INSERM U 668, 75015 Paris, France
| | - Nadine Cerf-Bensussan
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France.
| | - Bertrand Meresse
- INSERM UMR1163, Laboratory of Intestinal Immunity, Institut Imagine, 75015 Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France.
| |
Collapse
|