1
|
Yared N, Papadopoulou M, Barennes P, Pham HP, Quiniou V, Netzer S, Kaminski H, Burguet L, Demeste A, Colas P, Mora-Charrot L, Rousseau B, Izotte J, Zouine A, Gauthereau X, Vermijlen D, Déchanet-Merville J, Capone M. Long-lived central memory γδ T cells confer protection against murine cytomegalovirus reinfection. PLoS Pathog 2024; 20:e1010785. [PMID: 38976755 PMCID: PMC11257398 DOI: 10.1371/journal.ppat.1010785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 07/18/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
The involvement of γδ TCR-bearing lymphocytes in immunological memory has gained increasing interest due to their functional duality between adaptive and innate immunity. γδ T effector memory (TEM) and central memory (TCM) subsets have been identified, but their respective roles in memory responses are poorly understood. In the present study, we used subsequent mouse cytomegalovirus (MCMV) infections of αβ T cell deficient mice in order to analyze the memory potential of γδ T cells. As for CMV-specific αβ T cells, MCMV induced the accumulation of cytolytic, KLRG1+CX3CR1+ γδ TEM that principally localized in infected organ vasculature. Typifying T cell memory, γδ T cell expansion in organs and blood was higher after secondary viral challenge than after primary infection. Viral control upon MCMV reinfection was prevented when masking γδ T-cell receptor, and was associated with a preferential amplification of private and unfocused TCR δ chain repertoire composed of a combination of clonotypes expanded post-primary infection and, more unexpectedly, of novel expanded clonotypes. Finally, long-term-primed γδ TCM cells, but not γδ TEM cells, protected T cell-deficient hosts against MCMV-induced death upon adoptive transfer, probably through their ability to survive and to generate TEM in the recipient host. This better survival potential of TCM cells was confirmed by a detailed scRNASeq analysis of the two γδ T cell memory subsets which also revealed their similarity to classically adaptive αβ CD8 T cells. Overall, our study uncovered memory properties of long-lived TCM γδ T cells that confer protection in a chronic infection, highlighting the interest of this T cell subset in vaccination approaches.
Collapse
Affiliation(s)
- Nathalie Yared
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Maria Papadopoulou
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | | | | | - Sonia Netzer
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Hanna Kaminski
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Laure Burguet
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Amandine Demeste
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Pacôme Colas
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Lea Mora-Charrot
- Bordeaux University, Service Commun des Animaleries, Bordeaux, France
| | - Benoit Rousseau
- Bordeaux University, Service Commun des Animaleries, Bordeaux, France
| | - Julien Izotte
- Bordeaux University, Service Commun des Animaleries, Bordeaux, France
| | - Atika Zouine
- Bordeaux University, Centre National de la Recherche Scientifique, Institut national de la santé et de la recherche médicale, FACSility, TBM Core, Bordeaux, France
| | - Xavier Gauthereau
- Bordeaux University, Centre National de la Recherche Scientifique, Institut national de la santé et de la recherche médicale, OneCell, RT-PCR and Single Cell Libraries, TBM Core, Bordeaux, France
| | - David Vermijlen
- Department of Pharmacotherapy and Pharmaceutics, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Gosselies, Belgium
- Université Libre de Bruxelles Center for Research in Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO department, Walloon ExceLlence Research Institute, Wavre, Belgium
| | - Julie Déchanet-Merville
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| | - Myriam Capone
- Bordeaux University, Centre National de la Recherche Scientifique, Institut National de la Santé et de la Recherche Médicale, ImmunoConcEpt, UMR 5164, ERL 1303, ImmunoConcEpt, Bordeaux, France
| |
Collapse
|
2
|
Sumaria N, Fiala GJ, Inácio D, Curado-Avelar M, Cachucho A, Pinheiro R, Wiesheu R, Kimura S, Courtois L, Blankenhaus B, Darrigues J, Suske T, Almeida ARM, Minguet S, Asnafi V, Lhermitte L, Mullighan CG, Coffelt SB, Moriggl R, Barata JT, Pennington DJ, Silva-Santos B. Perinatal thymic-derived CD8αβ-expressing γδ T cells are innate IFN-γ producers that expand in IL-7R-STAT5B-driven neoplasms. Nat Immunol 2024; 25:1207-1217. [PMID: 38802512 PMCID: PMC11224017 DOI: 10.1038/s41590-024-01855-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/25/2024] [Indexed: 05/29/2024]
Abstract
The contribution of γδ T cells to immune responses is associated with rapid secretion of interferon-γ (IFN-γ). Here, we show a perinatal thymic wave of innate IFN-γ-producing γδ T cells that express CD8αβ heterodimers and expand in preclinical models of infection and cancer. Optimal CD8αβ+ γδ T cell development is directed by low T cell receptor signaling and through provision of interleukin (IL)-4 and IL-7. This population is pathologically relevant as overactive, or constitutive, IL-7R-STAT5B signaling promotes a supraphysiological accumulation of CD8αβ+ γδ T cells in the thymus and peripheral lymphoid organs in two mouse models of T cell neoplasia. Likewise, CD8αβ+ γδ T cells define a distinct subset of human T cell acute lymphoblastic leukemia pediatric patients. This work characterizes the normal and malignant development of CD8αβ+ γδ T cells that are enriched in early life and contribute to innate IFN-γ responses to infection and cancer.
Collapse
MESH Headings
- Animals
- Interferon-gamma/metabolism
- Interferon-gamma/immunology
- Mice
- Humans
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Thymus Gland/immunology
- Receptors, Interleukin-7/metabolism
- Immunity, Innate
- STAT5 Transcription Factor/metabolism
- Signal Transduction/immunology
- Mice, Inbred C57BL
- CD8-Positive T-Lymphocytes/immunology
- Mice, Knockout
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- CD8 Antigens/metabolism
- Female
- Intraepithelial Lymphocytes/immunology
- Intraepithelial Lymphocytes/metabolism
- Interleukin-7/metabolism
Collapse
Affiliation(s)
- Nital Sumaria
- Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK
| | - Gina J Fiala
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal.
- Faculty of Biology, University of Freiburg, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| | - Daniel Inácio
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Marta Curado-Avelar
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Ana Cachucho
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Rúben Pinheiro
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Robert Wiesheu
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Lucien Courtois
- Hôpital Necker Enfants-Malades, Université de Paris, Paris, France
| | - Birte Blankenhaus
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Julie Darrigues
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Tobias Suske
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - Afonso R M Almeida
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Susana Minguet
- Faculty of Biology, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
- Center of Chronic Immunodeficiency CCI, University Clinics and Medical Faculty, Freiburg, Germany
| | - Vahid Asnafi
- Hôpital Necker Enfants-Malades, Université de Paris, Paris, France
| | | | | | - Seth B Coffelt
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Richard Moriggl
- Department of Biosciences and Medical Biology, Paris Lodron University of Salzburg, Salzburg, Austria
| | - João T Barata
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| | - Daniel J Pennington
- Blizard Institute, Barts and The London School of Medicine, Queen Mary University of London, London, UK.
| | - Bruno Silva-Santos
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
| |
Collapse
|
3
|
Yu H, Yang W, Cao M, Lei Q, Yuan R, Xu H, Cui Y, Chen X, Su X, Zhuo H, Lin L. Mechanism study of ubiquitination in T cell development and autoimmune disease. Front Immunol 2024; 15:1359933. [PMID: 38562929 PMCID: PMC10982411 DOI: 10.3389/fimmu.2024.1359933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
T cells play critical role in multiple immune processes including antigen response, tumor immunity, inflammation, self-tolerance maintenance and autoimmune diseases et. Fetal liver or bone marrow-derived thymus-seeding progenitors (TSPs) settle in thymus and undergo T cell-lineage commitment, proliferation, T cell receptor (TCR) rearrangement, and thymic selections driven by microenvironment composed of thymic epithelial cells (TEC), dendritic cells (DC), macrophage and B cells, thus generating T cells with diverse TCR repertoire immunocompetent but not self-reactive. Additionally, some self-reactive thymocytes give rise to Treg with the help of TEC and DC, serving for immune tolerance. The sequential proliferation, cell fate decision, and selection during T cell development and self-tolerance establishment are tightly regulated to ensure the proper immune response without autoimmune reaction. There are remarkable progresses in understanding of the regulatory mechanisms regarding ubiquitination in T cell development and the establishment of self-tolerance in the past few years, which holds great potential for further therapeutic interventions in immune-related diseases.
Collapse
Affiliation(s)
- Hui Yu
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Wenyong Yang
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Min Cao
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Qingqiang Lei
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Renbin Yuan
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - He Xu
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Yuqian Cui
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Xuerui Chen
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Xu Su
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
- College of Medicine, Southwest Jiaotong University, Chengdu, China
| | - Hui Zhuo
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| | - Liangbin Lin
- Department of Urology, Medical Research Center, Department of Neurosurgery, The Third People’s Hospital of Chengdu, The Affiliated Hospital of Southwest Jiaotong University, The Second Chengdu Hospital Affiliated to Chongqing Medical University, Chengdu, China
| |
Collapse
|
4
|
Bosselut R. A Beginner's Guide to T Cell Development. Methods Mol Biol 2023; 2580:3-24. [PMID: 36374448 DOI: 10.1007/978-1-0716-2740-2_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
T lymphocytes (T cells) are essential components of the adaptive immune system; they serve multiple functions in responses to pathogens and to ensure immune homeostasis. Written for readers first entering this field of study, this chapter is a brief overview of the development of T cells in the thymus, from the entry of thymus-settling bone marrow-derived precursors to the egress of mature T cells. Surveyed topics include the differentiation and expansion of early precursors, the generation of the T cell antigen receptor repertoire, the selection of αβ T cell precursors, and their acquisition of functional competency.
Collapse
Affiliation(s)
- Rémy Bosselut
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
5
|
Salz A, Gurniak C, Jönsson F, Witke W. Cofilin1-driven actin dynamics controls migration of thymocytes and is essential for positive selection in the thymus. J Cell Sci 2020; 133:jcs238048. [PMID: 31974112 DOI: 10.1242/jcs.238048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 01/14/2020] [Indexed: 11/20/2022] Open
Abstract
Actin dynamics is essential for T-cell development. We show here that cofilin1 is the key molecule for controlling actin filament turnover in this process. Mice with specific depletion of cofilin1 in thymocytes showed increased steady-state levels of actin filaments, and associated alterations in the pattern of thymocyte migration and adhesion. Our data suggest that cofilin1 is controlling oscillatory F-actin changes, a parameter that influences the migration pattern in a 3-D environment. In a collagen matrix, cofilin1 controls the speed and resting intervals of migrating thymocytes. Cofilin1 was not involved in thymocyte proliferation, cell survival, apoptosis or surface receptor trafficking. However, in cofilin1 mutant mice, impaired adhesion and migration resulted in a specific block of thymocyte differentiation from CD4/CD8 double-positive thymocytes towards CD4 and CD8 single-positive cells. Our data suggest that tuning of the dwelling time of thymocytes in the thymic niches is tightly controlled by cofilin1 and essential for positive selection during T-cell differentiation. We describe a novel role of cofilin1 in the physiological context of migration-dependent cell differentiation.
Collapse
Affiliation(s)
- Andree Salz
- Institute of Genetics, University of Bonn, Karlrobert-Kreiten Strasse 13, 53115 Bonn, Germany
| | - Christine Gurniak
- Institute of Genetics, University of Bonn, Karlrobert-Kreiten Strasse 13, 53115 Bonn, Germany
| | - Friederike Jönsson
- Unit of Antibodies in Therapy and Pathology, Institut Pasteur, UMR 1222 INSERM, 75015 Paris, France
| | - Walter Witke
- Institute of Genetics, University of Bonn, Karlrobert-Kreiten Strasse 13, 53115 Bonn, Germany
| |
Collapse
|
6
|
Glynn RA, Bassing CH. From RAG2 to T Cell Riches and Future Fortunes. THE JOURNAL OF IMMUNOLOGY 2019; 202:1315-1316. [PMID: 30782849 DOI: 10.4049/jimmunol.1900010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Affiliation(s)
- Rebecca A Glynn
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Craig H Bassing
- Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104; and Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| |
Collapse
|
7
|
Danelli L, Donnarumma T, Kassiotis G. Correlates of Follicular Helper Bias in the CD4 T Cell Response to a Retroviral Antigen. Front Immunol 2018; 9:1260. [PMID: 29951052 PMCID: PMC6008654 DOI: 10.3389/fimmu.2018.01260] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/18/2018] [Indexed: 12/13/2022] Open
Abstract
CD4+ T cell differentiation is influenced by a plethora of intrinsic and extrinsic factors, providing the immune system with the ability to tailor its response according to specific stimuli. Indeed, different classes of pathogens may induce a distinct balance of CD4+ T cell differentiation programmes. Here, we report an uncommonly strong bias toward follicular helper (Tfh) differentiation of CD4+ T cells reactive with a retroviral envelope glycoprotein model antigen, presented in its natural context during retroviral infection. Conversely, the response to the same antigen, presented in different immunization regimens, elicited a response typically balanced between Tfh and T helper 1 cells. Comprehensive quantitation of variables known to influence Tfh differentiation revealed the closest correlation with the strength of T cell receptor (TCR) signaling, leading to PD-1 expression, but not with surface TCR downregulation, irrespective of TCR clonotypic avidity. In contrast, strong TCR signaling leading to TCR downregulation and induction of LAG3 expression in high TCR avidity clonotypes restrained CD4+ T cell commitment and further differentiation. Finally, stunted Th1 differentiation, correlating with limited IL-2 availability in retroviral infection, provided permissive conditions for Tfh development, suggesting that Tfh differentiation is the default program of envelope-reactive CD4+ T cells.
Collapse
Affiliation(s)
- Luca Danelli
- Retroviral Immunology, The Francis Crick Institute, London, United Kingdom
| | - Tiziano Donnarumma
- Retroviral Immunology, The Francis Crick Institute, London, United Kingdom
| | - George Kassiotis
- Retroviral Immunology, The Francis Crick Institute, London, United Kingdom
- Department of Medicine, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
8
|
Pao W, Ooi CH, Birzele F, Ruefli-Brasse A, Cannarile MA, Reis B, Scharf SH, Schubert DA, Hatje K, Pelletier N, Spleiss O, Reed JC. Tissue-Specific Immunoregulation: A Call for Better Understanding of the "Immunostat" in the Context of Cancer. Cancer Discov 2018; 8:395-402. [PMID: 29545369 DOI: 10.1158/2159-8290.cd-17-1320] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/26/2018] [Accepted: 02/01/2018] [Indexed: 11/16/2022]
Abstract
Checkpoint inhibitor therapy has been a breakthrough in cancer research, but only some patients with cancer derive substantial benefit. Although mechanisms underlying sensitivity and resistance to checkpoint inhibitors are being elucidated, the importance of organ-specific regulation of immunity is currently underappreciated. Here, we call for a greater understanding of tissue-specific immunoregulation, namely, "tissue-specific immunostats," to make advances in treatments for cancer. A better understanding of how individual organs at baseline regulate the immune system could enable an improved precision medicine approach to cancer immunotherapy. Cancer Discov; 8(4); 395-402. ©2018 AACR.
Collapse
Affiliation(s)
- William Pao
- Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland.
| | - Chia-Huey Ooi
- Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Fabian Birzele
- Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | | | | | - Bernhard Reis
- Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Sebastian H Scharf
- Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - David A Schubert
- Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Klas Hatje
- Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Nadege Pelletier
- Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - Olivia Spleiss
- Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| | - John C Reed
- Roche Innovation Center Basel, F. Hoffmann-La Roche AG, Basel, Switzerland
| |
Collapse
|
9
|
Driver JP, Racine JJ, Ye C, Lamont DJ, Newby BN, Leeth CM, Chapman HD, Brusko TM, Chen YG, Mathews CE, Serreze DV. Interferon-γ Limits Diabetogenic CD8 + T-Cell Effector Responses in Type 1 Diabetes. Diabetes 2017; 66:710-721. [PMID: 27920091 PMCID: PMC5319715 DOI: 10.2337/db16-0846] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 11/29/2016] [Indexed: 01/18/2023]
Abstract
Type 1 diabetes development in the NOD mouse model is widely reported to be dependent on high-level production by autoreactive CD4+ and CD8+ T cells of interferon-γ (IFN-γ), generally considered a proinflammatory cytokine. However, IFN-γ can also participate in tolerance-induction pathways, indicating it is not solely proinflammatory. This study addresses how IFN-γ can suppress activation of diabetogenic CD8+ T cells. CD8+ T cells transgenically expressing the diabetogenic AI4 T-cell receptor adoptively transferred disease to otherwise unmanipulated NOD.IFN-γnull , but not standard NOD, mice. AI4 T cells only underwent vigorous intrasplenic proliferation in NOD.IFN-γnull recipients. Disease-protective IFN-γ could be derived from any lymphocyte source and suppressed diabetogenic CD8+ T-cell responses both directly and through an intermediary nonlymphoid cell population. Suppression was not dependent on regulatory T cells, but was associated with increased inhibitory STAT1 to STAT4 expression levels in pathogenic AI4 T cells. Importantly, IFN-γ exposure during activation reduced the cytotoxicity of human-origin type 1 diabetes-relevant autoreactive CD8+ T cells. Collectively, these results indicate that rather than marking the most proinflammatory lymphocytes in diabetes development, IFN-γ production could represent an attempted limitation of pathogenic CD8+ T-cell activation. Thus, great care should be taken when designing possible diabetic intervention approaches modulating IFN-γ production.
Collapse
Affiliation(s)
- John P Driver
- Department of Animal Sciences, University of Florida, Gainesville, FL
| | | | - Cheng Ye
- Department of Animal Sciences, University of Florida, Gainesville, FL
| | | | - Brittney N Newby
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL
| | | | | | - Todd M Brusko
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL
| | - Yi-Guang Chen
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI
| | - Clayton E Mathews
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL
| | | |
Collapse
|
10
|
Antibody-independent mechanisms regulate the establishment of chronic Plasmodium infection. Nat Microbiol 2017; 2:16276. [PMID: 28165471 DOI: 10.1038/nmicrobiol.2016.276] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 12/22/2016] [Indexed: 01/15/2023]
Abstract
Malaria is caused by parasites of the genus Plasmodium. All human-infecting Plasmodium species can establish long-lasting chronic infections1-5, creating an infectious reservoir to sustain transmission1,6. It is widely accepted that the maintenance of chronic infection involves evasion of adaptive immunity by antigenic variation7. However, genes involved in this process have been identified in only two of five human-infecting species: Plasmodium falciparum and Plasmodium knowlesi. Furthermore, little is understood about the early events in the establishment of chronic infection in these species. Using a rodent model we demonstrate that from the infecting population, only a minority of parasites, expressing one of several clusters of virulence-associated pir genes, establishes a chronic infection. This process occurs in different species of parasites and in different hosts. Establishment of chronicity is independent of adaptive immunity and therefore different from the mechanism proposed for maintenance of chronic P. falciparum infections7-9. Furthermore, we show that the proportions of parasites expressing different types of pir genes regulate the time taken to establish a chronic infection. Because pir genes are common to most, if not all, species of Plasmodium10, this process may be a common way of regulating the establishment of chronic infections.
Collapse
|
11
|
Nitta T, Suzuki H. Thymic stromal cell subsets for T cell development. Cell Mol Life Sci 2016; 73:1021-37. [PMID: 26825337 PMCID: PMC11108406 DOI: 10.1007/s00018-015-2107-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/26/2015] [Accepted: 12/01/2015] [Indexed: 12/20/2022]
Abstract
The thymus provides a specialized microenvironment in which a variety of stromal cells of both hematopoietic and non-hematopoietic origin regulate development and repertoire selection of T cells. Recent studies have been unraveling the inter- and intracellular signals and transcriptional networks for spatiotemporal regulation of development of thymic stromal cells, mainly thymic epithelial cells (TECs), and the molecular mechanisms of how different TEC subsets control T cell development and selection. TECs are classified into two functionally different subsets: cortical TECs (cTECs) and medullary TECs (mTECs). cTECs induce positive selection of diverse and functionally distinct T cells by virtue of unique antigen-processing systems, while mTECs are essential for establishing T cell tolerance via ectopic expression of peripheral tissue-restricted antigens and cooperation with dendritic cells. In addition to reviewing the role of the thymic stroma in conventional T cell development, we will discuss recently discovered novel functions of TECs in the development of unconventional T cells, such as natural killer T cells and γδT cells.
Collapse
Affiliation(s)
- Takeshi Nitta
- Department of Immunology and Pathology, Research Institute, National Center for Global Health and Medicine, Chiba, 272-8516, Japan.
| | - Harumi Suzuki
- Department of Immunology and Pathology, Research Institute, National Center for Global Health and Medicine, Chiba, 272-8516, Japan.
| |
Collapse
|
12
|
Satoh R, Kakugawa K, Yasuda T, Yoshida H, Sibilia M, Katsura Y, Levi B, Abramson J, Koseki Y, Koseki H, van Ewijk W, Hollander GA, Kawamoto H. Requirement of Stat3 Signaling in the Postnatal Development of Thymic Medullary Epithelial Cells. PLoS Genet 2016; 12:e1005776. [PMID: 26789017 PMCID: PMC4720355 DOI: 10.1371/journal.pgen.1005776] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 12/08/2015] [Indexed: 01/20/2023] Open
Abstract
Thymic medullary regions are formed in neonatal mice as islet-like structures, which increase in size over time and eventually fuse a few weeks after birth into a continuous structure. The development of medullary thymic epithelial cells (TEC) is dependent on NF-κB associated signaling though other signaling pathways may contribute. Here, we demonstrate that Stat3-mediated signals determine medullary TEC cellularity, architectural organization and hence the size of the medulla. Deleting Stat3 expression selectively in thymic epithelia precludes the postnatal enlargement of the medulla retaining a neonatal architecture of small separate medullary islets. In contrast, loss of Stat3 expression in cortical TEC neither affects the cellularity or organization of the epithelia. Activation of Stat3 is mainly positioned downstream of EGF-R as its ablation in TEC phenocopies the loss of Stat3 expression in these cells. These results indicate that Stat3 meditated signal via EGF-R is required for the postnatal development of thymic medullary regions. Thymic medulla is known to be an essential site for the deletion of auto-reactive T cells. Whereas it has been well documented that the development of medullary thymic epithelial cells (mTECs) depends on NF-κB associated signaling, it remained unclear whether other signaling pathways are also involved. In this context, it had been reported that conditional deletion of Stat3 alleles in TECs using cytokeratin-5 (CK5) promoter controlled Cre expression results in a profound impairment in TEC development. However, a detailed analysis of phenotypes in mTECs remained unstudied. In the present study, we show that thymic medullary regions remain as small islets when Stat3 is conditionally deleted in thymic epithelial cells, while they normal fuse to form continuous structures during postnatal development. Furthermore, we identified EGF-R mediated signal to be placed upstream of Stat3 activation, as its ablation phenocopied the loss of Stat3 expression in TECs. Thus, the present study revealed that Stat3 is required for the postnatal development of medullary regions.
Collapse
Affiliation(s)
- Rumi Satoh
- Laboratory for Lymphocyte Development, RIKEN Research Center for Allergy and Immunology, Yokohama, Japan
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Kiyokazu Kakugawa
- Laboratory for Lymphocyte Development, RIKEN Research Center for Allergy and Immunology, Yokohama, Japan
- Laboratory for Immune Crosstalk, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Takuwa Yasuda
- Laboratory for Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokoham, Japan
| | - Hisahiro Yoshida
- Laboratory for Immunogenetics, RIKEN Center for Integrative Medical Sciences, Yokoham, Japan
| | - Maria Sibilia
- Department of Medicine I, Institute of Cancer Research, Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria
| | - Yoshimoto Katsura
- Division of Cell Regeneration and Transplantation, Advanced Medical Research Center, Nihon University School of Medicine, Tokyo, Japan
| | - Ben Levi
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Jakub Abramson
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yoko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Haruhiko Koseki
- Laboratory for Developmental Genetics, RIKEN Center for Integrative Medical Sciences, Yokohama, Japan
| | - Willem van Ewijk
- Department of Molecular Cell Biology and Department of Immunology, Leiden University Medical Center, RA Leiden, the Netherlands
| | - Georg A. Hollander
- Laboratory of Pediatric Immunology, Center for Biomedicine, University of Basel, and the University Children’s Hospital, Basel, Switzerland
- Laboratory of Developmental Immunology Weatherall Institute of Molecular Medicine and Department of Paediatrics, University of Oxford, Oxford, United Kingdom
| | - Hiroshi Kawamoto
- Laboratory for Lymphocyte Development, RIKEN Research Center for Allergy and Immunology, Yokohama, Japan
- Department of Immunology, Institute for Frontier Medical Sciences, Kyoto University, Kyoto, Japan
- * E-mail:
| |
Collapse
|
13
|
Stepwise B-cell-dependent expansion of T helper clonotypes diversifies the T-cell response. Nat Commun 2016; 7:10281. [PMID: 26728651 PMCID: PMC4728444 DOI: 10.1038/ncomms10281] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 11/24/2015] [Indexed: 01/07/2023] Open
Abstract
Antigen receptor diversity underpins adaptive immunity by providing the ground for clonal selection of lymphocytes with the appropriate antigen reactivity. Current models attribute T cell clonal selection during the immune response to T-cell receptor (TCR) affinity for either foreign or self peptides. Here, we report that clonal selection of CD4(+) T cells is also extrinsically regulated by B cells. In response to viral infection, the antigen-specific TCR repertoire is progressively diversified by staggered clonotypic expansion, according to functional avidity, which correlates with self-reactivity. Clonal expansion of lower-avidity T-cell clonotypes depends on availability of MHC II-expressing B cells, in turn influenced by B-cell activation. B cells clonotypically diversify the CD4(+) T-cell response also to vaccination or tumour challenge, revealing a common effect.
Collapse
|
14
|
Analyses of the TCR repertoire of MHC class II-restricted innate CD4⁺ T cells. Exp Mol Med 2015; 47:e154. [PMID: 25813222 PMCID: PMC4351420 DOI: 10.1038/emm.2015.7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 11/05/2014] [Accepted: 12/04/2014] [Indexed: 11/08/2022] Open
Abstract
Analysis of the T-cell receptor (TCR) repertoire of innate CD4(+) T cells selected by major histocompatibility complex (MHC) class II-dependent thymocyte-thymocyte (T-T) interaction (T-T CD4(+) T cells) is essential for predicting the characteristics of the antigens that bind to these T cells and for distinguishing T-T CD4(+) T cells from other types of innate T cells. Using the TCR(mini) Tg mouse model, we show that the repertoire of TCRα chains in T-T CD4(+) T cells was extremely diverse, in contrast to the repertoires previously described for other types of innate T cells. The TCRα chain sequences significantly overlapped between T-T CD4(+) T cells and conventional CD4(+) T cells in the thymus and spleen. However, the diversity of the TCRα repertoire of T-T CD4(+) T cells seemed to be restricted compared with that of conventional CD4(+) T cells. Interestingly, the frequency of the parental OT-II TCRα chains was significantly reduced in the process of T-T interaction. This diverse and shifted repertoire in T-T CD4(+) T cells has biological relevance in terms of defense against diverse pathogens and a possible regulatory role during peripheral T-T interaction.
Collapse
|
15
|
Pérez-Mazliah D, Ng DHL, Freitas do Rosário AP, McLaughlin S, Mastelic-Gavillet B, Sodenkamp J, Kushinga G, Langhorne J. Disruption of IL-21 signaling affects T cell-B cell interactions and abrogates protective humoral immunity to malaria. PLoS Pathog 2015; 11:e1004715. [PMID: 25763578 PMCID: PMC4370355 DOI: 10.1371/journal.ppat.1004715] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 01/29/2015] [Indexed: 12/22/2022] Open
Abstract
Interleukin-21 signaling is important for germinal center B-cell responses, isotype switching and generation of memory B cells. However, a role for IL-21 in antibody-mediated protection against pathogens has not been demonstrated. Here we show that IL-21 is produced by T follicular helper cells and co-expressed with IFN-γ during an erythrocytic-stage malaria infection of Plasmodium chabaudi in mice. Mice deficient either in IL-21 or the IL-21 receptor fail to resolve the chronic phase of P. chabaudi infection and P. yoelii infection resulting in sustained high parasitemias, and are not immune to re-infection. This is associated with abrogated P. chabaudi-specific IgG responses, including memory B cells. Mixed bone marrow chimeric mice, with T cells carrying a targeted disruption of the Il21 gene, or B cells with a targeted disruption of the Il21r gene, demonstrate that IL-21 from T cells signaling through the IL-21 receptor on B cells is necessary to control chronic P. chabaudi infection. Our data uncover a mechanism by which CD4+ T cells and B cells control parasitemia during chronic erythrocytic-stage malaria through a single gene, Il21, and demonstrate the importance of this cytokine in the control of pathogens by humoral immune responses. These data are highly pertinent for designing malaria vaccines requiring long-lasting protective B-cell responses. The importance of antibody and B-cell responses for control of the erythrocytic-stage of the malaria parasite, Plasmodium, was first described when immune serum, passively transferred into Plasmodium falciparum-infected children, reduced parasitemia. This was later confirmed in experimental models in which mice deficient in B cells were unable to eliminate erythrocytic-stage infections. The signals required to activate these protective long-lasting B cell responses towards Plasmodium have not been investigated. IL-21 has been shown to be important for development of B-cell responses after immunization; however, a direct requirement for IL-21 in the control of infection via B-cell dependent mechanisms has never been demonstrated. In this paper, we have used mouse models of erythrocytic P. chabaudi and P. yoelii 17X(NL) infections in combination with IL-21/IL-21R deficiency to show that IL-21 from CD4+ T cells is required to eliminate Plasmodium infection by activating protective, long-lasting B-cell responses. Disruption of IL-21 signaling in B cells prevents the elimination of the parasite resulting in sustained high parasitemias, with no development of memory B-cells, lack of antigen-specific plasma cells and antibodies, and thus no protective immunity against a second challenge infection. Our data demonstrate the absolute requirement of IL-21 for B-cell control of this systemic infection. This has important implications for the design of vaccines against Plasmodium.
Collapse
Affiliation(s)
- Damián Pérez-Mazliah
- Division of Parasitology, MRC National Institute for Medical Research (NIMR), London, United Kingdom
| | - Dorothy Hui Lin Ng
- Division of Parasitology, MRC National Institute for Medical Research (NIMR), London, United Kingdom
| | | | - Sarah McLaughlin
- Division of Parasitology, MRC National Institute for Medical Research (NIMR), London, United Kingdom
| | - Béatris Mastelic-Gavillet
- Division of Parasitology, MRC National Institute for Medical Research (NIMR), London, United Kingdom
| | - Jan Sodenkamp
- Division of Parasitology, MRC National Institute for Medical Research (NIMR), London, United Kingdom
| | - Garikai Kushinga
- Division of Parasitology, MRC National Institute for Medical Research (NIMR), London, United Kingdom
| | - Jean Langhorne
- Division of Parasitology, MRC National Institute for Medical Research (NIMR), London, United Kingdom
- * E-mail:
| |
Collapse
|
16
|
Khairallah C, Netzer S, Villacreces A, Juzan M, Rousseau B, Dulanto S, Giese A, Costet P, Praloran V, Moreau JF, Dubus P, Vermijlen D, Déchanet-Merville J, Capone M. γδ T cells confer protection against murine cytomegalovirus (MCMV). PLoS Pathog 2015; 11:e1004702. [PMID: 25747674 PMCID: PMC4352080 DOI: 10.1371/journal.ppat.1004702] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Accepted: 01/24/2015] [Indexed: 12/28/2022] Open
Abstract
Cytomegalovirus (CMV) is a leading infectious cause of morbidity in immune-compromised patients. γδ T cells have been involved in the response to CMV but their role in protection has not been firmly established and their dependency on other lymphocytes has not been addressed. Using C57BL/6 αβ and/or γδ T cell-deficient mice, we here show that γδ T cells are as competent as αβ T cells to protect mice from CMV-induced death. γδ T cell-mediated protection involved control of viral load and prevented organ damage. γδ T cell recovery by bone marrow transplant or adoptive transfer experiments rescued CD3ε−/− mice from CMV-induced death confirming the protective antiviral role of γδ T cells. As observed in humans, different γδ T cell subsets were induced upon CMV challenge, which differentiated into effector memory cells. This response was observed in the liver and lungs and implicated both CD27+ and CD27− γδ T cells. NK cells were the largely preponderant producers of IFNγ and cytotoxic granules throughout the infection, suggesting that the protective role of γδ T cells did not principally rely on either of these two functions. Finally, γδ T cells were strikingly sufficient to fully protect Rag−/−γc−/− mice from death, demonstrating that they can act in the absence of B and NK cells. Altogether our results uncover an autonomous protective antiviral function of γδ T cells, and open new perspectives for the characterization of a non classical mode of action which should foster the design of new γδ T cell based therapies, especially useful in αβ T cell compromised patients. γδ T cells are unconventional T lymphocytes that play a unique role in host protection against pathogens. Human Cytomegalovirus (HCMV) is a widespread virus that can cause severe organ disease such as hepatitis and pneumonitis in immune-compromised patients. Our decade-long study conveys compelling evidence for the implication of human γδ T cells in the immune response against HCMV, but their protective role could not be formally demonstrated in humans. In the present study we use the murine model of CMV infection which allows the spatial and temporal analysis of viral spread and anti-viral immune responses. We show that, in the absence of αβ T cells, γδ T cells control MCMV-induced hepatitis, pneumonitis and death by restricting viral load in the liver, lungs and spleen. γδ T cells expand in these organs and display memory features that could be further incorporated into vaccination strategies. In conclusion, γδ T cells represent an important arm in the immune response against CMV infection that could be particularly important in the context of αβ T cell immune-suppression.
Collapse
Affiliation(s)
- Camille Khairallah
- Université de Bordeaux, Bordeaux, France
- CNRS, UMR 5164, Bordeaux, France
| | - Sonia Netzer
- Université de Bordeaux, Bordeaux, France
- CNRS, UMR 5164, Bordeaux, France
| | - Arnaud Villacreces
- Université de Bordeaux, Bordeaux, France
- CNRS, UMR 5164, Bordeaux, France
| | - Marina Juzan
- Université de Bordeaux, Bordeaux, France
- CNRS, UMR 5164, Bordeaux, France
| | - Benoît Rousseau
- Université de Bordeaux, Bordeaux, France
- Laboratoire d’Immunologie et d’Immunogénétique, Animalerie A2, Bordeaux, France
| | - Sara Dulanto
- Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Alban Giese
- Université de Bordeaux, Bordeaux, France
- EA2406 Histologie et pathologie moléculaire des tumeurs, Bordeaux, France
| | - Pierre Costet
- Université de Bordeaux, Bordeaux, France
- Animalerie spécialisée, Bordeaux, France
| | - Vincent Praloran
- Université de Bordeaux, Bordeaux, France
- CNRS, UMR 5164, Bordeaux, France
- Laboratoire d’Hématologie, Centre Hospitalo-Universitaire, Bordeaux, France
| | - Jean-François Moreau
- Université de Bordeaux, Bordeaux, France
- CNRS, UMR 5164, Bordeaux, France
- Centre Hospitalo-Universitaire, Bordeaux, France
| | - Pierre Dubus
- Université de Bordeaux, Bordeaux, France
- EA2406 Histologie et pathologie moléculaire des tumeurs, Bordeaux, France
| | - David Vermijlen
- Faculty of Pharmacy, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Institute for Medical Immunology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Julie Déchanet-Merville
- Université de Bordeaux, Bordeaux, France
- CNRS, UMR 5164, Bordeaux, France
- * E-mail: (JDM); (MC)
| | - Myriam Capone
- Université de Bordeaux, Bordeaux, France
- CNRS, UMR 5164, Bordeaux, France
- * E-mail: (JDM); (MC)
| |
Collapse
|
17
|
Spidale NA, Wang B, Tisch R. Cutting edge: Antigen-specific thymocyte feedback regulates homeostatic thymic conventional dendritic cell maturation. THE JOURNAL OF IMMUNOLOGY 2014; 193:21-5. [PMID: 24890722 DOI: 10.4049/jimmunol.1400321] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Thymic dendritic cells (DC) mediate self-tolerance by presenting self-peptides to and depleting autoreactive thymocytes. Despite a significant role in negative selection, the events regulating thymic DC maturation and function under steady-state conditions are poorly understood. We report that cross-talk with thymocytes regulates thymic conventional DC (cDC) numbers, phenotype, and function. In mice lacking TCR-expressing thymocytes, thymic cDC were reduced and exhibited a less mature phenotype. Furthermore, thymic cDC in TCR-transgenic mice lacking cognate Ag expression in the thymus were also immature; notably, however, thymic cDC maturation was re-established by an Ag-specific cognate interaction with CD4+ or CD8+ single-positive thymocytes (SP). Blockade of CD40L during Ag-specific interactions with CD4 SP, but not CD8 SP, limited the effect on cDC maturation. Together, these novel findings demonstrate that homeostatic maturation and function of thymic cDC are regulated by feedback delivered by CD4 SP and CD8 SP via distinct mechanisms during a cognate Ag-specific interaction.
Collapse
Affiliation(s)
- Nicholas A Spidale
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599; and
| | - Bo Wang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599; and
| | - Roland Tisch
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599; and Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC 27599
| |
Collapse
|
18
|
Neunkirchner A, Schmetterer KG, Pickl WF. Lymphocyte-based model systems for allergy research: a historic overview. Int Arch Allergy Immunol 2014; 163:259-91. [PMID: 24777172 PMCID: PMC7617143 DOI: 10.1159/000360163] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
During the last decades, a multitude of studies applying distinct in vitro and in vivo model systems have contributed greatly to our better understanding of the initiation and regulation of inflammatory processes leading to allergic diseases. Over the years, it has become evident that among lymphocytes, not only IgE-producing B cells and allergy-orchestrating CD4(+) helper cells but also cytotoxic CD8(+) T cells, γδ-T cells and innate lymphoid cells, as well as regulatory lymphocytes, might critically shape the immune response towards usually innocuous allergens. In this review, we provide a historic overview of pioneering work leading to the establishment of important lymphocyte-based model systems for allergy research. Moreover, we contrast the original findings with our currently more refined knowledge to appreciate the actual validity of the respective models and to reassess the conclusions obtained from them. Conflicting studies and interpretations are identified and discussed. The tables are intended to provide an easy overview of the field not only for scientists newly entering the field but also for the broader readership interested in updating their knowledge. Along those lines, herein we discuss in vitro and in vivo approaches to the investigation of lymphocyte effector cell activation, polarization and regulation, and describe depletion and adoptive transfer models along with gene knockout and transgenic (tg) methodologies. In addition, novel attempts to establish humanized T cell antigen receptor tg mouse models for allergy research are described and discussed.
Collapse
Affiliation(s)
- Alina Neunkirchner
- Christian Doppler Laboratory for Immunomodulation, Medical University of Vienna, Vienna, Austria
| | | | | |
Collapse
|
19
|
Williams JA, Zhang J, Jeon H, Nitta T, Ohigashi I, Klug D, Kruhlak MJ, Choudhury B, Sharrow SO, Granger L, Adams A, Eckhaus MA, Jenkinson SR, Richie ER, Gress RE, Takahama Y, Hodes RJ. Thymic medullary epithelium and thymocyte self-tolerance require cooperation between CD28-CD80/86 and CD40-CD40L costimulatory pathways. THE JOURNAL OF IMMUNOLOGY 2013; 192:630-40. [PMID: 24337745 DOI: 10.4049/jimmunol.1302550] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
A critical process during thymic development of the T cell repertoire is the induction of self-tolerance. Tolerance in developing T cells is highly dependent on medullary thymic epithelial cells (mTEC), and mTEC development in turn requires signals from mature single-positive thymocytes, a bidirectional relationship termed thymus crosstalk. We show that CD28-CD80/86 and CD40-CD40L costimulatory interactions, which mediate negative selection and self-tolerance, upregulate expression of LTα, LTβ, and receptor activator for NF-κB in the thymus and are necessary for medullary development. Combined absence of CD28-CD80/86 and CD40-CD40L results in profound deficiency in mTEC development comparable to that observed in the absence of single-positive thymocytes. This requirement for costimulatory signaling is maintained even in a TCR transgenic model of high-affinity TCR-ligand interactions. CD4 thymocytes maturing in the altered thymic epithelial environment of CD40/CD80/86 knockout mice are highly autoreactive in vitro and are lethal in congenic adoptive transfer in vivo, demonstrating a critical role for these costimulatory pathways in self-tolerance as well as thymic epithelial development. These findings demonstrate that cooperativity between CD28-CD80/86 and CD40-CD40L pathways is required for normal medullary epithelium and for maintenance of self-tolerance in thymocyte development.
Collapse
Affiliation(s)
- Joy A Williams
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
TRAF3 enforces the requirement for T cell cross-talk in thymic medullary epithelial development. Proc Natl Acad Sci U S A 2013; 110:21107-12. [PMID: 24324158 DOI: 10.1073/pnas.1314859111] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Induction of self-tolerance in developing T cells depends on medullary thymic epithelial cells (mTECs), whose development, in turn, requires signals from single-positive (SP) thymocytes. Thus, the absence of SP thymocytes in Tcra(-/-) mice results in a profound deficiency in mTECs. Here, we have probed the mechanism that underlies this requirement for cross-talk with thymocytes in medullary development. Previous studies have implicated nonclassical NF-κB as a pathway important in the development of mTECs, because mice lacking RelB, NIK, or IKKα, critical components of this pathway, have an almost complete absence of mTECs, with resulting autoimmune pathology. We therefore assessed the effect of selective deletion in TEC of TNF receptor-associated factor 3 (TRAF3), an inhibitor of nonclassical NF-κB signaling. Deletion of TRAF3 in thymic epithelial cells allowed RelB-dependent development of normal numbers of AIRE-expressing mTECs in the complete absence of SP thymocytes. Thus, mTEC development can occur in the absence of cross-talk with SP thymocytes, and signals provided by SP T cells are needed to overcome TRAF3-imposed arrest in mTEC development mediated by inhibition of nonclassical NF-κB. We further observed that TRAF3 deletion is also capable of overcoming all requirements for LTβR and CD40, which are otherwise necessary for mTEC development, but is not sufficient to overcome the requirement for RANKL, indicating a role for RANKL that is distinct from the signals provided by SP thymocytes. We conclude that TRAF3 plays a central role in regulation of mTEC development by imposing requirements for SP T cells and costimulation-mediated cross-talk in generation of the medullary compartment.
Collapse
|
21
|
Linowes BA, Ligons DL, Nam AS, Hong C, Keller HR, Tai X, Luckey MA, Park JH. Pim1 permits generation and survival of CD4+ T cells in the absence of γc cytokine receptor signaling. Eur J Immunol 2013; 43:2283-94. [PMID: 23712827 DOI: 10.1002/eji.201242686] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Revised: 05/06/2013] [Accepted: 05/21/2013] [Indexed: 01/02/2023]
Abstract
γ-Chain (γc) cytokine receptor signaling is required for the development of all lymphocytes. Why γc signaling plays such an essential role is not fully understood, but induction of the serine/threonine kinase Pim1 is considered a major downstream event of γc as Pim1 prevents apoptosis and increases metabolic activity. Consequently, we asked whether Pim1 overexpression would suffice to restore lymphocyte development in γc-deficient mice. By analyzing Pim1-transgenic γc-deficient mice (Pim1(Tg) γc(KO) ), we show that Pim1 promoted T-cell development and survival in the absence of γc. Interestingly, such effects were largely limited to CD4(+) lineage αβ T cells as CD4(+) T-cell numbers improved to near normal levels but CD8(+) T cells remained severely lymphopenic. Notably, Pim1 over-expression failed to promote development and survival of any T-lineage cells other than αβ T cells, as we observed complete lack of γδ, NKT, FoxP3(+) T regulatory cells and TCR-β(+) CD8αα IELs in Pim1(Tg) γc(KO) mice. Collectively, these results uncover distinct requirements for γc signaling between CD4(+) αβ T cells and all other T-lineage cells, and they identify Pim1 as a novel effector molecule sufficient to drive CD4(+) αβ T-cell development and survival in the absence of γc cytokine receptor signaling.
Collapse
Affiliation(s)
- Brett A Linowes
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Division of Graduate Medical Sciences, Boston University School of Medicine, Boston, MA, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Dysregulated signaling pathways in the development of CNTRL-FGFR1-induced myeloid and lymphoid malignancies associated with FGFR1 in human and mouse models. Blood 2013; 122:1007-16. [PMID: 23777766 DOI: 10.1182/blood-2013-03-489823] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myeloid and lymphoid neoplasm associated with FGFR1 is an aggressive disease, and resistant to all the current chemotherapies. To define the molecular etiology of this disease, we have developed murine models of this disease, in syngeneic hosts as well as in nonobese diabetic/severe combined immunodeficiency/interleukin 2Rγ(null) mice engrafted with transformed human CD34+ hematopoietic stem/progenitor cells. Both murine models mimic the human disease with splenohepatomegaly, hypercellular bone marrow, and myeloproliferative neoplasms that progresses to acute myeloid leukemia. Molecular genetic analyses of these model mice, as well as primary human disease, demonstrated that CNTRL-FGFR1, through abnormal activation of several signaling pathways related to development and differentiation of both myeloid and T-lymphoid cells, contribute to overt leukemogenesis. Clonal evolution analysis indicates that myeloid related neoplasms arise from common myeloid precursor cells that retain potential for T-lymphoid differentiation. These data indicate that simultaneously targeting these pathways is essential to successfully treating this almost invariably lethal disease.
Collapse
|
23
|
Plasticity of Th17 cells in Peyer's patches is responsible for the induction of T cell-dependent IgA responses. Nat Immunol 2013; 14:372-9. [PMID: 23475182 PMCID: PMC3672955 DOI: 10.1038/ni.2552] [Citation(s) in RCA: 396] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2012] [Accepted: 01/22/2013] [Indexed: 12/12/2022]
Abstract
Intestinal Peyer’s patches are essential lymphoid organs for the generation of T cell-dependent immunoglobulin (Ig) A production for gut homeostasis. Using IL-17 fate reporter mice we show here that endogenous TH17 cells in lymphoid organs of naïve mice home preferentially to the intestine and are maintained independently of IL-23. In Peyer’s patches such TH17 cells acquire a T follicular helper (TFH) phenotype and induce the development of IgA-producing germinal center B cells. Mice deficient in TH17 cells fail to generate antigen specific IgA responses, providing evidence that TH17 cells are the crucial subset required for high affinity T cell-dependent IgA production.
Collapse
|
24
|
Napolitano A, Pittoni P, Beaudoin L, Lehuen A, Voehringer D, MacDonald HR, Dellabona P, Casorati G. Functional education of invariant NKT cells by dendritic cell tuning of SHP-1. THE JOURNAL OF IMMUNOLOGY 2013; 190:3299-308. [PMID: 23427253 DOI: 10.4049/jimmunol.1203466] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Invariant NKT (iNKT) cells play key roles in host defense by recognizing lipid Ags presented by CD1d. iNKT cells are activated by bacterial-derived lipids and are also strongly autoreactive toward self-lipids. iNKT cell responsiveness must be regulated to maintain effective host defense while preventing uncontrolled stimulation and potential autoimmunity. CD1d-expressing thymocytes support iNKT cell development, but thymocyte-restricted expression of CD1d gives rise to Ag hyperresponsive iNKT cells. We hypothesized that iNKT cells require functional education by CD1d(+) cells other than thymocytes to set their correct responsiveness. In mice that expressed CD1d only on thymocytes, hyperresponsive iNKT cells in the periphery expressed significantly reduced levels of tyrosine phosphatase SHP-1, a negative regulator of TCR signaling. Accordingly, heterozygous SHP-1 mutant mice displaying reduced SHP-1 expression developed a comparable population of Ag hyperresponsive iNKT cells. Restoring nonthymocyte CD1d expression in transgenic mice normalized SHP-1 expression and iNKT cell reactivity. Radiation chimeras revealed that CD1d(+) dendritic cells supported iNKT cell upregulation of SHP-1 and decreased responsiveness after thymic emigration. Hence, dendritic cells functionally educate iNKT cells by tuning SHP-1 expression to limit reactivity.
Collapse
Affiliation(s)
- Anna Napolitano
- Experimental Immunology Unit, Division of Immunology, Transplantation and Infectious Diseases, San Raffaele Scientific Institute, 20132 Milano, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Antigen recognition by autoreactive CD4⁺ thymocytes drives homeostasis of the thymic medulla. PLoS One 2012; 7:e52591. [PMID: 23300712 PMCID: PMC3531460 DOI: 10.1371/journal.pone.0052591] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Accepted: 11/16/2012] [Indexed: 01/07/2023] Open
Abstract
The thymic medulla is dedicated for purging the T-cell receptor (TCR) repertoire of self-reactive specificities. Medullary thymic epithelial cells (mTECs) play a pivotal role in this process because they express numerous peripheral tissue-restricted self-antigens. Although it is well known that medulla formation depends on the development of single-positive (SP) thymocytes, the mechanisms underlying this requirement are incompletely understood. We demonstrate here that conventional SP CD4+ thymocytes bearing autoreactive TCRs drive a homeostatic process that fine-tunes medullary plasticity in adult mice by governing the expansion and patterning of the medulla. This process exhibits strict dependence on TCR-reactivity with self-antigens expressed by mTECs, as well as engagement of the CD28-CD80/CD86 costimulatory axis. These interactions induce the expression of lymphotoxin α in autoreactive CD4+ thymocytes and RANK in mTECs. Lymphotoxin in turn drives mTEC development in synergy with RANKL and CD40L. Our results show that Ag-dependent interactions between autoreactive CD4+ thymocytes and mTECs fine-tune homeostasis of the medulla by completing the signaling axes implicated in mTEC expansion and medullary organization.
Collapse
|
26
|
Irla M, Guenot J, Sealy G, Reith W, Imhof BA, Sergé A. Three-dimensional visualization of the mouse thymus organization in health and immunodeficiency. THE JOURNAL OF IMMUNOLOGY 2012; 190:586-96. [PMID: 23248258 DOI: 10.4049/jimmunol.1200119] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Lymphoid organs exhibit complex structures tightly related to their function. Surprisingly, although the thymic medulla constitutes a specialized microenvironment dedicated to the induction of T cell tolerance, its three-dimensional topology remains largely elusive because it has been studied mainly in two dimensions using thymic sections. To overcome this limitation, we have developed an automated method for full organ reconstruction in three dimensions, allowing visualization of intact mouse lymphoid organs from a collection of immunolabeled slices. We validated full organ reconstruction in three dimensions by reconstructing the well-characterized structure of skin-draining lymph nodes, before revisiting the complex and poorly described corticomedullary organization of the thymus. Wild-type thymi contain ~200 small medullae that are connected to or separated from a major medullary compartment. In contrast, thymi of immunodeficient Rag2(-/-) mice exhibit only ~20 small, unconnected medullary islets. Upon total body irradiation, medullary complexity was partially reduced and then recovered upon bone marrow transplantation. This intricate topology presents fractal properties, resulting in a considerable corticomedullary area. This feature ensures short distances between cortex and medulla, hence efficient thymocyte migration, as assessed by mathematical models. Remarkably, this junction is enriched, particularly in neonates, in medullary thymic epithelial cells expressing the autoimmune regulator. The emergence of a major medullary compartment is induced by CD4(+) thymocytes via CD80/86 and lymphotoxin-α signals. This comprehensive three-dimensional view of the medulla emphasizes a complex topology favoring efficient interactions between developing T cells and autoimmune regulator-positive medullary thymic epithelial cells, a key process for central tolerance induction.
Collapse
Affiliation(s)
- Magali Irla
- Department of Pathology and Immunology, University of Geneva, 1211 Geneva, Switzerland.
| | | | | | | | | | | |
Collapse
|
27
|
Young GR, Eksmond U, Salcedo R, Alexopoulou L, Stoye JP, Kassiotis G. Resurrection of endogenous retroviruses in antibody-deficient mice. Nature 2012; 491:774-8. [PMID: 23103862 PMCID: PMC3511586 DOI: 10.1038/nature11599] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2012] [Accepted: 09/18/2012] [Indexed: 01/12/2023]
Abstract
The mammalian host has developed a long-standing symbiotic relationship with a considerable number of microbial species. These include the microbiota on environmental surfaces, such as the respiratory and gastrointestinal tracks1, and also endogenous retroviruses (ERVs), comprising a substantial fraction of the mammalian genome2,3. The long-term consequences for the host of interaction with these microbial species can range from mutualism to parasitism and are not always completely understood. The potential impact of one microbial symbiont on another is even less clear. We have studied the control of ERVs in the commonly-used C57BL/6 (B6) mouse strain, which lacks endogenous murine leukaemia viruses (MLVs) able to replicate in murine cells. We demonstrate the spontaneous emergence of fully infectious ecotropic4 MLV (eMLV) in B6 mice with a range of distinct immune deficiencies affecting antibody production. These recombinant retroviruses establish infection of immunodeficient mouse colonies, and ultimately result in retrovirus-induced lymphomas. Notably, ERV activation in immune-deficient mice is prevented in husbandry conditions associated with reduced or absent intestinal microbiota. Our results shed light onto a previously unappreciated role for immunity in the control of ERVs and provide a potential mechanistic link between immune activation by microbial triggers and a range of pathologies associated with ERVs, including cancer.
Collapse
Affiliation(s)
- George R Young
- Division of Immunoregulation, MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, UK
| | | | | | | | | | | |
Collapse
|
28
|
Abstract
The bias of αβ T cells for MHC ligands has been proposed to be intrinsic to the T-cell receptor (TCR). Equally, the CD4 and CD8 coreceptors contribute to ligand restriction by colocalizing Lck with the TCR when MHC ligands are engaged. To determine the importance of intrinsic ligand bias, the germ-line TCR complementarity determining regions were extensively diversified in vivo. We show that engagement with MHC ligands during thymocyte selection and peripheral T-cell activation imposes remarkably little constraint over TCR structure. Such versatility is more consistent with an opportunist, rather than a predetermined, mode of interface formation. This hypothesis was experimentally confirmed by expressing a hybrid TCR containing TCR-γ chain germ-line complementarity determining regions, which engaged efficiently with MHC ligands.
Collapse
|
29
|
Karnowski A, Chevrier S, Belz GT, Mount A, Emslie D, D'Costa K, Tarlinton DM, Kallies A, Corcoran LM. B and T cells collaborate in antiviral responses via IL-6, IL-21, and transcriptional activator and coactivator, Oct2 and OBF-1. ACTA ACUST UNITED AC 2012; 209:2049-64. [PMID: 23045607 PMCID: PMC3478936 DOI: 10.1084/jem.20111504] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Transcriptional activator Oct2 and cofactor OBF-1 regulate B cell IL-6 to induce T cell production of IL-21, to support Tfh cell development in antiviral immunity. A strong humoral response to infection requires the collaboration of several hematopoietic cell types that communicate via antigen presentation, surface coreceptors and their ligands, and secreted factors. The proinflammatory cytokine IL-6 has been shown to promote the differentiation of activated CD4+ T cells into T follicular helper cells (TFH cells) during an immune response. TFH cells collaborate with B cells in the formation of germinal centers (GCs) during T cell–dependent antibody responses, in part through secretion of critical cytokines such as IL-21. In this study, we demonstrate that loss of either IL-6 or IL-21 has marginal effects on the generation of TFH cells and on the formation of GCs during the response to acute viral infection. However, mice lacking both IL-6 and IL-21 were unable to generate a robust TFH cell–dependent immune response. We found that IL-6 production in follicular B cells in the draining lymph node was an important early event during the antiviral response and that B cell–derived IL-6 was necessary and sufficient to induce IL-21 from CD4+ T cells in vitro and to support TFH cell development in vivo. Finally, the transcriptional activator Oct2 and its cofactor OBF-1 were identified as regulators of Il6 expression in B cells.
Collapse
Affiliation(s)
- Alex Karnowski
- Molecular Immunology Division and 2 Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Romagnoli P, Dooley J, Enault G, Vicente R, Malissen B, Liston A, van Meerwijk JPM. The Thymic Niche Does Not Limit Development of the Naturally Diverse Population of Mouse Regulatory T Lymphocytes. THE JOURNAL OF IMMUNOLOGY 2012; 189:3831-7. [DOI: 10.4049/jimmunol.1201564] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
31
|
Kaminski WE, Beham AW, Kzhyshkowska J, Gratchev A, Puellmann K. On the horizon: flexible immune recognition outside lymphocytes. Immunobiology 2012; 218:418-26. [PMID: 22749215 DOI: 10.1016/j.imbio.2012.05.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Revised: 05/25/2012] [Accepted: 05/27/2012] [Indexed: 01/13/2023]
Abstract
Since decades there is consensus among immunologists that in jawless and jawed vertebrates flexible immune recognition is strictly confined to the lymphoid lineage. In jawed vertebrates the adaptive immune system is represented by two lineages of lymphocytes, B cells and T cells that express recombinatorial antigen receptors of enormous diversity known as immunoglobulins and the T cell receptor (TCR). The recent identification of recombined immune receptors that are structurally based on the TCR in subpopulations of neutrophils and eosinophils (referred to here as TCR-like immunoreceptors, "TCRL") provides unexpected evidence for the existence of flexible host defense mechanisms beyond the realm of lymphocytes. Consistent with this, subpopulations of monocytes and macrophages from humans and mice now have also been shown to constitutively express recombined TCR-like immunoreceptors. Available in vitro evidence suggests that the TCRL in macrophages may exert functions as facilitators of phagocytosis and self-recruitment. More importantly, our recent findings that the macrophage-TCRL is implicated in granuloma formation in tuberculosis and the neutrophil-TCRL is associated with autoimmune hemolytic anemia establish for the first time a link between myeloid recombinatorial immune receptors and clinical disease. The discovery of recombined TCR-like immune receptors in granulocytes and macrophages extends the principle of combinatorial immune recognition to phagocytic cells. Conceptually, this unifies the two hitherto disparate cardinal features of innate and adaptive immunity, phagocytic capacity and recombinatorial immune recognition on a common cellular platform. Moreover, it strongly suggests that flexible host defense in vertebrates may operate on a broader cellular basis than currently thought.
Collapse
Affiliation(s)
- Wolfgang E Kaminski
- Institute for Clinical Chemistry, University of Heidelberg Medical Faculty Mannheim, Mannheim, Germany.
| | | | | | | | | |
Collapse
|
32
|
Chen Y, Ci X, Gorentla B, Sullivan SA, Stone JC, Zhang W, Pereira P, Lu J, Zhong XP. Differential requirement of RasGRP1 for γδ T cell development and activation. THE JOURNAL OF IMMUNOLOGY 2012; 189:61-71. [PMID: 22623331 DOI: 10.4049/jimmunol.1103272] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
γδ T (γδT) cells belong to a distinct T cell lineage that performs immune functions different from αβ T (αβT) cells. Previous studies established that Erk1/2 MAPKs are critical for positive selection of αβT cells. Additional evidence suggests that increased Erk1/2 activity promotes γδT cell generation. RasGRP1, a guanine nucleotide-releasing factor for Ras, plays an important role in positive selection of αβT cells by activating the Ras-Erk1/2 pathway. In this article, we demonstrate that RasGRP1 is critical for TCR-induced Erk1/2 activation in γδT cells, but it exerts different roles for γδT cell generation and activation. Deficiency of RasGRP1 does not obviously affect γδT cell numbers in the thymus, but it leads to increased γδT cells, particularly CD4(-)CD8(+) γδT cells, in the peripheral lymphoid organs. The virtually unhindered γδT cell development in the RasGRP1(-/-) thymus proved to be cell intrinsic, whereas the increase in CD8(+) γδT cells is caused by non-cell-intrinsic mechanisms. Our data provide genetic evidence that decreased Erk1/2 activation in the absence of RasGRP1 is compatible with γδT cell generation. Although RasGRP1 is dispensable for γδT cell generation, RasGRP1-deficient γδT cells are defective in proliferation following TCR stimulation. Additionally, RasGRP1-deficient γδT cells are impaired to produce IL-17 but not IFNγ. Together, these observations revealed that RasGRP1 plays differential roles for γδ and αβ T cell development but is critical for γδT cell proliferation and production of IL-17.
Collapse
Affiliation(s)
- Yong Chen
- Division of Allergy and Immunology, Department of Pediatrics, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Young GR, Ploquin MJY, Eksmond U, Wadwa M, Stoye JP, Kassiotis G. Negative selection by an endogenous retrovirus promotes a higher-avidity CD4+ T cell response to retroviral infection. PLoS Pathog 2012; 8:e1002709. [PMID: 22589728 PMCID: PMC3349761 DOI: 10.1371/journal.ppat.1002709] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Accepted: 04/04/2012] [Indexed: 11/18/2022] Open
Abstract
Effective T cell responses can decisively influence the outcome of retroviral infection. However, what constitutes protective T cell responses or determines the ability of the host to mount such responses is incompletely understood. Here we studied the requirements for development and induction of CD4+ T cells that were essential for immunity to Friend virus (FV) infection of mice, according to their TCR avidity for an FV-derived epitope. We showed that a self peptide, encoded by an endogenous retrovirus, negatively selected a significant fraction of polyclonal FV-specific CD4+ T cells and diminished the response to FV infection. Surprisingly, however, CD4+ T cell-mediated antiviral activity was fully preserved. Detailed repertoire analysis revealed that clones with low avidity for FV-derived peptides were more cross-reactive with self peptides and were consequently preferentially deleted. Negative selection of low-avidity FV-reactive CD4+ T cells was responsible for the dominance of high-avidity clones in the response to FV infection, suggesting that protection against the primary infecting virus was mediated exclusively by high-avidity CD4+ T cells. Thus, although negative selection reduced the size and cross-reactivity of the available FV-reactive naïve CD4+ T cell repertoire, it increased the overall avidity of the repertoire that responded to infection. These findings demonstrate that self proteins expressed by replication-defective endogenous retroviruses can heavily influence the formation of the TCR repertoire reactive with exogenous retroviruses and determine the avidity of the response to retroviral infection. Given the overabundance of endogenous retroviruses in the human genome, these findings also suggest that endogenous retroviral proteins, presented by products of highly polymorphic HLA alleles, may shape the human TCR repertoire that reacts with exogenous retroviruses or other infecting pathogens, leading to interindividual heterogeneity.
Collapse
Affiliation(s)
- George R. Young
- Division of Immunoregulation, MRC National Institute for Medical Research, London, United Kingdom
| | - Mickaël J.-Y. Ploquin
- Division of Immunoregulation, MRC National Institute for Medical Research, London, United Kingdom
| | - Urszula Eksmond
- Division of Immunoregulation, MRC National Institute for Medical Research, London, United Kingdom
| | - Munisch Wadwa
- Division of Immunoregulation, MRC National Institute for Medical Research, London, United Kingdom
| | - Jonathan P. Stoye
- Division of Virology, MRC National Institute for Medical Research, London, United Kingdom
| | - George Kassiotis
- Division of Immunoregulation, MRC National Institute for Medical Research, London, United Kingdom
| |
Collapse
|
34
|
Mingueneau M, Jiang W, Feuerer M, Mathis D, Benoist C. Thymic negative selection is functional in NOD mice. ACTA ACUST UNITED AC 2012; 209:623-37. [PMID: 22329992 PMCID: PMC3302233 DOI: 10.1084/jem.20112593] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Based on analyses of multiple TCR transgenic (tg) models, the emergence of pathogenic T cells in diabetes-prone NOD mice has been ascribed to a failure to censure autoreactive clones in the thymus. In contrast, using isolated and preselected thymocytes, we show that nonobese diabetic (NOD) genetic variation impairs neither clonal deletion nor downstream transcriptional programs. However, we find that NOD genetic variation influences αβ/γδ-lineage decisions promoted by early expression of tg αβ-TCRs at the double-negative (DN) stage. In B6 and other genetic backgrounds, tg αβ-TCRs behave like γδ-TCRs and commit a large fraction of DNs toward the γδ-lineage, thereby decreasing the size of the double-positive (DP) pool, which is efficiently positively and negatively selected. In NOD DNs, αβ-TCR signalosomes instead behave like pre-TCRs, resulting in high numbers of DPs competing for limited selection niches, and poor positive and negative selection. Once niche effects are neutralized in mixed bone marrow chimeras, positive and negative selection are equally efficient on B6 and NOD backgrounds. Biochemical analysis revealed a selective defect in the activation of Erk1/2 downstream of NOD αβ-TCR signalosomes. Therefore, NOD genetic variation influences αβ/γδ-lineage decisions when the αβ-TCR heterodimer is prematurely expressed, but not the process of negative selection.
Collapse
Affiliation(s)
- Michael Mingueneau
- Division of Immunology, Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
35
|
Furmanski AL, Saldana JI, Rowbotham NJ, Ross SE, Crompton T. Role of Hedgehog signalling at the transition from double-positive to single-positive thymocyte. Eur J Immunol 2011; 42:489-99. [PMID: 22101858 PMCID: PMC3378705 DOI: 10.1002/eji.201141758] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2011] [Revised: 10/12/2011] [Accepted: 11/09/2011] [Indexed: 01/28/2023]
Abstract
In the thymus, developing T cells receive signals that determine lineage choice, specificity, MHC restriction and tolerance to self-antigen. One way in which thymocytes receive instruction is by secretion of Sonic hedgehog (Shh) from thymic epithelial cells. We have previously shown that Hedgehog (Hh) signalling in the thymus decreases the CD4:CD8 single-positive (SP) thymocyte ratio. Here, we present data indicating that double-positive (DP) thymocytes are Hh-responsive and that thymocyte-intrinsic Hh signalling plays a role in modulating the production of CD4(+) (SP4), CD8(+) (SP8) and unconventional T-cell subsets. Repression of physiological Hh signalling in thymocytes altered the proportions of DP and SP4 cells. Thymocyte-intrinsic Hh-dependent transcription also attenuated both the production of mature SP4 and SP8 cells, and the establishment of peripheral T-cell compartments in TCR-transgenic mice. Additionally, stimulation or withdrawal of Hh signals in the WT foetal thymus impaired or enhanced upregulation of the CD4 lineage-specific transcription factor Gata3 respectively. These data together suggest that Hh signalling may play a role in influencing the later stages of thymocyte development.
Collapse
Affiliation(s)
- Anna L Furmanski
- Immunobiology Unit, Institute of Child Health, University College London, London, UK.
| | | | | | | | | |
Collapse
|
36
|
Ploquin MJY, Eksmond U, Kassiotis G. B cells and TCR avidity determine distinct functions of CD4+ T cells in retroviral infection. THE JOURNAL OF IMMUNOLOGY 2011; 187:3321-30. [PMID: 21841129 DOI: 10.4049/jimmunol.1101006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The T cell-dependent B cell response relies on cognate interaction between B cells and CD4(+) Th cells. However, the consequences of this interaction for CD4(+) T cells are not entirely known. B cells generally promote CD4(+) T cell responses to pathogens, albeit to a variable degree. In contrast, CD4(+) T cell responses to self- or tumor Ags are often suppressed by B cells. In this study, we demonstrated that interaction with B cells dramatically inhibited the function of virus-specific CD4(+) T cells in retroviral infection. We have used Friend virus infection of mice as a model for retroviral infection, in which the behavior of virus-specific CD4(+) T cells was monitored according to their TCR avidity. We report that avidity for Ag and interaction with B cells determine distinct aspects of the primary CD4(+) T cell response to Friend virus infection. Virus-specific CD4(+) T cells followed exclusive Th1 and T follicular helper (Tfh) differentiation. High avidity for Ag facilitated expansion during priming and enhanced the capacity for IFN-γ and IL-21 production. In contrast, Tfh differentiation was not affected by avidity for Ag. By reducing or preventing B cell interaction, we found that B cells promoted Tfh differentiation, induced programmed death 1 expression, and inhibited IFN-γ production by virus-specific CD4(+) T cells. Ultimately, B cells protected hosts from CD4(+) T cell-mediated immune pathology, at the detriment of CD4(+) T cell-mediated protective immunity. Our results suggest that B cell presentation of vaccine Ags could be manipulated to direct the appropriate CD4(+) T cell response.
Collapse
Affiliation(s)
- Mickaël J-Y Ploquin
- Division of Immunoregulation, Medical Research Council National Institute for Medical Research, The Ridgeway, London NW7 1AA, United Kingdom
| | | | | |
Collapse
|
37
|
Chassaing B, Darfeuille-Michaud A. The commensal microbiota and enteropathogens in the pathogenesis of inflammatory bowel diseases. Gastroenterology 2011; 140:1720-28. [PMID: 21530738 DOI: 10.1053/j.gastro.2011.01.054] [Citation(s) in RCA: 344] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Revised: 01/14/2011] [Accepted: 01/20/2011] [Indexed: 02/07/2023]
Abstract
Intestinal inflammation arises from abnormal host-microbe interactions. The perturbations of homeostatic coexistence involve host genetic factors, barrier function, innate and adaptive immunity, as well as qualitative and quantitative changes in the composition of the microbiota. Dysbiosis toward selected micro-organisms and decreased complexity of commensal bacteria have been observed in patients with Crohn's disease and ulcerative colitis, but it is not clear whether the dysbiosis contributes to development of inflammatory bowel disease or is instead a consequence of the disease. Pathogens with virulence factors that allow them to breach the intestinal barrier and induce chronic inflammation might mediate the pathogenesis of these diseases. To identify new therapeutic approaches for inflammatory bowel disease, it is important to identify host susceptibility factors involved in the control of microbial infection, characterize potential pathogens, and eliminate them or block the expression of their virulence factors.
Collapse
Affiliation(s)
- Benoit Chassaing
- Clermont Université, Université d'Auvergne, Jeune Equipe JE 2526, Clermont-Ferrand, France
| | | |
Collapse
|
38
|
Constitutive Notch pathway activation in murine ZMYM2-FGFR1-induced T-cell lymphomas associated with atypical myeloproliferative disease. Blood 2011; 117:6837-47. [PMID: 21527531 DOI: 10.1182/blood-2010-07-295725] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
The ZMYM2-FGFR1 (formerly known as ZNF198-FGFR1) fusion kinase induces stem cell leukemia-lymphoma syndrome (SCLL), a hematologic malignancy characterized by rapid transformation to acute myeloid leukemia and T-lymphoblastic lymphoma. In the present study, we demonstrate frequent, constitutive activation of Notch1 and its downstream target genes in T-cell lymphomas that arose in a murine model of ZMYM2-FGFR1 SCLL. Notch up-regulation was also demonstrated in human SCLL- and FGFR1OP2-FGFR1-expressing KG-1 cells. To study the role of Notch in T-cell lymphomagenesis, we developed a highly tumorigenic cell line from ZMYM2-FGFR1-expressing cells. Pharmacologic inhibition of Notch signaling in these cells using γ-secretase inhibitors significantly delayed leukemogenesis in vivo. shRNA targeting of Notch1, as well as c-promoter-binding factor 1 (CBF1) and mastermind-like 1 (MAML1), 2 essential cofactors involved in transcriptional activation of Notch target genes, also significantly delayed or inhibited tumorigenesis in vivo. Mutation analysis demonstrated that 5' promoter deletions and alternative promoter usage were responsible for constitutive activation of Notch1 in all T-cell lymphomas. These data demonstrate the importance of Notch signaling in the etiology of SCLL, and suggest that targeting this pathway could provide a novel strategy for molecular therapies to treat SCLL patients.
Collapse
|
39
|
Zhang T, Sentman CL. Cancer immunotherapy using a bispecific NK receptor fusion protein that engages both T cells and tumor cells. Cancer Res 2011; 71:2066-76. [PMID: 21282338 PMCID: PMC3095211 DOI: 10.1158/0008-5472.can-10-3200] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
T-cell immunotherapy is a promising strategy to treat cancer, but its efficacy, complexity, and costs may pose challenges. In this study, we report the results of an investigation of a new approach to selectively activate a T-cell attack against tumor cells. The immunotherapeutic approach we developed utilizes a bifunctional fusion protein that binds tumor cells through NK (natural killer)-activating receptor NKG2D and that recruits and stimulates T cells through an anti-CD3 single-chain variable fragment (scFv-NKG2D). In vitro, this scFv-NKG2D fusion protein engaged both T cells and tumor cells, stimulating T cells to produce IFN-γ, and cytotoxicity against NKG2D ligand-positive tumor cells. In vivo, expression of scFv-NKG2D by NKG2D ligand-positive tumor cells reduced tumor burden and, in some cases, led to tumor-free survival. Administration of scFv-NKG2D in vivo also promoted survival in a murine lymphoma model. Tumor-free mice were resistant to rechallenge with cognate tumor cells, suggesting that a host-specific immunologic memory response had been generated. Host adaptive immunity including γδ T cells was required for scFv-NKG2D-mediated therapeutic efficacy. ScFv-NKG2D also inhibited the growth of NKG2D ligand-negative B16F10 tumors, reduced the percentage of myeloid-derived suppressor cells and regulatory T cells, and increased the infiltration of T cells, suggesting that scFv-NKG2D may target these immune suppressive cells. Together, these results establish scFv-NKG2D as a promising biological fusion protein to induce effective antitumor immunity.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire 03756, USA.
| | | |
Collapse
|
40
|
LaBranche TP, Hickman-Brecks CL, Meyer DM, Storer CE, Jesson MI, Shevlin KM, Happa FA, Barve RA, Weiss DJ, Minnerly JC, Racz JL, Allen PM. Characterization of the KRN cell transfer model of rheumatoid arthritis (KRN-CTM), a chronic yet synchronized version of the K/BxN mouse. THE AMERICAN JOURNAL OF PATHOLOGY 2010; 177:1388-96. [PMID: 20696780 DOI: 10.2353/ajpath.2010.100195] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In this study, a chronic yet synchronized version of the K/BxN mouse, the KRN-cell transfer model (KRN-CTM), was developed and extensively characterized. The transfer of purified splenic KRN T cells into T cell-deficient B6.TCR.Calpha(-/-)H-2(b/g7) mice induced anti-glucose 6-phosphate isomerase antibody-dependent chronic arthritis in 100% of the mice with uniform onset of disease 7 days after T cell transfer. Cellular infiltrations were assessed by whole-ankle transcript microarray, cytokine and chemokine levels, and microscopic and immunohistochemical analyses 7 through 42 days after T cell transfer. Transcripts identified an influx of monocytes/macrophages and neutrophils into the ankles and identified temporal progression of cartilage damage and bone resorption. In both serum and ankle tissue there was a significant elevation in interleukin-6, whereas macrophage inflammatory protein-1 alpha and monocyte chemotactic protein-1 were only elevated in tissue. Microscopic and immunohistochemical analyses revealed a time course for edema, synovial hypertrophy and hyperplasia, infiltration of F4/80-positive monocytes/macrophages and myeloperoxidase-positive neutrophils, destruction of articular cartilage, pannus invasion, bone resorption, extra-articular fibroplasia, and joint ankylosis. The KRN cell transfer model replicates many features of chronic rheumatoid arthritis in humans in a synchronized manner and lends itself to manipulation of adoptively transferred T cells and characterizing specific genes and T cell subsets responsible for rheumatoid arthritis pathogenesis and progression.
Collapse
|
41
|
Odumade OA, Weinreich MA, Jameson SC, Hogquist KA. Krüppel-like factor 2 regulates trafficking and homeostasis of gammadelta T cells. THE JOURNAL OF IMMUNOLOGY 2010; 184:6060-6. [PMID: 20427763 DOI: 10.4049/jimmunol.1000511] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
gammadelta T cells are generated in the thymus and traffic to secondary lymphoid organs and epithelial surfaces, where they regulate immune responses. alphabeta T cells require sphingosine 1-phosphate receptor type 1 (S1P(1)) and CD62L for thymic emigration and circulation through secondary lymphoid organs. Both of these genes are regulated by the transcription factor Krüppel-like factor 2 (KLF2) in conventional alphabeta T cells. It is unclear if gammadelta T cells use similar mechanisms. In this study, we show that thymic gammadelta T cells express S1P(1) and that it is regulated by KLF2. Furthermore, KLF2 and S1P(1)-deficient gammadelta T cells accumulate in the thymus and fail to populate the secondary lymphoid organs or gut, in contrast to the expectation from published work. Interestingly, KLF2 but not S1P(1) deficiency led to the expansion of a usually rare population of CD4(+) promyelocytic leukemia zinc finger(+) "gammadelta NKT" cells. Thus, KLF2 is critically important for the homeostasis and trafficking of gammadelta T cells.
Collapse
Affiliation(s)
- Oludare A Odumade
- Center for Immunology and Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA
| | | | | | | |
Collapse
|
42
|
Marques R, Williams A, Eksmond U, Wullaert A, Killeen N, Pasparakis M, Kioussis D, Kassiotis G. Generalized immune activation as a direct result of activated CD4+ T cell killing. J Biol 2010; 8:93. [PMID: 19943952 PMCID: PMC2790834 DOI: 10.1186/jbiol194] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2009] [Revised: 10/06/2009] [Accepted: 10/07/2009] [Indexed: 01/06/2023] Open
Abstract
Background In addition to progressive CD4+ T cell immune deficiency, HIV infection is characterized by generalized immune activation, thought to arise from increased microbial exposure resulting from diminishing immunity. Results Here we report that, in a virus-free mouse model, conditional ablation of activated CD4+ T cells, the targets of immunodeficiency viruses, accelerates their turnover and produces CD4+ T cell immune deficiency. More importantly, activated CD4+ T cell killing also results in generalized immune activation, which is attributable to regulatory CD4+ T cell insufficiency and preventable by regulatory CD4+ T cell reconstitution. Immune activation in this model develops independently of microbial exposure. Furthermore, microbial translocation in mice with conditional disruption of intestinal epithelial integrity affects myeloid but not T cell homeostasis. Conclusions Although neither ablation of activated CD4+ T cells nor disruption of intestinal epithelial integrity in mice fully reproduces every aspect of HIV-associated immune dysfunction in humans, ablation of activated CD4+ T cells, but not disruption of intestinal epithelial integrity, approximates the two key immune alterations in HIV infection: CD4+ T cell immune deficiency and generalized immune activation. We therefore propose activated CD4+ T cell killing as a common etiology for both immune deficiency and activation in HIV infection. See minireview http://www.jbiol.com/content/8/10/91
Collapse
Affiliation(s)
- Rute Marques
- Division of Immunoregulation, MRC National Institute for Medical Research, The Ridgeway, London NW7 1AA, UK
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Irla M, Hollander G, Reith W. Control of central self-tolerance induction by autoreactive CD4+ thymocytes. Trends Immunol 2010; 31:71-9. [DOI: 10.1016/j.it.2009.11.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2009] [Revised: 11/05/2009] [Accepted: 11/10/2009] [Indexed: 01/08/2023]
|
44
|
Perona-Wright G, Mohrs K, Mayer KD, Mohrs M. Differential regulation of IL-4Ralpha expression by antigen versus cytokine stimulation characterizes Th2 progression in vivo. THE JOURNAL OF IMMUNOLOGY 2009; 184:615-23. [PMID: 20018622 DOI: 10.4049/jimmunol.0902408] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
IL-4 promotes Th2 differentiation and provides immunity to helminth infections but is also associated with allergy and asthma. This suggests that precise adjustment of IL-4 responsiveness is needed to correctly balance immune responses. The IL-4Ralpha chain is an essential component of the IL-4 receptor and signals via STAT6. In this study, we show that infection with a helminth pathogen elicited broad upregulation of IL-4Ralpha on bystander CD4+ T cells in the draining lymph node, while simultaneously resulting in the loss of IL-4Ralpha expression on activated Th2 cells. IL-4Ralpha upregulation was restricted to the reactive lymph node, occurred within 4 d of infection, and was driven by an IL-4- and STAT6-dependent mechanism. Mice heterozygous for Stat6 exhibited reduced IL-4Ralpha upregulation and a correspondingly attenuated Th2 response. Indeed, the enhanced IL-4Ralpha upregulation in BALB/c mice, compared with that in C57BL6 mice, predicted their stronger Th2 response. The selective downregulation of IL-4Ralpha on highly activated Th cells was triggered by antigenic stimulation, was accompanied by loss of IL-7Ralpha, and rendered the cells unresponsive to IL-4. Together these data reveal a tightly controlled program of changing IL-4 responsiveness that characterizes the initiation, amplification, and restriction of a Th2 response in vivo.
Collapse
|
45
|
Furmanski AL, Bartok I, Chai JG, Singh Y, Ferreira C, Scott D, Holland SJ, Bourdeaux C, Crompton T, Dyson J. Peptide-specific, TCR-alpha-driven, coreceptor-independent negative selection in TCR alpha-chain transgenic mice. THE JOURNAL OF IMMUNOLOGY 2009; 184:650-7. [PMID: 19995903 DOI: 10.4049/jimmunol.0902291] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
As thymocytes differentiate, Ag sensitivity declines, with immature CD4-CD8- double-negative (DN) cells being most susceptible to TCR signaling events. We show that expression of alphabetaTCR from the DN3 stage lowers the threshold for activation, allowing recognition of MHC peptides independently of the TCR beta-chain and without either T cell coreceptor. The MHC class I-restricted C6 TCR recognizes the Y-chromosome-derived Ag HYK(k)Smcy. Positive selection in C6 alphabetaTCR females is skewed to the CD8 compartment, whereas transgenic male mice exhibit early clonal deletion of thymocytes. We investigated the effect of the HYK(k)Smcy complex on developing thymocytes expressing the C6 TCR alpha-chain on a TCR-alpha(-/-) background. On the original selecting haplotype, the skew to the CD8 lineage is preserved. This is MHC dependent, as the normal bias to the CD4 subset is seen on an H2b background. In male H2k C6 alpha-only mice, the presence of the HYK(k)Smcy complex leads to a substantial deletion of thymocytes from the DN subset. This phenotype is replicated in H2k C6 alpha-only female mice expressing an Smcy transgene. Deletion is not dependent on the beta variable segment of the C6 TCR or on a restricted TCR-beta repertoire. In contrast, binding of HYK(k)Smcy and Ag-specific activation of mature CD8+ T cells is strictly dependent on the original C6 beta-chain. These data demonstrate that, in comparison with mature T cells, alphabetaTCR+ immature thymocytes can recognize and transduce signals in response to specific MHC-peptide complexes with relaxed binding requirements.
Collapse
Affiliation(s)
- Anna L Furmanski
- Department of Immunology, Hammersmith Hospital, Imperial College London, London, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Wong FS, Siew LK, Scott G, Thomas IJ, Chapman S, Viret C, Wen L. Activation of insulin-reactive CD8 T-cells for development of autoimmune diabetes. Diabetes 2009; 58:1156-64. [PMID: 19208910 PMCID: PMC2671054 DOI: 10.2337/db08-0800] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
OBJECTIVE We have previously reported a highly diabetogenic CD8 T-cell clone, G9C8, in the nonobese diabetic (NOD) mouse, specific to low-avidity insulin peptide B15-23, and cells responsive to this antigen are among the earliest islet infiltrates. We aimed to study the selection, activation, and development of the diabetogenic capacity of these insulin-reactive T-cells. RESEARCH DESIGN AND METHODS We generated a T-cell receptor (TCR) transgenic mouse expressing the cloned TCR Valpha18/Vbeta6 receptor of the G9C8 insulin-reactive CD8 T-cell clone. The mice were crossed to TCRCalpha-/- mice so that the majority of the T-cells expressed the clonotypic TCR, and the phenotype and function of the cells was investigated. RESULTS There was good selection of CD8 T-cells with a predominance of CD8 single-positive thymocytes, in spite of thymic insulin expression. Peripheral lymph node T-cells had a naïve phenotype (CD44lo, CD62Lhi) and proliferated to insulin B15-23 peptide and to insulin. These cells produced interferon-gamma and tumor necrosis factor-alpha in response to insulin peptide and were cytotoxic to insulin peptide-coated targets. In vivo, the TCR transgenic mice developed insulitis but not spontaneous diabetes. However, the mice developed diabetes on immunization, and the activated transgenic T-cells were able to transfer diabetes to immunodeficient NOD.scid mice. CONCLUSIONS Autoimmune CD8 T-cells responding to a low-affinity insulin B-chain peptide escape from thymic negative selection and require activation in vivo to cause diabetes.
Collapse
Affiliation(s)
- F Susan Wong
- Department of Cellular and Molecular Medicine, University of Bristol, Bristol, UK.
| | | | | | | | | | | | | |
Collapse
|
47
|
Masuda K, Germeraad WTV, Satoh R, Itoi M, Ikawa T, Minato N, Katsura Y, van Ewijk W, Kawamoto H. Notch activation in thymic epithelial cells induces development of thymic microenvironments. Mol Immunol 2009; 46:1756-67. [PMID: 19250680 DOI: 10.1016/j.molimm.2009.01.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 01/25/2009] [Indexed: 01/18/2023]
Abstract
The development and maintenance of thymic microenvironments depends on sustained crosstalk signals derived from developing thymocytes. However, the molecular basis for the initial phase in the lymphoid dependent development of thymic epithelial cells (TECs) remains unclear. Here we show that similarly to regular thymocytes, developing B cells enforced to express the Notch ligand Delta-like-1 (DLL1) efficiently induce the non-polarized, three-dimensional (3D) meshwork architecture of cortical TECs in fetal thymic organ culture. Moreover, the DLL1-overexpressing B cells induce well-developed distinct medullae. Such medullae also arose in lobes reconstituted with Rag2(-/-) thymocytes overexpressing DLL1. Our present findings thus strongly suggest that Notch signaling from thymocytes to TECs induces TEC development at an early phase of thymic organogenesis. The present approach using non-T lineage cells for the in vitro construction of thymic environments may also provide a novel tool for thymus regeneration and T cell production in immunocompromised individuals.
Collapse
Affiliation(s)
- Kyoko Masuda
- Laboratory for Lymphocyte Development, RIKEN Research Center for Allergy and Immunology, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Retrovirus-specificity of regulatory T cells is neither present nor required in preventing retrovirus-induced bone marrow immune pathology. Immunity 2008; 29:782-94. [PMID: 19006695 PMCID: PMC2631611 DOI: 10.1016/j.immuni.2008.09.016] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Revised: 08/06/2008] [Accepted: 09/15/2008] [Indexed: 01/19/2023]
Abstract
Chronic viral infections of the hematopoietic system are associated with bone marrow dysfunction, to which both virus-mediated and immune-mediated effects may contribute. Using unresolving noncytopathic Friend virus (FV) infection in mice, we showed that unregulated CD4+ T cell response to FV caused IFN-γ-mediated bone marrow pathology and anemia. Importantly, bone marrow pathology was triggered by relative insufficiency in regulatory T (Treg) cells and was prevented by added Treg cells, which suppressed the local IFN-γ production by FV-specific CD4+ T cells. We further showed that the T cell receptor (TCR) repertoire of transgenic Treg cells expressing the β chain of an FV-specific TCR was virtually devoid of FV-specific clones. Moreover, anemia induction by virus-specific CD4+ T cells was efficiently suppressed by virus-nonspecific Treg cells. Thus, sufficient numbers of polyclonal Treg cells may provide substantial protection against bone marrow pathology in chronic viral infections.
Collapse
|
49
|
Hikosaka Y, Nitta T, Ohigashi I, Yano K, Ishimaru N, Hayashi Y, Matsumoto M, Matsuo K, Penninger JM, Takayanagi H, Yokota Y, Yamada H, Yoshikai Y, Inoue JI, Akiyama T, Takahama Y. The cytokine RANKL produced by positively selected thymocytes fosters medullary thymic epithelial cells that express autoimmune regulator. Immunity 2008; 29:438-50. [PMID: 18799150 DOI: 10.1016/j.immuni.2008.06.018] [Citation(s) in RCA: 333] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2007] [Revised: 04/08/2008] [Accepted: 06/20/2008] [Indexed: 12/15/2022]
Abstract
The thymic medulla provides a microenvironment where medullary thymic epithelial cells (mTECs) express autoimmune regulator and diverse tissue-restricted genes, contributing to launching self-tolerance. Positive selection is essential for thymic medulla formation via a previously unknown mechanism. Here we show that the cytokine RANK ligand (RANKL) was produced by positively selected thymocytes and regulated the cellularity of mTEC by interacting with RANK and osteoprotegerin. Forced expression of RANKL restored thymic medulla in mice lacking positive selection, whereas RANKL perturbation impaired medulla formation. These results indicate that RANKL produced by positively selected thymocytes is responsible for fostering thymic medulla formation, thereby establishing central tolerance.
Collapse
Affiliation(s)
- Yu Hikosaka
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Protein phosphatase subunit G5PR that regulates the JNK-mediated apoptosis signal is essential for the survival of CD4 and CD8 double-positive thymocytes. Mol Immunol 2008; 45:2028-37. [DOI: 10.1016/j.molimm.2007.10.028] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2007] [Revised: 10/11/2007] [Accepted: 10/12/2007] [Indexed: 11/22/2022]
|