1
|
Honan AM, Vazquez EN, Chen Z. Lymph Node Stromal Cell-Intrinsic MHC Class II Expression Promotes MHC Class I-Restricted CD8 T Cell Lineage Conversion to Regulatory CD4 T Cells. THE JOURNAL OF IMMUNOLOGY 2021; 207:1530-1544. [PMID: 34408011 DOI: 10.4049/jimmunol.2100396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/07/2021] [Indexed: 12/29/2022]
Abstract
MHC class I (MHC-I)-restricted CD4+ T cells have long been discovered in the natural repertoire of healthy humans as well as patients with autoimmune diseases or cancer, but the exact origin of these cells remains to be fully characterized. In mouse models, mature peripheral CD8+ T cells have the potential to convert to CD4+ T cells in the mesenteric lymph nodes. This conversion can produce a unique population of MHC-I-restricted CD4+ T cells including Foxp3+ regulatory T cells termed MHC-I-restricted CD4+Foxp3+ T (CI-Treg) cells. In this study we examined the cellular and molecular elements that promote CD8-to-CD4 lineage conversion and the development of CI-Treg cells in mice. Using adoptive transfer and bone marrow chimera experiments, we found that the differentiation of CI-Treg cells was driven by lymph node stromal cell (LNSC)-intrinsic MHC-II expression as opposed to transcytosis of MHC-II from bone marrow-derived APCs. The lineage conversion was accompanied by Runx3 versus ThPOK transcriptional switch. This finding of a new role for LNSCs in vivo led us to develop an efficient tissue culture method using LNSCs to generate and expand CI-Treg cells in vitro. CI-Treg cells expanded in vitro with LNSCs effectively suppressed inflammatory tissue damage caused by pathogenic CD4+ T cells in mouse models of colitis. This study identified a novel role of MHC-II expressed by LNSCs in immune regulation and the potential utilization of LNSCs to generate novel subsets of immune regulatory cells for therapeutic applications.
Collapse
Affiliation(s)
- Amanda M Honan
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL; and
| | - Emily N Vazquez
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL; and
| | - Zhibin Chen
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL; and .,Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| |
Collapse
|
2
|
Maccari ME, Fuchs S, Kury P, Andrieux G, Völkl S, Bengsch B, Lorenz MR, Heeg M, Rohr J, Jägle S, Castro CN, Groß M, Warthorst U, König C, Fuchs I, Speckmann C, Thalhammer J, Kapp FG, Seidel MG, Dückers G, Schönberger S, Schütz C, Führer M, Kobbe R, Holzinger D, Klemann C, Smisek P, Owens S, Horneff G, Kolb R, Naumann-Bartsch N, Miano M, Staniek J, Rizzi M, Kalina T, Schneider P, Erxleben A, Backofen R, Ekici A, Niemeyer CM, Warnatz K, Grimbacher B, Eibel H, Mackensen A, Frei AP, Schwarz K, Boerries M, Ehl S, Rensing-Ehl A. A distinct CD38+CD45RA+ population of CD4+, CD8+, and double-negative T cells is controlled by FAS. J Exp Med 2021; 218:211525. [PMID: 33170215 PMCID: PMC7658692 DOI: 10.1084/jem.20192191] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 08/06/2020] [Accepted: 10/09/2020] [Indexed: 12/16/2022] Open
Abstract
The identification and characterization of rare immune cell populations in humans can be facilitated by their growth advantage in the context of specific genetic diseases. Here, we use autoimmune lymphoproliferative syndrome to identify a population of FAS-controlled TCRαβ+ T cells. They include CD4+, CD8+, and double-negative T cells and can be defined by a CD38+CD45RA+T-BET− expression pattern. These unconventional T cells are present in healthy individuals, are generated before birth, are enriched in lymphoid tissue, and do not expand during acute viral infection. They are characterized by a unique molecular signature that is unambiguously different from other known T cell differentiation subsets and independent of CD4 or CD8 expression. Functionally, FAS-controlled T cells represent highly proliferative, noncytotoxic T cells with an IL-10 cytokine bias. Mechanistically, regulation of this physiological population is mediated by FAS and CTLA4 signaling, and its survival is enhanced by mTOR and STAT3 signals. Genetic alterations in these pathways result in expansion of FAS-controlled T cells, which can cause significant lymphoproliferative disease.
Collapse
Affiliation(s)
- Maria Elena Maccari
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sebastian Fuchs
- Roche Pharma Research and Early Development, Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Innovation Center Basel, Basel, Switzerland
| | - Patrick Kury
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Geoffroy Andrieux
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Freiburg, and German Cancer Research Center, Heidelberg, Germany
| | - Simon Völkl
- Department of Internal Medicine 5-Hematology/Oncology, University of Erlangen, Erlangen, Germany
| | - Bertram Bengsch
- Department of Medicine II, Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Integrative Biological Signaling Studies, Albert-Ludwigs University, Freiburg, Germany.,Bioss Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany
| | - Myriam Ricarda Lorenz
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany.,Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Wuerttemberg-Hessen, Ulm, Germany
| | - Maximilian Heeg
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Jan Rohr
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sabine Jägle
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carla N Castro
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Miriam Groß
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ursula Warthorst
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christoph König
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Ilka Fuchs
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carsten Speckmann
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Julian Thalhammer
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Friedrich G Kapp
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Markus G Seidel
- Division of Pediatric Hematology-Oncology, Department of Pediatrics and Adolescent Medicine, Medical University Graz, Graz, Austria
| | - Gregor Dückers
- Helios Kliniken Krefeld, Children's Hospital, Krefeld, Germany
| | - Stefan Schönberger
- University of Bonn, Department of Paediatric Haematology and Oncology, University Children's Hospital Bonn, Germany
| | - Catharina Schütz
- Department of Pediatrics, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Marita Führer
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany.,Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Wuerttemberg-Hessen, Ulm, Germany
| | - Robin Kobbe
- First Department of Medicine, Division of Infectious Diseases, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dirk Holzinger
- Department of Pediatric Hematology-Oncology, University of Duisburg-Essen, Essen, Germany
| | - Christian Klemann
- Department of Pediatric Pulmonology, Allergy and Neonatology, Hannover Medical School, Hannover, Germany
| | - Petr Smisek
- Department of Pediatric Hematology and Oncology, University Hospital Motol and Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Stephen Owens
- Great North Children's Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.,Institute of Health and Society, Newcastle University, Newcastle upon Tyne, UK
| | - Gerd Horneff
- Department of General Paediatrics, Clinic Sankt Augustin, Sankt Augustin, Germany.,Department of Pediatric and Adolescent Medicine, University Hospital of Cologne, Cologne, Germany
| | - Reinhard Kolb
- Department of General Paediatrics, Clinic Oldenburg, Oldenburg, Germany
| | - Nora Naumann-Bartsch
- Department of Pediatrics, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Maurizio Miano
- Haematology Unit, Istituto di Ricovero e Cura a Carattere Scientifico Istituto Giannina Gaslini, Genoa, Italy
| | - Julian Staniek
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Department of Rheumatology and Clinical Immunology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Marta Rizzi
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Department of Rheumatology and Clinical Immunology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tomas Kalina
- Childhood Leukemia Investigation Prague, Department of Pediatric Hematology and Oncology, Second Medical School, Charles University and University Hospital Motol, Prague, Czech Republic
| | - Pascal Schneider
- Department of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | - Anika Erxleben
- Bioinformatics, Institute for Computer Science, Faculty of Engineering, University of Freiburg, Germany
| | - Rolf Backofen
- Bioinformatics, Institute for Computer Science, Faculty of Engineering, University of Freiburg, Germany
| | - Arif Ekici
- Institute of Human Genetics, University of Erlangen, Erlangen, Germany
| | - Charlotte M Niemeyer
- Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus Warnatz
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bodo Grimbacher
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Integrative Biological Signaling Studies, Albert-Ludwigs University, Freiburg, Germany.,German Center for Infection Research, Satellite Center, Freiburg, Germany.,Resolving Infection Susceptibility Cluster of Excellence 2155, Hanover Medical School, Satellite Center, Freiburg, Germany
| | - Hermann Eibel
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Andreas Mackensen
- Department of Internal Medicine 5-Hematology/Oncology, University of Erlangen, Erlangen, Germany
| | - Andreas Philipp Frei
- Roche Pharma Research and Early Development, Immunology, Infectious Diseases and Ophthalmology (I2O) Discovery and Translational Area, Roche Innovation Center Basel, Basel, Switzerland
| | - Klaus Schwarz
- Institute for Transfusion Medicine, University of Ulm, Ulm, Germany.,Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Wuerttemberg-Hessen, Ulm, Germany
| | - Melanie Boerries
- Institute of Medical Bioinformatics and Systems Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,German Cancer Consortium, Freiburg, and German Cancer Research Center, Heidelberg, Germany
| | - Stephan Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Division of Pediatric Hematology and Oncology, Department of Pediatrics and Adolescent Medicine, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.,Center for Integrative Biological Signaling Studies, Albert-Ludwigs University, Freiburg, Germany
| | - Anne Rensing-Ehl
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
3
|
Akiyama C, Tsumiyama K, Uchimura C, Honda E, Miyazaki Y, Sakurai K, Miura Y, Hashiramoto A, Felsher DW, Shiozawa S. Conditional Upregulation of IFN-α Alone Is Sufficient to Induce Systemic Lupus Erythematosus. THE JOURNAL OF IMMUNOLOGY 2019; 203:835-843. [PMID: 31324723 DOI: 10.4049/jimmunol.1801617] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 06/17/2019] [Indexed: 11/19/2022]
Abstract
The cause of systemic lupus erythematosus (SLE) is unknown. IFN-α has been suggested as a causative agent of SLE; however, it was not proven, and to what extent and how IFN-α contributes to the disease is unknown. We studied the contribution of IFN-α to SLE by generating inducible IFN-α transgenic mice and directly show that conditional upregulation of IFN-α alone induces a typical manifestation of SLE in the mice not prone to autoimmunity, such as serum immune complex, autoantibody against dsDNA (anti-dsDNA Ab), and the organ manifestations classical to SLE, such as immune complex-deposited glomerulonephritis, classical splenic onion-skin lesion, alopecia, epidermal liquefaction, and positive lupus band test of the skin. In the spleen of mice, activated effector CD4 T cells, IFN-γ-producing CD8 T cells, B220+CD86+ cells, and CD11c+CD86+ cells were increased, and the T cells produced increased amounts of IL-4, IL-6, IL-17, and IFN-γ and decreased IL-2. In particular, activated CD3+CD4-CD8- double-negative T cells positive for TCRαβ, B220, CD1d-teteramer, PD-1, and Helios (that produced increased amounts of IFN-γ, IL-4, IL-17, and TNF-α) were significantly expanded. They infiltrated into kidney and induced de novo glomerulonephritis and alopecia when transferred into naive recipients. Thus, sole upregulation of IFN-α is sufficient to induce SLE, and the double-negative T cells expanded by IFN-α are directly responsible for the organ manifestations, such as lupus skin disease or nephritis.
Collapse
Affiliation(s)
- Chieri Akiyama
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Ken Tsumiyama
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan.,Institute for Rheumatic Diseases, Ashiya 659-0004, Japan.,Kyushu University Beppu Hospital, Beppu 874-0838, Japan; and
| | - Chiaki Uchimura
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Eriko Honda
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Yumi Miyazaki
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Keiichi Sakurai
- Institute for Rheumatic Diseases, Ashiya 659-0004, Japan.,Kyushu University Beppu Hospital, Beppu 874-0838, Japan; and
| | - Yasushi Miura
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Akira Hashiramoto
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan
| | - Dean W Felsher
- Division of Oncology, Department of Medicine and Pathology, School of Medicine, Stanford University, Stanford, CA 94305
| | - Shunichi Shiozawa
- Department of Biophysics, Kobe University Graduate School of Health Science, Kobe 654-0142, Japan; .,Institute for Rheumatic Diseases, Ashiya 659-0004, Japan.,Kyushu University Beppu Hospital, Beppu 874-0838, Japan; and
| |
Collapse
|
4
|
Rodríguez-Rodríguez N, Apostolidis SA, Fitzgerald L, Meehan BS, Corbett AJ, Manuel Martín-Villa J, McCluskey J, Tsokos GC, Crispín JC. Pro-inflammatory self-reactive T cells are found within murine TCR-αβ(+) CD4(-) CD8(-) PD-1(+) cells. Eur J Immunol 2016; 46:1383-1391. [PMID: 27060346 PMCID: PMC4913481 DOI: 10.1002/eji.201546056] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 02/23/2016] [Accepted: 03/30/2016] [Indexed: 11/07/2022]
Abstract
TCR-αβ(+) double negative (DN) T cells (CD3(+) TCR-αβ(+) CD4(-) CD8(-) NK1.1(-) CD49b(-) ) represent a minor heterogeneous population in healthy humans and mice. These cells have been ascribed pro-inflammatory and regulatory capacities and are known to expand during the course of several autoimmune diseases. Importantly, previous studies have shown that self-reactive CD8(+) T cells become DN after activation by self-antigens, suggesting that self-reactive T cells may exist within the DN T-cell population. Here, we demonstrate that programmed cell death 1 (PD-1) expression in unmanipulated mice identifies a subset of DN T cells with expression of activation-associated markers and a phenotype that strongly suggests they are derived from self-reactive CD8(+) cells. We also found that, within DN T cells, the PD-1(+) subset generates the majority of pro-inflammatory cytokines. Finally, using a TCR-activation reporter mouse (Nur77-GFP), we confirmed that in the steady-state PD-1(+) DN T cells engage endogenous antigens in healthy mice. In conclusion, we provide evidence that indicates that the PD-1(+) fraction of DN T cells represents self-reactive cells.
Collapse
Affiliation(s)
- Noé Rodríguez-Rodríguez
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sokratis A. Apostolidis
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Lauren Fitzgerald
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Bronwyn S. Meehan
- The Department of Microbiology and Immunology, The University of Melbourne and The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Alexandra J. Corbett
- The Department of Microbiology and Immunology, The University of Melbourne and The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - José Manuel Martín-Villa
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| | - James McCluskey
- The Department of Microbiology and Immunology, The University of Melbourne and The Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - George C. Tsokos
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - José C. Crispín
- Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Departamento de Inmunología y Reumatología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| |
Collapse
|
5
|
Rodríguez-Rodríguez N, Apostolidis SA, Penaloza-MacMaster P, Martín Villa JM, Barouch DH, Tsokos GC, Crispín JC. Programmed cell death 1 and Helios distinguish TCR-αβ+ double-negative (CD4-CD8-) T cells that derive from self-reactive CD8 T cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2015; 194:4207-4214. [PMID: 25825451 PMCID: PMC4503929 DOI: 10.4049/jimmunol.1402775] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 03/01/2015] [Indexed: 01/20/2023]
Abstract
TCR-αβ(+) double-negative (DN; CD4(-)CD8(-)) T cells represent a poorly understood cellular subset suggested to contribute to the pathogenesis of the autoimmune disease systemic lupus erythematosus. DN T cells have been proposed to derive from CD8(+) cells. However, the conditions that govern the loss of CD8 expression after Ag encounter are unknown. In this study, we tracked the fate of CD8 T cells from transgenic TCR mice exposed to their cognate Ags as self or in the context of infection. We demonstrate that CD8 T cells lose CD8 expression and become DN only when cognate Ag is sensed as self. This process is restricted to tissues where the Ag is present. We also show that DN T cells derived from self-reactive CD8 cells express the inhibitory molecules PD-1 and Helios. These molecules identify a subset of DN T cells in normal mice. A similar population expands when CD8 T cells from repertoires enriched in self-reactive cells (Aire-deficient) are transferred into cognate hosts. Collectively, our data suggest that a subset of DN T cells, identified by the expression of PD-1 and Helios, represent self-reactive cells. Our results provide an explanation for the origin of DN T cells and introduce CD8 loss as a process associated with self-Ag encounter.
Collapse
Affiliation(s)
- Noé Rodríguez-Rodríguez
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215; Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Sokratis A Apostolidis
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215
| | - Pablo Penaloza-MacMaster
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and
| | - José Manuel Martín Villa
- Departamento de Inmunología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, 28040, Spain
| | - Dan H Barouch
- Center for Virology and Vaccine Research, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215; and Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139
| | - George C Tsokos
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215;
| | - José C Crispín
- Division of Rheumatology, Department of Medicine, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA 02215;
| |
Collapse
|
6
|
Double negative (DN) αβ T cells: misperception and overdue recognition. Immunol Cell Biol 2014; 93:305-10. [PMID: 25420721 DOI: 10.1038/icb.2014.99] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 10/14/2014] [Accepted: 10/15/2014] [Indexed: 12/24/2022]
Abstract
CD4(-)CD8(-)double negative (DN) αβ T cells are legitimate components of the normal immune system. However, they are poorly understood and largely ignored by immunologists because of their historical association with the lymphoproliferation that occurs in mice (lpr and gld) and humans (autoimmune lymphoproliferative syndromes patients) with impaired Fas-mediated apoptosis where they are considered abnormal T cells. We believe that the traditional view that DN T cells that cause lymphoproliferation (hereafter referred to as lpr DN T cells) are CD4 and CD8 T cells that lost their coreceptor, conceived more than two decades ago, is flawed and that conflating lpr DN T cells with DN T cells found in normal immune system (hereafter referred to as nDN T cells) is unnecessarily dampening interest of this potentially important cell type. To begin rectifying these misperceptions, we will revisit the traditional view of lpr DN T cells and show that it does not hold true in light of recent immunological advances. In lieu of it, we offer a new model proposing that Fas-mediated apoptosis actively removes normally existing DN T cells from the periphery and that impaired Fas-mediated apoptosis leads to accumulation of these cells rather than de novo generation of DN T cells from activated CD4 or CD8 T cells. By doing so, we hope to provoke a new discussion that may lead to a consensus about the origin of lpr DN T cells and regulation of their homeostasis by the Fas pathway and reignite wider interest in nDN T cells.
Collapse
|
7
|
Abnormally differentiated CD4+ or CD8+ T cells with phenotypic and genetic features of double negative T cells in human Fas deficiency. Blood 2014; 124:851-60. [DOI: 10.1182/blood-2014-03-564286] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Key Points
Lack of KLRG1 and T-bet expression is a unique feature of DNT and subsets of single positive T cells in ALPS patients. Genetic, phenotypic, and transcriptional evidence indicates that DNT in ALPS patients derive from both CD4+ and CD8+ T cells.
Collapse
|
8
|
DNA methylation and regulation of the CD8A after duck hepatitis virus type 1 infection. PLoS One 2014; 9:e88023. [PMID: 24505360 PMCID: PMC3913717 DOI: 10.1371/journal.pone.0088023] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 01/02/2014] [Indexed: 11/19/2022] Open
Abstract
Background Cluster of differentiation 8 (CD8) is expressed in cytotoxic T cells, where it functions as a co-receptor for the T-cell receptor by binding to major histocompatibility complex class I (MHCI) proteins, which present peptides on the cell surface. CD8A is critical for cell-mediated immune defense and T-cell development. CD8A transcription is controlled by several cis-acting elements and trans-acting elements and is also regulated by DNA methylation. However, the epigenetic regulation of CD8A in the duck and its relationship with virus infection are still unclear. Results We investigated the epigenetic transcriptional regulatory mechanisms, such as DNA methylation, for the expression of the CD8A and further evaluated the contribution of such epigenetic regulatory mechanisms to DHV-I infection in the duck. Real-time quantitative polymerase chain reaction (RT-qPCR) revealed the highest level of CD8A expression to be in the thymus, followed by the lungs, spleen, and liver, and the levels of CD8A expression were very low in the kidney, cerebrum, cerebellum, and muscle in the duck. RT-qPCR also demonstrated that the CD8A mRNA was down-regulated significantly in morbid ducklings treated with DHV-1 and up-regulated significantly in non-morbid ducklings in all the tissues tested. In addition, hypermethylation of CD8A was detected in the morbid ducklings, whereas relatively low methylation of CD8A was evident in the non-morbid ducklings. The CD8A mRNA level was negatively associated with the CpG methylation level of CD8A and global methylation status. Conclusions We concluded that the mRNA level of the CD8A was negatively associated with the CpG methylation level of CD8A and global methylation status in the duck, suggesting that the hypermethylation of CD8A may be associated with DHV-1 infection. The first two CpG sites of the CD8A promoter region could be considered as epigenetic biomarkers for resistance breeding against duckling hepatitis disease in the duck.
Collapse
|
9
|
Olver S, Apte SH, Baz A, Kelso A, Kienzle N. Interleukin-4-induced loss of CD8 expression and cytolytic function in effector CD8 T cells persists long term in vivo. Immunology 2013; 139:187-96. [PMID: 23311920 DOI: 10.1111/imm.12068] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2012] [Revised: 01/07/2013] [Accepted: 01/08/2013] [Indexed: 01/28/2023] Open
Abstract
Activation of naive CD8(+) T cells in the presence of interleukin-4 modulates their CD8 co-receptor expression and functional differentiation, resulting in the generation of CD8(low) cells that produce type 2 cytokines and display poor cytolytic and anti-tumour activity. Although this CD8(low) phenotype becomes stable after about a week and can persist with further stimulation in vitro, it is not known whether it can be maintained long term in vivo. Here we report that CD8(low) cells derived from oval-bumin(257-264) -specific T-cell receptor-transgenic CD8(+) T cells activated in the presence of interleukin-4 could be detected in the spleen for at least 4 months after adoptive transfer into normal mice. A significant proportion of the long-term surviving cells retained their CD8(low) phenotype in vivo and after clonal re-activation in vitro. Although long-term surviving CD8(low) cells lacked detectable cytolytic activity or perforin expression, they showed some anti-tumour function in vivo. The persistence of functional cells with a CD8(low) phenotype in vivo raises the possibility that such cells can contribute to effector or regulatory responses to tumours or pathogens.
Collapse
Affiliation(s)
- Stuart Olver
- The Cooperative Research Centre for Vaccine Technology and the Queensland Institute of Medical Research, Herston, Qld, Australia
| | | | | | | | | |
Collapse
|
10
|
Alli R, Nguyen P, Geiger TL. Altered differentiation, diminished pathogenicity, and regulatory activity of myelin-specific T cells expressing an enhanced affinity TCR. THE JOURNAL OF IMMUNOLOGY 2011; 187:5521-31. [PMID: 22025553 DOI: 10.4049/jimmunol.1102202] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Whereas increased affinity enhances T cell competitiveness after immunization, the role of affinity in modulating the pathogenicity of self-reactive T cells is less established. To assess this, we generated two myelin-specific, class II MHC-restricted TCR that differ only in a buried hydroxymethyl that forms a common TCR β-chain V region variant. The variation, predicted to increase TCR stability, resulted in a ~3log(10) difference in TCR sensitivity with preserved fine specificity. The high-affinity TCR markedly diminished T cell pathogenicity. T cells were not deleted, did not upregulate Foxp3, and barring disease induction were predominantly naive. However, high-affinity CD4(+) T cells showed an altered cytokine profile characterized by the production of protective cytokines prior to experimental allergic encephalomyelitis induction and decreased effector cytokines after. Further, the high-affinity TCR promoted the development of CD4(-)CD8(-) and CD8(+) T cells that possessed low intrinsic pathogenicity, were protective even in small numbers when transferred into wild-type mice and in mixed chimeras, and outcompete CD4(+) T cells during disease development. Therefore, TCR affinities exceeding an upper affinity threshold may impede the development of autoimmunity through altered development and functional maturation of T cells, including diminished intrinsic CD4(+) T cell pathogenicity and the development of CD4(-)Foxp3(-) regulatory populations.
Collapse
Affiliation(s)
- Rajshekhar Alli
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN 38105, USA
| | | | | |
Collapse
|
11
|
Fortner KA, Lees RK, MacDonald HR, Budd RC. Fas (CD95/APO-1) limits the expansion of T lymphocytes in an environment of limited T-cell antigen receptor/MHC contacts. Int Immunol 2011; 23:75-88. [PMID: 21266499 DOI: 10.1093/intimm/dxq466] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Fas-deficient mice (Fas(lpr/lpr)) and humans have profoundly dysregulated T lymphocyte homeostasis, which manifests as an accumulation of CD4(+) and CD8(+) T cells as well as an unusual population of CD4(-)CD8(-)TCRαβ(+) T cells. To date, no unifying model has explained both the increased T-cell numbers and the origin of the CD4(-)CD8(-)TCRαβ(+) T cells. As Fas(lpr/lpr) mice raised in a germ-free environment still manifest lymphadenopathy, we considered that this process is primarily driven by recurrent low-avidity TCR signaling in response to self-peptide/MHC as occurs during homeostatic proliferation. In these studies, we developed two independent systems to decrease the number of self-peptide/MHC contacts. First, expression of MHC class I was reduced in OT-I TCR transgenic mice. Although OT-I Fas(lpr/lpr) mice did not develop lymphadenopathy characteristic of Fas(lpr/lpr) mice, in the absence of MHC class I, OT-I Fas(lpr/lpr) T cells accumulated as both CD8(+) and CD4(-)CD8(-) T cells. In the second system, re-expression of β(2)m limited to thymic cortical epithelial cells of Fas(lpr/lpr) β(2)m-deficient mice yielded a model in which polyclonal CD8(+) thymocytes entered a peripheral environment devoid of MHC class I. These mice accumulated significantly greater numbers of CD4(-)CD8(-)TCRαβ(+) T cells than conventional Fas(lpr/lpr) mice. Thus, Fas shapes the peripheral T-cell repertoire by regulating the survival of a subset of T cells proliferating in response to limited self-peptide/MHC contacts.
Collapse
Affiliation(s)
- Karen A Fortner
- Immunobiology Program, Department of Medicine, The University of Vermont College of Medicine, Burlington, VT 05405-0068, USA.
| | | | | | | |
Collapse
|
12
|
Kinjyo I, Gordon SM, Intlekofer AM, Dowdell K, Mooney EC, Caricchio R, Grupp SA, Teachey DT, Rao VK, Lindsten T, Reiner SL. Cutting edge: Lymphoproliferation caused by Fas deficiency is dependent on the transcription factor eomesodermin. THE JOURNAL OF IMMUNOLOGY 2010; 185:7151-5. [PMID: 21076068 DOI: 10.4049/jimmunol.1003193] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A hallmark of autoimmune lymphoproliferative syndrome (ALPS), caused by mutation of the Fas death receptor, is massive lymphadenopathy from aberrant expansion of CD4(-)CD8(-) (double-negative [DN]) T cells. Eomesodermin (Eomes) is a member of the T-box family of transcription factors and plays critical roles in effector cell function and memory cell fitness of CD8(+) T lymphocytes. We provide evidence in this study that DN T cells exhibit dysregulated expression of Eomes in humans and mice with ALPS. We also find that T cell-specific deletion of Eomes prevents lymphoid hypertrophy and accumulation of DN T cells in Fas-mutant mice. Although Eomes has critical physiological roles in the function and homeostasis of CD8(+) T cells, overexpression of Eomes appears to enable pathological induction or expansion of unusual CD8-related T cell subsets. Thus, antagonism of Eomes emerges as a therapeutic target for DN T cell ablation in ALPS.
Collapse
Affiliation(s)
- Ichiko Kinjyo
- Abramson Family Cancer Research Institute, Division of Infectious Diseases, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Crispín JC, Tsokos GC. Human TCR-alpha beta+ CD4- CD8- T cells can derive from CD8+ T cells and display an inflammatory effector phenotype. THE JOURNAL OF IMMUNOLOGY 2009; 183:4675-81. [PMID: 19734235 DOI: 10.4049/jimmunol.0901533] [Citation(s) in RCA: 127] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The origin and function of human double negative (DN) TCR-alphabeta+ T cells is unknown. They are thought to contribute to the pathogenesis of systemic lupus erythematosus because they expand and accumulate in inflamed organs. In this study, we provide evidence that human TCR-alphabeta+ CD4- CD8- DN T cells can derive from activated CD8+ T cells. Freshly isolated TCR-alphabeta+ DN T cells display a distinct gene expression and cytokine production profile. DN cells isolated from peripheral blood as well as DN cells derived in vitro from CD8+ T cells produce a defined array of proinflammatory mediators that includes IL-1beta, IL-17, IFN-gamma, CXCL3, and CXCL2. These results indicate that, upon activation, CD8+ T cells have the capacity to acquire a distinct phenotype that grants them inflammatory capacity.
Collapse
Affiliation(s)
- José C Crispín
- Department of Medicine, Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
14
|
Abstract
The lpr mutation in the Fas gene leads to symptoms of autoimmune disease, including polyclonal B-lymphocyte activation and lymphoproliferation. The expanding T-lymphocyte populations in the disease are characterized by high expression levels of the memory marker CD44. It has not been known whether CD44 expression contributes to the pathophysiology of the condition or merely reflects a consequence of excessive lymphocyte activation. We therefore tested the role of CD44 gene products in Fas(lpr/lpr) disease in gene targeted mice. We bred CD44 knockout mice and C57Bl/6-lpr mice to generate the Fas(lpr/lpr) CD44+/+, and Fas(lpr/lpr) CD44-/- genotypes and analyzed the disease manifestations around 215 days of age. The absence of CD44 substantially reduced the immunoglobulin secretion and lymphocyte expansion that are characteristic of the Fas(lpr/lpr) syndrome. Surprisingly, the percentage of CD3+ CD4- CD8- (double negative) cells in peripheral lymphoid organs increased in Fas(lpr/lpr) CD44-/- mice almost to the same extent as in Fas(lpr/lpr) CD44+/+ mice. These results indicate that the expansion of the fraction of double negative cells in spleens and lymph nodes, believed to be generated by down-regulation of CD8, does not depend on increased lymphocyte numbers. Furthermore, they corroborate an essential role for CD44 gene products in the T-cell expansion and polyclonal B-cell activation that constitute the Fas(lpr/lpr) syndrome. The CD44 receptor may be a suitable therapeutic target for inhibition of lymphoproliferation in autoimmune diseases.
Collapse
Affiliation(s)
- Georg F Weber
- Department of Radiation Oncology, New England Medical Center, 750 Washington Street, NEMC #824, Boston, MA 02111, USA.
| |
Collapse
|
15
|
Prevention of autoimmunity and control of recall response to exogenous antigen by Fas death receptor ligand expression on T cells. Immunity 2008; 29:922-33. [PMID: 19013083 DOI: 10.1016/j.immuni.2008.10.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Revised: 10/03/2008] [Accepted: 10/22/2008] [Indexed: 12/29/2022]
Abstract
Mice with mutations in the gene encoding Fas ligand (FasL) develop lymphoproliferation and systemic autoimmune diseases. However, the cellular subset responsible for the prevention of autoimmunity in FasL-deficient mice remains undetermined. Here, we show that mice with FasL loss on either B or T cells had identical life span as littermates, and both genotypes developed signs of autoimmunity. In addition, we show that T cell-dependent death was vital for the elimination of aberrant T cells and for controlling the numbers of B cells and dendritic cells that dampen autoimmune responses. Furthermore, we show that the loss of FasL on T cells affected the follicular dentritic cell network in the germinal centers, leading to an impaired recall response to exogenous antigen. These results disclose the distinct roles of cellular subsets in FasL-dependent control of autoimmunity and provide further insight into the role of FasL in humoral immunity.
Collapse
|
16
|
Bristeau-Leprince A, Mateo V, Lim A, Magerus-Chatinet A, Solary E, Fischer A, Rieux-Laucat F, Gougeon ML. Human TCR alpha/beta+ CD4-CD8- double-negative T cells in patients with autoimmune lymphoproliferative syndrome express restricted Vbeta TCR diversity and are clonally related to CD8+ T cells. THE JOURNAL OF IMMUNOLOGY 2008; 181:440-8. [PMID: 18566410 DOI: 10.4049/jimmunol.181.1.440] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The peripheral expansion of alpha/beta+-CD4-CD8- double negative (DN) T cells in patients with autoimmune lymphoproliferative syndrome (ALPS) is a consistent feature of this disease, and part of the diagnostic criteria of ALPS. The origin of these cells remains undetermined. They could derive from mature T cells that have lost coreceptor expression, or represent a special minor cell lineage. To investigate relationship of DN and single positive (SP) T cells in ALPS, we used Immunoscope technology to analyze the TCRVbeta repertoire diversity of sorted DN and SP T cells, and we performed CDR3 sequence analyses of matching clonotypes. We show that DN T cells express all the Vbeta gene families that are used by their SP counterparts, though they dominantly use some Vbeta genes. Analysis of CDR3 length distribution revealed a diverse polyclonal TCR repertoire for sorted CD4+ T cells, whereas both DN and CD8+ T cells showed a skewed TCR repertoire with oligoclonal expansions throughout most of the Vbeta families. CDR3 sequencing of matching clonotypes revealed a significant sharing of CDR3 sequences from selected Vbeta-Jbeta transcripts between DN and CD8+ T cells. Altogether, these data strongly argue for a CD8 origin of DN T cells in ALPS.
Collapse
Affiliation(s)
- Anne Bristeau-Leprince
- Antiviral Immunity, Biotherapy and Vaccine Unit, Infection and Epidemiology Department, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Mateo V, Ménager M, de Saint-Basile G, Stolzenberg MC, Roquelaure B, André N, Florkin B, le Deist F, Picard C, Fischer A, Rieux-Laucat F. Perforin-dependent apoptosis functionally compensates Fas deficiency in activation-induced cell death of human T lymphocytes. Blood 2007; 110:4285-92. [PMID: 17724145 DOI: 10.1182/blood-2007-05-088286] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Activation-induced cell death (AICD) is involved in peripheral tolerance by controlling the expansion of repeatedly stimulated T cells via an apoptotic Fas (CD95; APO-1)-dependent pathway. The TNFRSF-6 gene encoding Fas is mutated in children suffering from autoimmune lymphoproliferative syndrome (ALPS), which is characterized by lymphoproliferation and autoimmunity. We examined AICD in Fas-deficient T cells from ALPS patients. We showed that primary activated Fas-deficient T cells die by apoptosis after repeated T cell antigen receptor (TCR) stimulation despite resistance to Fas-mediated cell death. This Fas-independent AICD was found to be mediated through a cytotoxic granules-dependent pathway. Cytotoxic granules-mediated AICD was also detected in normal T lymphocytes though to a lesser extent. As expected, the cytotoxic granules-dependent AICD was abolished in T cells from Rab27a- or perforin-deficient patients who exhibited defective granules-dependent cytotoxicity. Supporting an in vivo relevance of the cytotoxic granules-dependent AICD in ALPS patients, we detected an increased number of circulating T lymphocytes expressing granzymes A and B. Altogether, these data indicated that the cytotoxic granules-dependent cell death in ALPS may compensate for Fas deficiency in T lymphocytes. Furthermore, they identified a novel AICD pathway as a unique alternative to Fas apoptosis in human peripheral T lymphocytes.
Collapse
|
18
|
Mohamood AS, Guler ML, Xiao Z, Zheng D, Hess A, Wang Y, Yagita H, Schneck JP, Hamad ARA. Protection from autoimmune diabetes and T-cell lymphoproliferation induced by FasL mutation are differentially regulated and can be uncoupled pharmacologically. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 171:97-106. [PMID: 17591957 PMCID: PMC1941609 DOI: 10.2353/ajpath.2007.070148] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/28/2007] [Indexed: 12/16/2022]
Abstract
Spontaneous mutation of Fas (lpr) or FasL (gld) completely protects nonobese diabetic mice from autoimmune diabetes but also causes massive double-negative T-cell lymphoproliferation. In this study, we used bone marrow chimeras and adoptive transfer analysis to investigate further the role of FasL in the pathogenesis of autoimmune diabetes and to determine whether gld-induced tolerance and double-negative T-cell lymphoproliferation can be uncoupled from each other. We show that FasL expressed on hematopoietic and nonhematopoietic compartments plays nonredundant roles in the pathogenesis of autoimmune diabetes. Mutation of FasL in either compartment interferes with the autoimmune process and prevents onset of diabetes, but FasL expressed in the hematopoietic compartment is the dominant regulator of T-cell homeostasis. Furthermore, pathogenesis of diabetes is dependent on normal FasL expression in both compartments, whereas only minimal FasL function is required to maintain T-cell homeostasis. Consequently, partial disruption of FasL protects from autoimmune diabetes without causing T-cell lymphoproliferation. This is demonstrated genetically in nonobese diabetic-gld/+ mice and pharmacologically by using FasL-neutralizing antibody. These results have important implications for understanding the role of the Fas pathway in pathogenesis of autoimmune diseases and for designing novel FasL-modulating therapies.
Collapse
Affiliation(s)
- Abdiaziz S Mohamood
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Rieux-Laucat F. [Autoimmune lymphoproliferative syndrome: an inherited or a somatic defect of apoptosis]. Med Sci (Paris) 2006; 22:645-50. [PMID: 16828042 DOI: 10.1051/medsci/20062267645] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Control of lymphocyte homeostasis is essential to ensure efficient immune responses and to prevent autoimmunity. Expansion followed by contraction of the lymphocyte pool are the basis of adaptive immune responses, and apoptosis is a crucial cellular modus operandi of the contraction phase. The death receptor Fas is a key player in lymphocyte apoptosis induction and patients lacking a functional Fas receptor develop a chronic lymphoproliferation termed autoimmune lymphoproliferative syndrome (ALPS). In rare instances, defects of the Fas signaling pathway have been associated with the ALPS condition. Although these defects with familial history are usually caused by inherited mutations of the corresponding genes, somatic mosaicism of these Fas mutations were also found in sporadic cases of ALPS. These findings might have important implications in deciphering the pathophysiological bases of other autoimmune diseases.
Collapse
Affiliation(s)
- Frédéric Rieux-Laucat
- Inserm U768, Université Paris V, Hôpital Necker, 149, rue de Sèvres, 75015 Paris, France.
| |
Collapse
|
20
|
Ford MS, Zhang ZX, Chen W, Zhang L. Double-negative T regulatory cells can develop outside the thymus and do not mature from CD8+ T cell precursors. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:2803-2809. [PMID: 16920915 DOI: 10.4049/jimmunol.177.5.2803] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Recent studies have demonstrated that activated peripheral alphabeta TCR+ CD3+ CD4- CD8- NK1.1- (double-negative, DN) regulatory T cells (Tregs) from both mice and humans are able to down-regulate immune responses in vitro and in vivo. However, the origin and developmental requirements of functional DN Tregs remain unclear. In this study, we investigated the requirement for CD8 expression as well as the presence of a thymus for the development of functional DN Tregs. We demonstrate that DN Tregs exist in CD8-deficient mice and that stimulation of CD8+ T cells in vivo with TCR-specific Ag does not convert CD8+ T cells into DN Tregs. In addition, we found that DN T cells are present in the spleens and lymph nodes of thymectomized mice that are irradiated and reconstituted with T cell-depleted bone marrow cells. Interestingly, DN Tregs that develop in thymectomized mice can suppress syngeneic CD8+ T cells more effectively than those that develop in sham-thymectomized mice. Taken together, our data suggest that DN Tregs are not derived from CD8+ T cell precursors and that functional DN Tregs may preferentially develop outside of the thymus. These data suggest that DN Tregs may represent a developmentally and functionally unique cell population.
Collapse
Affiliation(s)
- Megan S Ford
- Multiorgan Transplantation Program, Toronto General Research Institute, University Health Network, and Department of Laboratory Medicine and Pathobiology, University of Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
21
|
Wilson CB, Makar KW, Shnyreva M, Fitzpatrick DR. DNA methylation and the expanding epigenetics of T cell lineage commitment. Semin Immunol 2005; 17:105-19. [PMID: 15737572 DOI: 10.1016/j.smim.2005.01.005] [Citation(s) in RCA: 105] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
During their development from progenitors, lymphocytes make a series of cell fate decisions. These decisions reflect and require changes in overall programs of gene expression. To maintain cellular identity, programs of gene expression must be iterated through mitosis in a heritable manner by epigenetic processes, which include DNA methylation, methyl-CpG-binding proteins, histone modifications, transcription factors and higher order chromatin structure. Current evidence is consistent with the notion that DNA methylation acts in concert with other epigenetic processes to limit the probability of aberrant gene expression and to stabilize, rather than to initiate, cell fate decisions. In particular, DNA methylation appears to be a non-redundant repressor of CD8 expression in TCR-gammadelta T cells and Th2 cytokine expression in Th1 and CD8 T cells, and is required to enforce clonally restricted Ly49 and KIR gene expression in NK cells. However, most of our knowledge is derived from in vitro studies, and the importance of DNA methylation in memory cell lineage fidelity in vivo remains to be shown convincingly.
Collapse
|
22
|
Kienzle N, Olver S, Buttigieg K, Groves P, Janas ML, Baz A, Kelso A. Progressive differentiation and commitment of CD8+ T cells to a poorly cytolytic CD8low phenotype in the presence of IL-4. THE JOURNAL OF IMMUNOLOGY 2005; 174:2021-9. [PMID: 15699131 DOI: 10.4049/jimmunol.174.4.2021] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Exposure to IL-4 during activation of naive murine CD8+ T cells leads to generation of IL-4-producing effector cells with reduced surface CD8, low perforin, granzyme B and granzyme C mRNA, and poor cytolytic function. We show in this study that maximal development of these cells depended on exposure to IL-4 for the first 5 days of activation. Although IL-4 was not required at later times, CD8 T cell clones continued to lose surface CD8 expression with prolonged culture, suggesting commitment to the CD8low phenotype. This state was reversible in early differentiation. When single CD8low cells from 4-day cultures were cultured without IL-4, 65% gave rise to clones that partly or wholly comprised CD8high cells; the proportion of reverted clones was reduced or increased when the cells were cloned in the presence of IL-4 or anti-IL-4 Ab, respectively. CD8 expression positively correlated with perforin and granzyme A, B, and C mRNA, and negatively correlated with IL-4 mRNA levels among these clones. By contrast, most CD8low cells isolated at later time points maintained their phenotype, produced IL-4, and exhibited poor cytolytic function after many weeks in the absence of exogenous IL-4. We conclude that IL-4-dependent down-regulation of CD8 is associated with progressive differentiation and commitment to yield IL-4-producing cells with little cytolytic activity. These data suggest that the CD4-CD8- cells identified in some disease states may be the product of a previously unrecognized pathway of effector differentiation from conventional CD8+ T cells.
Collapse
Affiliation(s)
- Norbert Kienzle
- Cooperative Research Centre for Vaccine Technology and Queensland Institute of Medical Research, Brisbane, Queensland, Australia.
| | | | | | | | | | | | | |
Collapse
|
23
|
Sia C, Homo-Delarche F. Tolerance induction and endogenous regeneration of pancreatic beta-cells in established autoimmune diabetes. Rev Diabet Stud 2005; 1:198-206. [PMID: 17491705 PMCID: PMC1783694 DOI: 10.1900/rds.2004.1.198] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Studies aimed at the understanding of the multifactorial development of autoimmune diabetes have made substantial contributions toward elucidating the molecular mechanisms that open the road to an effective prevention of defective immune responses. Immunomodulatory regimens capable of inducing tolerance are shown to be effective even in the reversal of established autoimmune diabetes in animal models. Experimental trials including the reeducation of autoreactive T cells, depletion of macrophages, dendritic cells, and T cells, as well as the use of monoclonal antibodies, have yielded encouraging results, but have not yet been translated into beneficial clinical outcomes. In addition, we are now seeing an emergence of promising new directions aimed at the induction of islet regeneration by endogenous factors, suggesting that the repair of pancreatic tissue is possible without the need for an engraftment of donor tissue. These recent waves of technological progress have injected new hope for a combined therapy to offer diabetic patients long-term benefits of insulin independence. This article reviews the latest findings on diabetic pathogenesis and discusses promising avenues to tolerance induction and islet regeneration.
Collapse
Affiliation(s)
- Charles Sia
- Department of Immunology, United Biomedical Inc., 25 Davids Drive, Hauppage, New York 11788, USA.
| | | |
Collapse
|
24
|
Makar KW, Wilson CB. DNA methylation is a nonredundant repressor of the Th2 effector program. THE JOURNAL OF IMMUNOLOGY 2004; 173:4402-6. [PMID: 15383570 DOI: 10.4049/jimmunol.173.7.4402] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The extent to which DNA methylation contributes to proper regulation of murine T cell effector function is unclear. In this study, we show that in the absence of the maintenance DNA methyltransferase Dnmt1, silencing of IL-4, IL-5, IL-13, and IL-10 in CD8 T cells was abolished, and expression of these Th2 cytokines increased as much as 1000-fold compared with that of control CD8 T cells. Th2 cytokine expression also increased in Dnmt1(-/-) CD4 T cells, but the increase ( approximately 20-40-fold for IL-4 and IL-10, </=5-fold for IL-5 and IL-13) was less than for CD8 T cells. As a result, both Dnmt1(-/-) CD4 and CD8 T cells expressed high and comparable amounts of Th2 cytokines. Loss of Dnmt1 had more subtle effects on IL-2 (</=5-fold increase) and IFN-gamma ( approximately 5-10-fold increase) expression and did not affect the normal bias for greater IL-2 expression by CD4 T cells and greater IFN-gamma expression by CD8 T cells, nor the exclusive expression of perforin and granzyme B by the CD8 T cells. These results indicate that Dnmt1 and DNA methylation are necessary to prevent cell autonomous Th2 cytokine expression in CD8 T cells but are not essential for maintaining proper T cell subset-specific expression of Th1 or CTL effectors. We also found that the expression of Th2 cytokines by Dnmt1(-/-) T cells was appropriately up-regulated in Th2 conditions and down-regulated in Th1 conditions, indicating that transcription factors and DNA methylation are complementary and nonredundant mechanisms by which the Th2 effector program is regulated.
Collapse
Affiliation(s)
- Karen W Makar
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | | |
Collapse
|
25
|
Abstract
The factors affecting T cell viability vary depending on the type and status of the T cell involved. Naive T cells die via a Bcl-2/Bim dependent route. Their deaths are prevented in animals by IL-7 and contact with MHC. Activated T cells die in many different ways. Among these is a pathway involving signals that come from outside the T cell and affect it via surface receptors such as Fas. Activated T cells also die through a pathway driven by signals generated within the T cell itself, a cell autonomous route. This pathway involves members of the Bcl-2 family, in particular Bcl-2, Bcl-xl, Bim, and probably Bak. The viability of CD8+ and CD4+ memory T cells is controlled in different ways. CD8+ memory T cells are maintained by IL-15 and IL-7. The control of CD4+ memory T cells is more mysterious, with roles reported for IL-7 and/or contact via the TCR.
Collapse
Affiliation(s)
- Philippa Marrack
- Howard Hughes Medical Institute and Integrated Department of Immunology, National Jewish Medical and Research Center, and Department of Medicine, University of Colorado Health Sciences Center, Denver, Colorado 80206, USA.
| | | |
Collapse
|
26
|
Kong PL, Morel L, Croker BP, Craft J. The centromeric region of chromosome 7 from MRL mice (Lmb3) is an epistatic modifier of Fas for autoimmune disease expression. THE JOURNAL OF IMMUNOLOGY 2004; 172:2785-94. [PMID: 14978078 DOI: 10.4049/jimmunol.172.5.2785] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Lupus is a prototypic systemic autoimmune disease that has a significant genetic component in its etiology. Several genome-wide screens have identified multiple loci that contribute to disease susceptibility in lupus-prone mice, including the Fas-deficient MRL/Fas(lpr) strain, with each locus contributing in a threshold liability manner. The centromeric region of chromosome 7 was identified as a lupus susceptibility locus in MRL/Fas(lpr) mice as Lmb3. This locus was backcrossed onto the resistant C57BL/6 (B6) background, in the presence or absence of Fas, resulting in the generation of B6.MRLc7 congenic animals. Detailed analysis of these animals showed that Lmb3 enhances and accelerates several characteristics of lupus, including autoantibody production, kidney disease, and T cell activation, as well as accumulation of CD4(-)CD8(-) double-negative T cells, the latter a feature of Fas-deficient mice. These effects appeared to be dependent on the interaction between Lmb3 and Fas deficiency, as Lmb3 on the B6/+(Fas-lpr) background did not augment any of the lupus traits measured. These findings confirm the role of Lmb3 in lupus susceptibility, as a modifier of Fas(lpr) phenotype, and illustrate the importance of epistatic interaction between genetic loci in the etiology of lupus. Furthermore, they suggest that the genetic lesion(s) in MRLc7 is probably different from those in NZMc7 (Sle3/5), despite a significant overlap of these two intervals.
Collapse
Affiliation(s)
- Philip L Kong
- Section of Rheumatology, Department of Internal Medicine, and Section of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | | | |
Collapse
|
27
|
McCarty N, Shinohara ML, Lu L, Cantor H. Detailed analysis of gene expression during development of T cell lineages in the thymus. Proc Natl Acad Sci U S A 2004; 101:9339-44. [PMID: 15190182 PMCID: PMC438978 DOI: 10.1073/pnas.0402654101] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The genetic mechanisms that promote lineage commitment and eliminate autoreactive cells in the thymus are not well understood. To better understand this process, we have identified and quantitated transcripts in the two major thymocyte lineages by using serial analysis of gene expression. Approximately 25 genes displayed almost complete segregation to one or the other T cell lineage. Commitment to the CD4 lineage was marked by up-regulation of genes associated with increased survival and chaperone function followed by expression of genes that regulate nucleosome remodeling and T cell receptor signaling. Differentiation within the CD8 lineage, on the other hand, was marked by up-regulation of genes that regulate lymphocyte homing, followed by quenching of genes that inhibit apoptosis. Definition of differential gene expression during development of the two major thymocyte lineages will allow insight into mechanisms of T cell development after positive and negative selection.
Collapse
Affiliation(s)
- Nami McCarty
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, and Department of Pathology, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
28
|
Rush LJ, Raval A, Funchain P, Johnson AJ, Smith L, Lucas DM, Bembea M, Liu TH, Heerema NA, Rassenti L, Liyanarachchi S, Davuluri R, Byrd JC, Plass C. Epigenetic profiling in chronic lymphocytic leukemia reveals novel methylation targets. Cancer Res 2004; 64:2424-33. [PMID: 15059895 DOI: 10.1158/0008-5472.can-03-2870] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
CpG island methylation is an epigenetic alteration that contributes to tumorigenesis by transcriptional inactivation of genes. Little is known about the overall levels of CpG island methylation in chronic lymphocytic leukemia (CLL). To provide a baseline estimate of global aberrant methylation and identify target sequences for additional investigation, we performed Restriction Landmark Genomic Scanning on 10 CLL samples. Two methylation-sensitive landmark enzymes were used (NotI and AscI), allowing assessment of over 3000 CpG islands in each sample. Tumor-derived Restriction Landmark Genomic Scanning profiles were compared with profiles from CD19-selected B cells from normal volunteers and matched normal neutrophils from 4 CLL patients. We found 2.5-8.1% (mean 4.8%) of the CpG islands in CLL samples were aberrantly methylated compared with controls, and the methylation events had a nonrandom distribution (P < 0.0001). Furthermore, we identified 193 aberrantly methylated sequences, of which 93% have CpG island characteristics and 90% have homology to genes or expressed sequences. One such gene, the G protein-coupled metabotropic glutamate receptor 7 (GRM7), possibly inhibits cyclic AMP signaling in the induction of apoptosis. Bisulfite sequencing of GRM7 confirmed extensive CpG island methylation, and treatment with 5-aza-2'-deoxycytidine (decitabine) resulted in up-regulated expression of several genes in vitro with concurrent cellular depletion of DNMT1 protein. Our dual-enzyme global methylation study shows that CLL is characterized by widespread nonrandom CpG island methylation similar to other tumors and provides a panel of novel methylation targets that can be used in larger studies designed to assess impact on disease progression and survival.
Collapse
Affiliation(s)
- Laura J Rush
- Department of Veterinary Biosciences, Comprehensive Cancer Center and The Ohio State University, Columbus, Ohio 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Aliahmad P, O'Flaherty E, Han P, Goularte OD, Wilkinson B, Satake M, Molkentin JD, Kaye J. TOX provides a link between calcineurin activation and CD8 lineage commitment. ACTA ACUST UNITED AC 2004; 199:1089-99. [PMID: 15078895 PMCID: PMC2211890 DOI: 10.1084/jem.20040051] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
T cell development is dependent on the integration of multiple signaling pathways, although few links between signaling cascades and downstream nuclear factors that play a role in thymocyte differentiation have been identified. We show here that expression of the HMG box protein TOX is sufficient to induce changes in coreceptor gene expression associated with β-selection, including CD8 gene demethylation. TOX expression is also sufficient to initiate positive selection to the CD8 lineage in the absence of MHC–TCR interactions. TOX-mediated positive selection is associated with up-regulation of Runx3, implicating CD4 silencing in the process. Interestingly, a strong T cell receptor–mediated signal can modify this cell fate. We further demonstrate that up-regulation of TOX in double positive thymocytes is calcineurin dependent, linking this critical signaling pathway to nuclear changes during positive selection.
Collapse
Affiliation(s)
- Parinaz Aliahmad
- Department of Immunology IMM-8, The Scripps Research Institute, 10550 N. Torrey Pines Rd., La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Prins RM, Incardona F, Lau R, Lee P, Claus S, Zhang W, Black KL, Wheeler CJ. Characterization of Defective CD4−CD8−T Cells in Murine Tumors Generated Independent of Antigen Specificity. THE JOURNAL OF IMMUNOLOGY 2004; 172:1602-11. [PMID: 14734741 DOI: 10.4049/jimmunol.172.3.1602] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immune-based therapy confers limited benefits to hosts bearing late-stage tumors. Mounting evidence points to local suppression of T cell function as the most substantial barrier to effective antitumor immunity in hosts with large tumor burdens. Despite this, events responsible for locally defective T cells and immune suppression in tumors remain unclear. We describe in this study a predominant T cell population localized within two murine tumors that is characterized by expression of apoptotic markers and TCRalphabeta/CD3, but not CD4, CD8, or NK-associated markers. These defective cells resembled double negative (DN) T cells in lpr mice, harbored defects in the expression of T cell signaling molecules, and produced the anti-inflammatory cytokine, IL-10. Conditions known to increase or decrease the accumulation of lpr DN T cells had corresponding effects on local DN tumor infiltrating lymphocyte (TIL) levels and inversely impacted host survival. Adoptive transfer into s.c. tumors demonstrated that naive CD8(+) T cells were highly susceptible to becoming DN TIL, and local supplementation of tumors with nontumor Ag-bearing MHC class I-expressing fibroblasts decreased both this susceptibility and endogenous DN TIL levels. These findings identify a major defective T cell population with suppressive potential within tumors. The data also suggest that local T cell defectiveness is controlled by the tumor environment independent of cognate Ag specificity per se. Decreasing defective DN TIL levels by increasing noncognate peptide MHC class I availability, or modulating TCR or cytokine signaling may facilitate host survival by bolstering endogenous immunity to late-stage tumors, and may help improve therapeutic tumor vaccines.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- CD4 Antigens/metabolism
- CD8 Antigens/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/transplantation
- Cell Aggregation/immunology
- Cell Division/immunology
- Cell Line, Tumor
- Epitopes, T-Lymphocyte/physiology
- Female
- Glioma/immunology
- Glioma/mortality
- Glioma/pathology
- Histocompatibility Antigens Class I/physiology
- Immunohistochemistry
- Immunophenotyping
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/metabolism
- Lymphocytes, Tumor-Infiltrating/pathology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/mortality
- Melanoma, Experimental/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Protein-Tyrosine Kinases/deficiency
- Protein-Tyrosine Kinases/genetics
- Protein-Tyrosine Kinases/physiology
- Proto-Oncogene Proteins/deficiency
- Proto-Oncogene Proteins/genetics
- Proto-Oncogene Proteins/physiology
- Proto-Oncogene Proteins c-fyn
- Signal Transduction/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocyte Subsets/pathology
- fas Receptor/physiology
Collapse
Affiliation(s)
- Robert M Prins
- Maxine Dunitz Neurosurgical Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Xiao S, Sung SSJ, Fu SM, Ju ST. Combining Fas Mutation with Interleukin-2 Deficiency Prevents Colitis and Lupus. J Biol Chem 2003; 278:52730-8. [PMID: 14525977 DOI: 10.1074/jbc.m308707200] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Both the lpr gene defect and interleukin 2-targeted mutation (IL-2 KO) in mice are lethal. Interestingly, mice bearing both mutations live significantly longer than mice with either of the single mutant genes, approximating the life span of normal controls. They do not display the major disease phenotypes of lpr and IL-2 KO mice. Systemic autoimmune response, the accumulation of the abnormal CD4-CD8-B220+ double-negative T cells, kidney disease pathology, anemia, colon damage, and lethality are prevented. Our data indicate that IL-2 is mandatory for the expansion of auto-reactive T cells in lpr mice and that CD95 (Fas) is the critical target for the development of anemia and ulcerative colitis in IL-2 KO mice in which CD178 (FasL) on intraepithelial T cells is the major effector responsible for colon damage and lethality.
Collapse
Affiliation(s)
- Sheng Xiao
- Division of Rheumatology and Immunology, Department of Internal Medicine, and Special Center of Research on Systemic Lupus Erythematosus, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | |
Collapse
|
32
|
Fitzpatrick DR, Wilson CB. Methylation and demethylation in the regulation of genes, cells, and responses in the immune system. Clin Immunol 2003; 109:37-45. [PMID: 14585274 DOI: 10.1016/s1521-6616(03)00205-5] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
DNA methylation is a focus of epigenetic research in the immune system. This overview begins with a synopsis of the players and processes involved in DNA methylation, demethylation, methyl-CpG-recognition, histone modification, and chromatin remodeling. The role of these mechanisms in immune responses, with a focus on T lymphocytes, is then reviewed. There is evidence for epigenetic regulation of several key immune processes including thymocyte development, antigen presentation, differentiation, cytokine expression, effector function, and memory. DNA methylation contributes, along with other epigenetic mechanisms, to the establishment of transcriptional thresholds that vary between genes and T cell types. The immune system is a fertile field for studies of epigenetic regulation of cell fate and function.
Collapse
Affiliation(s)
- David R Fitzpatrick
- Immunological Systems Department, Amgen Inc, 51 University St, Seattle, WA 98101, USA.
| | | |
Collapse
|
33
|
Abstract
FasL (CD95L) is a well-known and well-characterized death-inducing ligand. Spontaneous mutations in FasL and its cognate receptor Fas (CD95) have helped understand the role of these molecules in the disease. Once thought to be mainly involved in the homeostasis of immune system, the territory of FasL regulation has been expanded to angiogenesis and tumor progression. Here, we review what is currently known about the role of FasL in many areas of biology.
Collapse
Affiliation(s)
- Hae-ock Lee
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, St Louis, MO 63110, USA
| | | |
Collapse
|
34
|
Guillaume P, Legler DF, Boucheron N, Doucey MA, Cerottini JC, Luescher IF. Soluble major histocompatibility complex-peptide octamers with impaired CD8 binding selectively induce Fas-dependent apoptosis. J Biol Chem 2003; 278:4500-9. [PMID: 12407102 DOI: 10.1074/jbc.m208863200] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Fluorescence-labeled soluble major histocompatibility complex class I-peptide "tetramers" constitute a powerful tool to detect and isolate antigen-specific CD8(+) T cells by flow cytometry. Conventional "tetramers" are prepared by refolding of heavy and light chains with a specific peptide, enzymatic biotinylation at an added C-terminal biotinylation sequence, and "tetramerization" by reaction with phycoerythrin- or allophycocyanin-labeled avidin derivatives. We show here that such preparations are heterogeneous and describe a new procedure that allows the preparation of homogeneous tetra- or octameric major histocompatibility complex-peptide complexes. These compounds were tested on T1 cytotoxic T lymphocytes (CTLs), which recognize the Plasmodium berghei circumsporzoite peptide 252-260 (SYIPSAEKI) containing photoreactive 4-azidobenzoic acid on Lys(259) in the context of H-2K(d). We report that mutation of the CD8 binding site of K(d) greatly impairs the binding of tetrameric but not octameric or multimeric K(d)-PbCS(ABA) complexes to CTLs. This mutation abolishes the ability of the octamer to elicit significant phosphorylation of CD3, intracellular calcium mobilization, and CTL degranulation. Remarkably, however, this octamer efficiently activates CTLs for Fas (CD95)-dependent apoptosis.
Collapse
Affiliation(s)
- Philippe Guillaume
- Ludwig Institute for Cancer Research, Lausanne Branch, University of Lausanne, 1066 Epalinges, Switzerland
| | | | | | | | | | | |
Collapse
|
35
|
Penfornis A, Yan G, Shi L, Faustman DL. Polymorphisms of human TAP2 detected by denaturing gradient gel electrophoresis. Hum Immunol 2003; 64:156-67. [PMID: 12507827 DOI: 10.1016/s0198-8859(02)00687-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The human transporter associated with antigen processing (TAP1 and TAP2) genes are located in the human leukocyte antigen (HLA) class II region of the genome and encode proteins that form a heterodimer essential for the transport of endogenous peptides into the endoplasmic reticulum for assembly with HLA class I molecules. Type 1 diabetes is an autoimmune disease that is associated with the HLA region of the genome, with HLA class II genes conferring the greatest statistical risk. The presentation of self-peptides by HLA class I molecules is defective in individuals with this disease, and both TAP1 and TAP2 are potential contributors to this defect. Denaturing gradient gel electrophoresis (DGGE) was applied to screen all 11 exons and the 3' flanking region of TAP2 for polymorphisms in individuals with type 1 diabetes patients and controls. Seventy polymorphisms, including 51 in introns, 4 in the 3' flanking region, and 15 in exons, were identified. Sequencing of polymorphic DNA fragments revealed several new polymorphisms, including a Gln --> Arg substitution at codon 611 and a GT --> GC polymorphism affecting the donor splice site of intron 4, that might be of functional significance. None of the polymorphisms examined differed in frequency between individuals with type 1 diabetes and controls.
Collapse
Affiliation(s)
- Alfred Penfornis
- Immunobiology Laboratory, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | |
Collapse
|
36
|
Abstract
The DNA methylation profile of cancer cells is frequently characterized by global hypomethylation and simultaneous hypermethylation of selected CpG island gene promoters. In recent years, the epigenetic phenomenon of DNA promoter methylation has gained increasing recognition as an important mechanism for transcriptional inactivation of cancer related genes. Studies on both liquid and solid tumors have revealed myriad aberrant methylation events, some of which may provide important clues to the pathogenesis of these tumors. The identification of these methylation alterations and elucidation of the mechanistic events surrounding them are of prime importance, as the methylation status of cancer cells can now be manipulated in vivo with demethylating chemotherapeutics.
Collapse
Affiliation(s)
- Laura J Rush
- Department of Veterinary Biosciences, The Ohio State University, Columbus, OH, USA.
| | | |
Collapse
|
37
|
König R, Shen X, Maroto R, Denning TL. The role of CD4 in regulating homeostasis of T helper cells. Immunol Res 2002; 25:115-30. [PMID: 11999166 DOI: 10.1385/ir:25:2:115] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Intrathymic T cell selection and peripheral activation of mature T cells are crucial for self-recognition and the general immune response to viral, bacterial, and tumor antigens. The T cell coreceptors, CD4 and CD8, contribute to the regulation of these processes. The importance of interactions between CD4 and molecules encoded by the class II major histocompatibility complex (MHC) for thymic T cell selection has been clearly established, however, the role of CD4-MHC class II interactions in T helper (TH) cell differentiation, in the maintenance of homeostasis in the peripheral immune system, and in the generation of memory TH cells is largely unclear. Here, we present evidence for a role of CD4 in controlling homeostasis in the peripheral immune system. We also demonstrate the importance of CD4-MHC class II interactions in inducing these previously not recognized functions of CD4.
Collapse
Affiliation(s)
- Rolf König
- Department of Microbiology and Immunology and the Sealy Center for Molecular Science, The University of Texas Medical Branch, Galveston 77555-1070, USA.
| | | | | | | |
Collapse
|
38
|
Santourlidis S, Trompeter HI, Weinhold S, Eisermann B, Meyer KL, Wernet P, Uhrberg M. Crucial role of DNA methylation in determination of clonally distributed killer cell Ig-like receptor expression patterns in NK cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:4253-61. [PMID: 12370356 DOI: 10.4049/jimmunol.169.8.4253] [Citation(s) in RCA: 195] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Human NK cells are characterized by the expression of surface receptors of the killer cell Ig-like receptor (KIR) family, which are involved in the specific recognition of pathogenic target cells. Each NK cell expresses and maintains an individual subset of inhibitory and stimulatory KIR and in this way contributes to a diversified NK cell repertoire. To date, the molecular basis for generation of clonally distributed KIR expression patterns has been elusive. Here, analyses of DNA methylation patterns of KIR genes in NK cell lines as well as in NK cells, freshly isolated from peripheral blood, demonstrated that a small CpG island surrounding the transcriptional start site of each KIR gene is consistently demethylated in expressed KIR and methylated in unexpressed KIR. DNA-demethylating treatment resulted in a rapid and stable induction of transcription and cell surface expression of all formerly unexpressed KIR in NK cell lines, NK cell clones, and freshly isolated NK cells, but not in other cell types. In vitro methylation of KIR CpG islands repressed reporter gene expression in NK cells. We conclude that clonal patterns of KIR expression are mainly epigenetically determined and maintained through DNA methylation.
Collapse
Affiliation(s)
- Simeon Santourlidis
- Institute for Transplantation Diagnostics and Cell Therapeutics, Heinrich Heine University, Moorenstrasse 5, D-40225 Dusseldorf, Germany
| | | | | | | | | | | | | |
Collapse
|
39
|
Shi X, Xie C, Kreska D, Richardson JA, Mohan C. Genetic dissection of SLE: SLE1 and FAS impact alternate pathways leading to lymphoproliferative autoimmunity. J Exp Med 2002; 196:281-92. [PMID: 12163557 PMCID: PMC2193943 DOI: 10.1084/jem.20010955] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Genetic dissection of lupus pathogenesis in the NZM2410 strain has recently revealed that Sle1 is a potent locus that triggers the formation of IgG anti-histone/DNA antibodies, when expressed on the B6 background as a congenic interval. B6.lpr mice, in contrast, exhibit distinctly different cellular and serological phenotypes. Both strains, however, do not usually exhibit pathogenic autoantibodies, or succumb to lupus nephritis. In this study, we show that the epistatic interaction of Sle1 (in particular, Sle1/Sle1) with FAS(lpr) leads to massive lymphosplenomegaly (with elevated numbers of activated CD4 T cells, CD4(-)CD8(-) double negative (DN) T cells, and B1a cells), high levels of IgG and IgM antinuclear (including anti-ssDNA, anti-dsDNA, and anti-histone/DNA), and antiglomerular autoantibodies, histological, and clinical evidence of glomerulonephritis, and >80% mortality by 5-6 mo of age. Whereas FAS(lpr) functions as a recessive gene, Sle1 exhibits a gene dosage effect. These studies indicate that Sle1 and FAS(lpr) must be impacting alternate pathways leading to lymphoproliferative autoimmunity.
Collapse
Affiliation(s)
- Xiaoyan Shi
- Simmon's Arthritis Research Center and the Center for Immunology, University of Texas Southwestern Medical School, Dallas 75235, USA
| | | | | | | | | |
Collapse
|
40
|
Surquin M, Le Moine A, Flamand V, Nagy N, Rombaut K, Demoor FX, Stordeur P, Salmon I, Guéry JC, Goldman M, Abramowicz D. Skin graft rejection elicited by beta 2-microglobulin as a minor transplantation antigen involves multiple effector pathways: role of Fas-Fas ligand interactions and Th2-dependent graft eosinophil infiltrates. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:500-6. [PMID: 12077281 DOI: 10.4049/jimmunol.169.1.500] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Beta(2)-microglobulin (beta(2)m)-derived peptides are minor transplantation Ags in mice as beta(2)m-positive skin grafts (beta(2)m(+/+)) are rejected by genetically beta(2)m-deficient recipient mice (beta(2)m(-/-)). We studied the effector pathways responsible for the rejection induced by beta(2)-microglobulin-derived minor transplantation Ags. The rejection of beta(2)m(+/+) skin grafts by naive beta(2)m(-/-) mice was dependent on both CD4 and CD8 T cells as shown by administration of depleting mAbs. Experiments performed with beta(2)m(-/-)CD8(-/-) double knockout mice grafted with a beta(2)m(+/+) MHC class I-deficient skin showed that sensitized CD4 T cells directed at beta(2)m peptides-MHC class II complexes are sufficient to trigger rapid rejection. Rejection of beta(2)m(+/+) grafts was associated with the production of IL-5 in vitro, the expression of IL-4 and IL-5 mRNAs in the grafted tissue, and the presence within rejected grafts of a considerable eosinophil infiltrate. Blocking IL-4 and IL-5 in vivo and depleting eosinophils with an anti-CCR3 mAb prevented graft eosinophil infiltration and prolonged beta(2)m(+/+) skin graft survival. Lymphocytes from rejecting beta(2)m(-/-) mice also displayed an increased production of IFN-gamma after culture with beta(2)m(+/+) minor alloantigens. In vivo neutralization of IFN-gamma inhibited skin graft rejection. Finally, beta(2)m(+/+) skin grafts harvested from B6(lpr/lpr) donor mice, which lack a functional Fas molecule, survived longer than wild-type beta(2)m(+/+) skin grafts, showing that Fas-Fas ligand interactions are involved in the rejection process. We conclude that IL-4- and IL-5-dependent eosinophilic rejection, IFN-gamma-dependent mechanisms, and Fas-Fas ligand interactions are effector pathways in the acute rejection of minor transplantation Ags.
Collapse
Affiliation(s)
- Murielle Surquin
- Laboratory of Experimental Immunology, Université Libre de Bruxelles, and Department of Nephrology, Hôpital Erasme, Brussels, Belgium.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Trimble LA, Prince KA, Pestano GA, Daley J, Cantor H. Fas-dependent elimination of nonselected CD8 cells and lpr disease. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4960-7. [PMID: 11994447 DOI: 10.4049/jimmunol.168.10.4960] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
MHC/self peptide interactions with cognate coreceptor/TCR complexes are central to homeostasis of the T cell repertoire. Recent reports have also underlined the critical role of IL-15/IL-2 cytokines in regulating this homeostatic process. In this study, we investigate mechanisms that regulate potentially autoreactive CD8 cells that have escaped intrathymic selection. These cells, upon exit from the thymus, express high levels of CD44, B220, and the IL-15R/IL-2R, and undergo fas-dependent apoptosis. Defects in fas signaling allow increased IL-15/IL-2-dependent survival of these CD44/B220(+) CD8(+) as well as the double-negative T cells characteristic of lpr disease.
Collapse
Affiliation(s)
- Linda A Trimble
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, 44 Binney Street, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
42
|
Aranami T, Iwabuchi K, Onoé K. Syngeneic mixed lymphocyte reaction (SMLR) with dendritic cells: direct visualization of dividing T cell subsets in SMLR. Cell Immunol 2002; 217:67-77. [PMID: 12426002 DOI: 10.1016/s0008-8749(02)00520-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Syngeneic mixed lymphocyte reaction (SMLR) has been considered to represent T cell response to self antigens. In this study using stimulator dendritic cells (DC), we analyzed cellular components responding to the syngeneic DC. It was shown that the predominant dividing cells were CD8(+) T cells although the response of CD4(+) T cells was essential for initiation of SMLR. In spite of the vigorous proliferation and expression of several activation markers, these SMLR-activated CD8(+) T cells hardly killed syngeneic targets and most of the CD8(+) T cells produced no interferon-gamma upon restimulation with DC. Furthermore, in SMLR where CD8(+) T cells were absent or inhibited, a considerable proliferation of CD4(-) CD8(-) double negative-T cells that included TCRalpha/beta(+) natural killer-T cells (NKT cells), TCRgamma/delta(+) NKT cells and TCRgamma/delta(+) T cells was observed.
Collapse
MESH Headings
- Animals
- Antigens, Differentiation, T-Lymphocyte/analysis
- Autoantigens/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cell Division
- Cells, Cultured
- Coculture Techniques
- Cytotoxicity Tests, Immunologic
- Dendritic Cells/immunology
- Female
- Fluoresceins
- Fluorescent Dyes
- Killer Cells, Natural/immunology
- Lymphocyte Activation
- Lymphocyte Culture Test, Mixed
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Antigen, T-Cell/analysis
- Succinimides
- T-Lymphocyte Subsets/classification
- T-Lymphocyte Subsets/immunology
- beta 2-Microglobulin/genetics
Collapse
Affiliation(s)
- Toshimasa Aranami
- Division of Immunobiology, Research Section of Pathophysiology, Institute for Genetic Medicine, Hokkaido University, Kita-15 Nishi-7 Kita-ku, Sapporo 060 0815, Japan
| | | | | |
Collapse
|
43
|
Kienzle N, Buttigieg K, Groves P, Kawula T, Kelso A. A clonal culture system demonstrates that IL-4 induces a subpopulation of noncytolytic T cells with low CD8, perforin, and granzyme expression. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:1672-81. [PMID: 11823496 DOI: 10.4049/jimmunol.168.4.1672] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Immune deviation of cytolytic T cell function, induced by type 2 cytokines like IL-4, is an attractive concept to explain failure of the immune system in some diseases. However, this concept is challenged by previous conflicting results on whether type 2 cytokine-producing CD8(+) T cells are cytolytic. Therefore, we have analyzed the relationship between cytolytic activity and cytokine production among large numbers of primary CD8(+) T cell clones. Single murine CD8(+) T cells of naive phenotype were activated at high efficiency with immobilized Abs to CD3, CD8, and CD11a in the presence of IL-2 (neutral conditions) or IL-2, IL-4, and anti-IFN-gamma Ab (type 2-polarizing conditions) for 8-9 days. Under neutral conditions, most clones produced IFN-gamma without IL-4 and were cytolytic. Under type 2-polarizing conditions, most clones produced IFN-gamma and IL-4 but displayed variable cytolytic activity and CD8 expression. Separation on the basis of surface CD8 levels revealed that, compared with CD8(high) cells from the same cultures, CD8(low) cells were poorly cytolytic and expressed low levels of perforin mRNA and protein and granzyme A, B, and C mRNA. A similar, smaller population of noncytolytic CD8(low) cells was identified among CD8(+) T cells activated in mixed lymphocyte reaction with IL-4. Variable efficiency of generation of the noncytolytic cells may account for the differing results of earlier studies. We conclude that IL-4 promotes the development of a noncytolytic CD8(low) T cell phenotype that might be important in tumor- or pathogen-induced immune deviation.
Collapse
Affiliation(s)
- Norbert Kienzle
- Queensland Institute of Medical Research, Brisbane, Queensland, Australia.
| | | | | | | | | |
Collapse
|
44
|
Abstract
The cytokine osteopontin (Eta-1) leads to macrophage-dependent polyclonal B-cell activation and is induced early in autoimmune prone mice with the lpr mutation, suggesting a significant pathogenic role for this molecule. Indeed, C57BL/6-Fas(lpr/lpr) mice crossed with osteopontin(-/-) mice display delayed onset of polyclonal B-cell activation, as judged by serum immunoglobulin levels. In contrast, they are subject to normal onset, but late exacerbation of lymphoproliferation and evidence of kidney disease. These observations define two stages of Fas(lpr/lpr) disease with respect to osteopontin-dependent pathogenesis that should be taken into account in the design of therapeutic approaches to the clinical disease.
Collapse
Affiliation(s)
- G F Weber
- Department of Cancer Immunology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| | | |
Collapse
|
45
|
Smith K, Seddon B, Purbhoo MA, Zamoyska R, Fisher AG, Merkenschlager M. Sensory adaptation in naive peripheral CD4 T cells. J Exp Med 2001; 194:1253-61. [PMID: 11696591 PMCID: PMC2195983 DOI: 10.1084/jem.194.9.1253] [Citation(s) in RCA: 132] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
T cell receptor interactions with peptide/major histocompatibility complex (pMHC) ligands control the selection of T cells in the thymus as well as their homeostasis in peripheral lymphoid organs. Here we show that pMHC contact modulates the expression of CD5 by naive CD4 T cells in a process that requires the continued expression of p56(lck). Reduced CD5 levels in T cells deprived of pMHC contact are predictive of elevated Ca(2)+ responses to subsequent TCR engagement by anti-CD3 or nominal antigen. Adaptation to peripheral pMHC contact may be important for regulating naive CD4 T cell responsiveness.
Collapse
Affiliation(s)
- K Smith
- Lymphocyte Development Group, MRC Clinical Sciences Centre, ICSM Hammersmith Hospital, London W12 0NN, UK
| | | | | | | | | | | |
Collapse
|
46
|
Xiao S, Marshak-Rothstein A, Ju ST. Sp1 is the major fasl gene activator in abnormal CD4(-)CD8(-)B220(+) T cells of lpr and gld mice. Eur J Immunol 2001; 31:3339-48. [PMID: 11745351 DOI: 10.1002/1521-4141(200111)31:11<3339::aid-immu3339>3.0.co;2-u] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The abnormal CD4(-)CD8(-)TCRalpha beta(+)B220(+) double-negative (DN) T cells that accumulate in lpr and gld mice are refractory to TCR cross-linking and IL-2 stimulation, yet they have an activated phenotype and express a high level of fasl mRNA. Specific binding sites for Sp1, NFAT, Egr, and NF-kappaB have been identified in the promoter region of the fasl gene. To determine the critical factor for fasl gene activation, fasl promoter reporter and mutant constructs were transiently transfected into the abnormal DN T cells. The data demonstrate that the Sp1 binding site is the major response element that regulates fasl promoter activity. Moreover, the abnormal DN T cells contain in their nuclei a high level of Sp1, a low level of NFAT and NF-kappaB, and a very low level of Egr. Ectopic expression of Egr-3 but not Sp1 protein in the abnormal DN T cells enhanced fasl promoter activity, suggesting that the Egr but not Sp1 was limiting for fasl gene activation. Comparison between the abnormal DN T cells and the Sertoli TM4 cells showed a strong correlation between Sp1 expression and fasl mRNA level and FasL function. Our study has identified Sp1 as the major transcription factor responsible for fasl gene activation in the abnormal DN T cells that are defective in signal transduction through TCR and IL-2R, thereby, implicating a novel regulatory pathway for fasl gene activation during the physiological development and elimination of the abnormal DN T cells.
Collapse
Affiliation(s)
- S Xiao
- Department of Medicine, Boston University School of Medicine, Boston, USA
| | | | | |
Collapse
|
47
|
Hayashi K, Abe N, Watanabe T, Obinata M, Ito M, Sato T, Habu S, Satake M. Overexpression of AML1 transcription factor drives thymocytes into the CD8 single-positive lineage. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:4957-65. [PMID: 11673502 DOI: 10.4049/jimmunol.167.9.4957] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
To understand the gene regulation involved in the development of single-positive (SP) thymocytes, we generated transgenic mice in which the AML1 transcription factor is overexpressed. In these mice the number of CD8 SP thymocytes was greatly increased, and this continued to be true even when MHC class I was absent. This promotion to the CD8 SP lineage was not, however, observed when both class I and class II were absent. Furthermore, even thymocytes carrying MHC class II-restricted TCR differentiated into the CD8 SP lineage when AML1 was overexpressed. The selected CD8 SP cells were, however, unable to mature, as judged by the expression level of heat-stable Ag. Thus, overexpression of AML1 is able to skew class II-restricted thymocytes into the CD8 SP lineage, but not to drive the maturation of resulting selected CD8 SP cells.
Collapse
Affiliation(s)
- K Hayashi
- Department of Molecular Immunology, Institute of Development, Aging, and Cancer, Tohoku University, Sendai, Japan
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Panoutsakopoulou V, Sanchirico ME, Huster KM, Jansson M, Granucci F, Shim DJ, Wucherpfennig KW, Cantor H. Analysis of the relationship between viral infection and autoimmune disease. Immunity 2001; 15:137-47. [PMID: 11485745 DOI: 10.1016/s1074-7613(01)00172-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The clinical association between viral infection and onset or exacerbation of autoimmune disorders remains poorly understood. Here, we examine the relative roles of molecular mimicry and nonspecific inflammatory stimuli in progression from infection to autoimmune disease. Murine herpes virus 1 (HSV-1 KOS) infection triggers T cell-dependent autoimmune reactions to corneal tissue. We generated an HSV-1 KOS point mutant containing a single amino acid exchange within the putative mimicry epitope as well as mice expressing a TCR transgene specific for the self-peptide mimic to allow dissection of two pathogenic mechanisms in disease induction. These experiments indicate that viral mimicry is essential for disease induction after low-level viral infection of animals containing limited numbers of autoreactive T cells, while innate immune mechanisms become sufficient to provoke disease in animals containing relatively high numbers of autoreactive T cells.
Collapse
Affiliation(s)
- V Panoutsakopoulou
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Ryu S, Kodama S, Ryu K, Schoenfeld DA, Faustman DL. Reversal of established autoimmune diabetes by restoration of endogenous beta cell function. J Clin Invest 2001; 108:63-72. [PMID: 11435458 PMCID: PMC209340 DOI: 10.1172/jci12335] [Citation(s) in RCA: 120] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2001] [Accepted: 05/14/2001] [Indexed: 11/17/2022] Open
Abstract
In NOD (nonobese diabetic) mice, a model of autoimmune diabetes, various immunomodulatory interventions prevent progression to diabetes. However, after hyperglycemia is established, such interventions rarely alter the course of disease or allow sustained engraftment of islet transplants. A proteasome defect in lymphoid cells of NOD mice impairs the presentation of self antigens and increases the susceptibility of these cells to TNF-alpha-induced apoptosis. Here, we examine the hypothesis that induction of TNF-alpha expression combined with reeducation of newly emerging T cells with self antigens can interrupt autoimmunity. Hyperglycemic NOD mice were treated with CFA to induce TNF-alpha expression and were exposed to functional complexes of MHC class I molecules and antigenic peptides either by repeated injection of MHC class I matched splenocytes or by transplantation of islets from nonautoimmune donors. Hyperglycemia was controlled in animals injected with splenocytes by administration of insulin or, more effectively, by implantation of encapsulated islets. These interventions reversed the established beta cell-directed autoimmunity and restored endogenous pancreatic islet function to such an extent that normoglycemia was maintained in up to 75% of animals after discontinuation of treatment and removal of islet transplants. A therapy aimed at the selective elimination of autoreactive cells and the reeducation of T cells, when combined with control of glycemia, is thus able to effect an apparent cure of established type 1 diabetes in the NOD mouse.
Collapse
Affiliation(s)
- S Ryu
- Immunobiology Laboratory, Harvard Medical School and Massachusetts General Hospital, Charlestown, Massachusetts 02129, USA
| | | | | | | | | |
Collapse
|
50
|
Abstract
The death of T lymphocytes following their activation involves several signal pathways that converge on a series of proteases, known as caspases, that degrade cellular proteins and activate a DNAse. Caspases are activated through ligation of cell surface death receptors as well as via direct activation of downstream caspases, often through metabolic stress such as cytokine withdrawal or generation of oxygen radicals, that culminates in mitochondrial dysfunction and release of the pro-apoptotic molecules, cytochrome c and Smac/DIABLO. The Bcl-2 family members serve to regulate the mitochondrial membrane integrity. Recent studies are now revealing the significant contribution to the activation-induced cell death of T cells by downstream caspases such as caspase-3 and Bcl-2-homology domain 3 (BH3)-only members of the Bcl-2 family.
Collapse
Affiliation(s)
- R C Budd
- Immunobiology Program, The University of Vermont College of Medicine, Given Medical Building, D-305 05405-0068, Burlington, VT, USA.
| |
Collapse
|