1
|
Hirata SI, Sawane K, Adachi J, Isoyama J, Sugiura Y, Matsunaga A, Hosomi K, Tomonaga T, Suematsu M, Nagatake T, Kunisawa J. Vitamin B1 Supports the Differentiation of T Cells through TGF-β Superfamily Production in Thymic Stromal Cells. iScience 2020; 23:101426. [PMID: 32818907 PMCID: PMC7452312 DOI: 10.1016/j.isci.2020.101426] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 06/12/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
Homeostatic generation of T cells, which occurs in the thymus, is controlled at least in part by endogenous cytokines and ligands. In addition, nutritional factors are other key regulators for the homeostasis of host immunity, but whether and how nutrition affects the homeostatic generation of thymocytes remains to be established. Here, we showed that vitamin B1 deficiency resulted in a bias toward the maturation of γδ thymocytes accompanied by decreased differentiation into double-positive thymocytes during thymic involution. These events were mediated through the increased production of TGF-β superfamily members due to the accumulation of branched-chain α-keto acids in thymic stromal cells. These findings revealed essential roles of vitamin B1 in the appropriate differentiation of T cells through the metabolism of thymic stromal cells.
Collapse
Affiliation(s)
- So-ichiro Hirata
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Asagi Saito, Ibaraki-city, Osaka 567-0085, Japan
- Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe-city, Hyogo 650-0017, Japan
| | - Kento Sawane
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Asagi Saito, Ibaraki-city, Osaka 567-0085, Japan
- Nippon Flour Mills Co., Ltd., Innovation Center, Midorigaoka, Atsugi-city, Kanagawa 243-0041, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamadaoka, Suita-city, Osaka 565-0871, Japan
| | - Jun Adachi
- Laboratory of Proteome Research, NIBIOHN, Asagi Saito, Ibaraki-city, Osaka 567-0085, Japan
| | - Junko Isoyama
- Laboratory of Proteome Research, NIBIOHN, Asagi Saito, Ibaraki-city, Osaka 567-0085, Japan
| | - Yuki Sugiura
- Japan Science and Technology Agency, PRESTO, Honcho, Kawaguchi-city, Saitama 332-0012, Japan
- Department of Biochemistry, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Ayu Matsunaga
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Asagi Saito, Ibaraki-city, Osaka 567-0085, Japan
| | - Koji Hosomi
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Asagi Saito, Ibaraki-city, Osaka 567-0085, Japan
| | - Takeshi Tomonaga
- Laboratory of Proteome Research, NIBIOHN, Asagi Saito, Ibaraki-city, Osaka 567-0085, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | - Takahiro Nagatake
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Asagi Saito, Ibaraki-city, Osaka 567-0085, Japan
| | - Jun Kunisawa
- Laboratory of Vaccine Materials, Center for Vaccine and Adjuvant Research, and Laboratory of Gut Environmental System, National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Asagi Saito, Ibaraki-city, Osaka 567-0085, Japan
- Department of Microbiology and Immunology, Kobe University Graduate School of Medicine, Kusunoki-cho, Chuo-ku, Kobe-city, Hyogo 650-0017, Japan
- Graduate School of Pharmaceutical Sciences, Osaka University, Yamadaoka, Suita-city, Osaka 565-0871, Japan
- International Research and Development Center for Mucosal Vaccines, The Institute of Medical Science, The University of Tokyo, Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Graduate School of Medicine and Graduate School of Dentistry, Osaka University, Yamadaoka, Suita-city, Osaka 565-0871, Japan
| |
Collapse
|
2
|
Jouan Y, Patin EC, Hassane M, Si-Tahar M, Baranek T, Paget C. Thymic Program Directing the Functional Development of γδT17 Cells. Front Immunol 2018; 9:981. [PMID: 29867959 PMCID: PMC5951931 DOI: 10.3389/fimmu.2018.00981] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/20/2018] [Indexed: 12/27/2022] Open
Abstract
γδT cells comprise a unique T cell sublineage endowed with a wide functional repertoire, which allow them to play important—sometimes opposite—roles in many immune responses associated with infection, cancer, and inflammatory processes. This is largely dependent on the existence of pre-programmed discrete functional subsets that differentiate within the thymus at specific temporal windows of life. Since they represent a major early source of interleukin-17A in many models of immune responses, the γδT17 cell population has recently gained considerable interest. Thus, a better dissection of the developmental program of this effector γδT subset appears critical in understanding their associated immune functions. Several recent reports have provided new exciting insights into the developmental mechanisms that control γδT cell lineage commitment and differentiation. Here, we review the importance of thymic cues and intrinsic factors that shape the developmental program of γδT17 cells. We also discuss the potential future areas of research in γδT17 cell development especially in regards to the recently provided data from deep RNA sequencing technology. Pursuing our understanding into this complex mechanism will undoubtedly provide important clues into the biology of this particular T cell sublineage.
Collapse
Affiliation(s)
- Youenn Jouan
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France.,Service de Médecine Intensive Réanimation, Centre Hospitalier Régional Universitaire de Tours, Tours, France
| | - Emmanuel C Patin
- Division of Radiotherapy and Imaging, Targeted Therapy Team, The Institute of Cancer Research, London, United Kingdom
| | - Maya Hassane
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Mustapha Si-Tahar
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Thomas Baranek
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| | - Christophe Paget
- INSERM, Centre d'Etude des Pathologies Respiratoires (CEPR), UMR 1100, Tours, France.,Université de Tours, Tours, France
| |
Collapse
|
3
|
Identification of lineage-specifying cytokines that signal all CD8 +-cytotoxic-lineage-fate 'decisions' in the thymus. Nat Immunol 2017; 18:1218-1227. [PMID: 28945245 PMCID: PMC5659273 DOI: 10.1038/ni.3847] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 08/29/2017] [Indexed: 12/11/2022]
Abstract
T cell antigen receptor (TCR) signaling in the thymus initiates positive selection but CD8 lineage fate is thought to be induced by cytokines after TCR signaling has ceased, although this remains controversial and unproven. We now identify four non-common gamma chain (γc) receptor-signaling cytokines (IL-6, IFN-γ, TSLP, TGF-β) that, like IL-7 and IL-15, induce expression of the lineage-specifying transcription factor Runx3d and signal the generation of CD8 T cells. Remarkably, elimination of in vivo signaling by all ‘lineage-specifying cytokines’ during positive selection eliminated Runx3d expression and completely abrogated CD8 single-positive thymocyte generation. Thus, this study proves that signaling during positive selection by lineage-specifying cytokines is responsible for all CD8 lineage fate decisions in the thymus.
Collapse
|
4
|
Ohigashi I, Kozai M, Takahama Y. Development and developmental potential of cortical thymic epithelial cells. Immunol Rev 2016; 271:10-22. [DOI: 10.1111/imr.12404] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Izumi Ohigashi
- Division of Experimental Immunology; Institute for Genome Research; University of Tokushima; Tokushima Japan
| | - Mina Kozai
- Division of Experimental Immunology; Institute for Genome Research; University of Tokushima; Tokushima Japan
| | - Yousuke Takahama
- Division of Experimental Immunology; Institute for Genome Research; University of Tokushima; Tokushima Japan
| |
Collapse
|
5
|
Nitta T, Suzuki H. Thymic stromal cell subsets for T cell development. Cell Mol Life Sci 2016; 73:1021-37. [PMID: 26825337 PMCID: PMC11108406 DOI: 10.1007/s00018-015-2107-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 11/26/2015] [Accepted: 12/01/2015] [Indexed: 12/20/2022]
Abstract
The thymus provides a specialized microenvironment in which a variety of stromal cells of both hematopoietic and non-hematopoietic origin regulate development and repertoire selection of T cells. Recent studies have been unraveling the inter- and intracellular signals and transcriptional networks for spatiotemporal regulation of development of thymic stromal cells, mainly thymic epithelial cells (TECs), and the molecular mechanisms of how different TEC subsets control T cell development and selection. TECs are classified into two functionally different subsets: cortical TECs (cTECs) and medullary TECs (mTECs). cTECs induce positive selection of diverse and functionally distinct T cells by virtue of unique antigen-processing systems, while mTECs are essential for establishing T cell tolerance via ectopic expression of peripheral tissue-restricted antigens and cooperation with dendritic cells. In addition to reviewing the role of the thymic stroma in conventional T cell development, we will discuss recently discovered novel functions of TECs in the development of unconventional T cells, such as natural killer T cells and γδT cells.
Collapse
Affiliation(s)
- Takeshi Nitta
- Department of Immunology and Pathology, Research Institute, National Center for Global Health and Medicine, Chiba, 272-8516, Japan.
| | - Harumi Suzuki
- Department of Immunology and Pathology, Research Institute, National Center for Global Health and Medicine, Chiba, 272-8516, Japan.
| |
Collapse
|
6
|
Akiyama T, Tateishi R, Akiyama N, Yoshinaga R, Kobayashi TJ. Positive and Negative Regulatory Mechanisms for Fine-Tuning Cellularity and Functions of Medullary Thymic Epithelial Cells. Front Immunol 2015; 6:461. [PMID: 26441966 PMCID: PMC4568481 DOI: 10.3389/fimmu.2015.00461] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 08/24/2015] [Indexed: 01/10/2023] Open
Abstract
Self-tolerant T cells and regulatory T cells develop in the thymus. A wide variety of cell-cell interactions in the thymus is required for the differentiation, proliferation, and repertoire selection of T cells. Various secreted and cell surface molecules expressed in thymic epithelial cells (TECs) mediate these processes. Moreover, cytokines expressed by cells of hematopoietic origin regulate the cellularity of TECs. Tumor necrosis factor (TNF) family RANK ligand, lymphotoxin, and CD40 ligand, expressed in T cells and innate lymphoid cells (ILCs), promote the differentiation and proliferation of medullary TECs (mTECs) that play critical roles in the induction of immune tolerance. A recent study suggests that interleukin-22 (IL-22) produced by ILCs promotes regeneration of TECs after irradiation. Intriguingly, tumor growth factor-β and osteoprotegerin limit cellularity of mTECs, thereby attenuating regulatory T cell generation. We will review recent insights into the molecular basis for cell-cell interactions regulating differentiation and proliferation of mTECs and also discuss about a perspective on use of mathematical models for understanding this complicated system.
Collapse
Affiliation(s)
- Taishin Akiyama
- Division of Cellular and Molecular Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan
| | - Ryosuke Tateishi
- Division of Cellular and Molecular Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan
| | - Nobuko Akiyama
- Division of Cellular and Molecular Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan
| | - Riko Yoshinaga
- Division of Cellular and Molecular Biology, Institute of Medical Science, University of Tokyo , Tokyo , Japan
| | | |
Collapse
|
7
|
Linhares-Lacerda L, Palu CC, Ribeiro-Alves M, Paredes BD, Morrot A, Garcia-Silva MR, Cayota A, Savino W. Differential Expression of microRNAs in Thymic Epithelial Cells from Trypanosoma cruzi Acutely Infected Mice: Putative Role in Thymic Atrophy. Front Immunol 2015; 6:428. [PMID: 26347748 PMCID: PMC4543887 DOI: 10.3389/fimmu.2015.00428] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/06/2015] [Indexed: 12/21/2022] Open
Abstract
A common feature seen in acute infections is a severe atrophy of the thymus. This occurs in the murine model of acute Chagas disease. Moreover, in thymuses from Trypanosoma cruzi acutely infected mice, thymocytes exhibit an increase in the density of fibronectin and laminin integrin-type receptors, with an increase in migratory response ex vivo. Thymic epithelial cells (TEC) play a major role in the intrathymic T cell differentiation. To date, the consequences of molecular changes promoted by parasite infection upon thymus have not been elucidated. Considering the importance of microRNA for gene expression regulation, 85 microRNAs (mRNAs) were analyzed in TEC from T. cruzi acutely infected mice. The infection significantly modulated 29 miRNAs and modulation of 9 was also dependent whether TEC sorted out from the thymus exhibited cortical or medullary phenotype. In silico analysis revealed that these miRNAs may control target mRNAs known to be responsible for chemotaxis, cell adhesion, and cell death. Considering that we sorted TEC in the initial phase of thymocyte loss, it is conceivable that changes in TEC miRNA expression profile are functionally related to thymic atrophy, providing new clues to better understanding the mechanisms of the thymic involution seen in experimental Chagas disease.
Collapse
Affiliation(s)
- Leandra Linhares-Lacerda
- Laboratory on Thymus Research, Institute Oswaldo Cruz, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| | - Cintia Cristina Palu
- Laboratory on Thymus Research, Institute Oswaldo Cruz, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| | - Marcelo Ribeiro-Alves
- HIV/AIDS Clinical Research Center, National Institute of Infectious Diseases, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| | - Bruno Diaz Paredes
- The National Institute of Science and Technology for Structural Biology and Bioimaging, Federal University of Rio de Janeiro , Rio de Janeiro , Brazil
| | - Alexandre Morrot
- Department of Immunology, Microbiology Institute, Federal University of Rio de Janeiro , Rio de Janeiro , Brazil
| | | | - Alfonso Cayota
- Functional Genomics Unit, Institut Pasteur de Montevideo , Montevideo , Uruguay
| | - Wilson Savino
- Laboratory on Thymus Research, Institute Oswaldo Cruz, Oswaldo Cruz Foundation , Rio de Janeiro , Brazil
| |
Collapse
|
8
|
Lepletier A, Chidgey AP, Savino W. Perspectives for Improvement of the Thymic Microenvironment through Manipulation of Thymic Epithelial Cells: A Mini-Review. Gerontology 2015; 61:504-14. [DOI: 10.1159/000375160] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/13/2015] [Indexed: 11/19/2022] Open
|
9
|
A regulatory role for TGF-β signaling in the establishment and function of the thymic medulla. Nat Immunol 2014; 15:554-61. [DOI: 10.1038/ni.2869] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2013] [Accepted: 03/11/2014] [Indexed: 12/16/2022]
|
10
|
Gentek R, Munneke JM, Helbig C, Blom B, Hazenberg MD, Spits H, Amsen D. Modulation of Signal Strength Switches Notch from an Inducer of T Cells to an Inducer of ILC2. Front Immunol 2013; 4:334. [PMID: 24155745 PMCID: PMC3804867 DOI: 10.3389/fimmu.2013.00334] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 10/02/2013] [Indexed: 11/20/2022] Open
Abstract
Innate lymphoid cells (ILCs) are emerging key players of the immune system with close lineage relationship to T cells. ILC2 play an important role in protective immunity against multicellular parasites, but are also involved in the pathogenesis of type 2 immune diseases. Here, we have studied the developmental requirements for human ILC2. We report that ILC2 are present in the thymus of young human donors, possibly reflecting local differentiation. Furthermore, we show that uncommitted lineage−CD34+CD1a−human thymic progenitors have the capacity to develop into ILC2 in vitro under the influence of Notch signaling, either by stimulation with the Notch ligand Delta like 1 (Dll1) or by expression of the active intracellular domain of NOTCH1 (NICD1). The capacity of NICD1 to mobilize the ILC2 differentiation program was sufficiently potent to override commitment to the T cell lineage in CD34+CD1a+ progenitors and force them into the ILC2 lineage. As Notch is an important factor also for T cell development, these results raise the question how one and the same signaling pathway can elicit such distinct developmental outcomes from the same precursors. We provide evidence that Notch signal strength is a critical determinant in this decision: by tuning signal amplitude, Notch can be converted from a T cell inducer (low signal strength) to an ILC2 inducer (high signal strength). Thus, this study enhances our understanding of human ILC2 development and identifies a mechanism determining specificity of Notch signal output during T cell and ILC2 differentiation.
Collapse
Affiliation(s)
- Rebecca Gentek
- Department of Cell Biology and Histology, Academic Medical Center , Amsterdam , Netherlands
| | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
The thymic cortex provides a microenvironment that supports the generation and T cell antigen receptor (TCR)-mediated selection of CD4(+)CD8(+)TCRαβ(+) thymocytes. Cortical thymic epithelial cells (cTECs) are the essential component that forms the architecture of the thymic cortex and induces the generation as well as the selection of newly generated T cells. Here we summarize current knowledge on the development, function, and heterogeneity of cTECs, focusing on the expression and function of β5t, a cTEC-specific subunit of the thymoproteasome.
Collapse
|
12
|
Do JS, Fink PJ, Li L, Spolski R, Robinson J, Leonard WJ, Letterio JJ, Min B. Cutting edge: spontaneous development of IL-17-producing gamma delta T cells in the thymus occurs via a TGF-beta 1-dependent mechanism. THE JOURNAL OF IMMUNOLOGY 2010; 184:1675-9. [PMID: 20061408 DOI: 10.4049/jimmunol.0903539] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In naive animals, gammadelta T cells are innate sources of IL-17, a potent proinflammatory cytokine mediating bacterial clearance as well as autoimmunity. However, mechanisms underlying the generation of these cells in vivo remain unclear. In this study, we show that TGF-beta1 plays a key role in the generation of IL-17(+) gammadelta T cells and that it mainly occurs in the thymus particularly during the postnatal period. Interestingly, IL-17(+) gammadelta TCR(+) thymocytes were mainly CD44(high)CD25(low) cells, which seem to derive from double-negative 4 gammadelta TCR(+) cells that acquired CD44 and IL-17 expression. Our findings identify a novel developmental pathway during which IL-17-competent gammadelta T cells arise in the thymus by a TGF-beta1-dependent mechanism.
Collapse
Affiliation(s)
- Jeong-su Do
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Xu M, Sharma A, Hossain MZ, Wiest DL, Sen JM. Sustained expression of pre-TCR induced beta-catenin in post-beta-selection thymocytes blocks T cell development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:759-65. [PMID: 19124718 PMCID: PMC2701226 DOI: 10.4049/jimmunol.182.2.759] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Pre-TCR and IL-7R signals regulate beta-selection of thymocytes and then must be down-regulated for further development. However, the molecular events that control down-regulation remain unknown. We and others have previously shown that beta-catenin in cooperation with TCF regulates beta-selection. In this paper, we demonstrate that beta-catenin expression is stringently regulated by intrathymic signals, it is expressed at the highest levels in the pre-TCR signaled thymocytes, and is down-regulated in post-beta-selection thymocytes. Pre-TCR-induced beta-catenin regulates initial stages of pre-TCR signaling including expression of early growth response (Egr) genes but must be down-regulated to express RORgammat, which is essential for maturation to the CD4+CD8+ double positive (DP) stage. Sustained expression of beta-catenin results in the generation of IL-7R-, Egr-, and TGFbeta-expressing pre-DP thymocytes that are blocked in development. These data are consistent with a model in which post-beta-selection, pre-TCR-induced beta-catenin expression must return to background levels for efficient transition to the DP stage.
Collapse
Affiliation(s)
- Mai Xu
- Lymphocyte Development Unit, Laboratory of Immunology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | | | | | |
Collapse
|
14
|
Maintenance of a normal thymic microenvironment and T-cell homeostasis require Smad4-mediated signaling in thymic epithelial cells. Blood 2008; 112:3688-95. [PMID: 18695001 DOI: 10.1182/blood-2008-04-150532] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Signals mediated by the transforming growth factor-beta superfamily of growth factors have been implicated in thymic epithelial cell (TEC) differentiation, homeostasis, and function, but a direct reliance on these signals has not been established. Here we demonstrate that a block in canonical transforming growth factor-beta signaling by the loss of Smad4 expression in TECs leads to qualitative changes in TEC function and a progressively disorganized thymic microenvironment. Moreover, the number of thymus resident early T-lineage progenitors is severely reduced in the absence of Smad4 expression in TECs and directly correlates with extensive thymic and peripheral lymphopenia. Our observations hence place Smad4 within the signaling events in TECs that determine total thymus cellularity by controlling the number of early T-lineage progenitors.
Collapse
|
15
|
Hauri-Hohl MM, Zuklys S, Keller MP, Jeker LT, Barthlott T, Moon AM, Roes J, Holländer GA. TGF-beta signaling in thymic epithelial cells regulates thymic involution and postirradiation reconstitution. Blood 2008; 112:626-34. [PMID: 18474727 PMCID: PMC2481556 DOI: 10.1182/blood-2007-10-115618] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 03/28/2008] [Indexed: 01/21/2023] Open
Abstract
The thymus constitutes the primary lymphoid organ responsible for the generation of naive T cells. Its stromal compartment is largely composed of a scaffold of different subsets of epithelial cells that provide soluble and membrane-bound molecules essential for thymocyte maturation and selection. With senescence, a steady decline in the thymic output of T cells has been observed. Numeric and qualitative changes in the stromal compartment of the thymus resulting in reduced thymopoietic capacity have been suggested to account for this physiologic process. The precise cellular and molecular mechanisms underlying thymic senescence are, however, only incompletely understood. Here, we demonstrate that TGF-beta signaling in thymic epithelial cells exerts a direct influence on the cell's capacity to support thymopoiesis in the aged mouse as the physiologic process of thymic senescence is mitigated in mice deficient for the expression of TGF-beta RII on thymic epithelial cells. Moreover, TGF-beta signaling in these stromal cells transiently hinders the early phase of thymic reconstitution after myeloablative conditioning and hematopoietic stem cell transplantation. Hence, inhibition of TGF-beta signaling decelerates the process of age-related thymic involution and may hasten the reconstitution of regular thymopoiesis after hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Mathias M Hauri-Hohl
- Laboratory of Pediatric Immunology, Center for Biomedicine, Department of Clinical-Biological Sciences, University of Basel and The University Children's Hospital (UKBB), Basel, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Sonmez MF, Colakoglu N, Kukner A, Ozan E, Dabak DO. Immunolocalization of TGF-beta2 in the rat thymus during late stages of prenatal development. Acta Histochem 2008; 111:68-73. [PMID: 18554691 DOI: 10.1016/j.acthis.2008.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2007] [Revised: 04/09/2008] [Accepted: 04/16/2008] [Indexed: 10/22/2022]
Abstract
The aim of this study was to investigate the immunolocalization of transforming growth factor beta (TGF-beta2) in rat thymic stromal cells and thymocytes and investigate the roles of TGF-beta2 in thymopoiesis during the late stages of fetal development. Twelve adult pregnant female Wistar rats weighing 250-270 g were used in this study. The rats were killed by cervical dislocation on gestation days 16 (GD16), 18 (GD18) and 20 (GD20). Fetal thymus glands were prepared and examined by an immunohistochemical technique to reveal binding of an anti-TGF-beta2 rabbit polyclonal antibody. The thymic primordium was surrounded with a connective tissue capsule at GD16 and at this stage TGF-beta2 immunoreactivity was not observed. At GD18, the connective tissue capsule had formed septa which subdivided the tissue into lobules and at this stage TGF-beta2 immunolocalization was detected in the capsule and in thymocytes. Lobulation was more evident at GD20 and TGF-beta2 immunoreactivity of thymocytes was more extensive than on GD18. Results indicate that TGF-beta2 may play an important role in the organization or development of thymocytes in the late stages of thymopoiesis.
Collapse
|
17
|
Nitta T, Murata S, Ueno T, Tanaka K, Takahama Y. Thymic microenvironments for T-cell repertoire formation. Adv Immunol 2008; 99:59-94. [PMID: 19117532 DOI: 10.1016/s0065-2776(08)00603-2] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Functionally competent immune system includes a functionally competent T-cell repertoire that is reactive to foreign antigens but is tolerant to self-antigens. The repertoire of T cells is primarily formed in the thymus through positive and negative selection of developing thymocytes. Immature thymocytes that undergo V(D)J recombination of T-cell antigen receptor (TCR) genes and that express the virgin repertoire of TCRs are generated in thymic cortex. The recent discovery of thymoproteasomes, a molecular complex specifically expressed in cortical thymic epithelial cells (cTEC), has revealed a unique role of cTEC in cuing the further development of immature thymocytes in thymic cortex, possibly by displaying unique self-peptides that induce positive selection. Cortical thymocytes that receive TCR-mediated positive selection signals are destined to survive for further differentiation and are induced to express CCR7, a chemokine receptor. Being attracted to CCR7 ligands expressed by medullary thymic epithelial cells (mTEC), CCR7-expressing positively selected thymocytes relocate to thymic medulla. The medullary microenvironment displays another set of unique self-peptides for trimming positively selected T-cell repertoire to establish self-tolerance, via promiscuous expression of tissue-specific antigens by mTEC and efficient antigen presentation by dendritic cells. Recent results demonstrate that tumor necrosis factor (TNF) superfamily ligands, including receptor activating NF-kappaB ligand (RANKL), CD40L, and lymphotoxin, are produced by positively selected thymocytes and pivotally regulate mTEC development and thymic medulla formation.
Collapse
Affiliation(s)
- Takeshi Nitta
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, Tokushima 770-8503, Japan
| | | | | | | | | |
Collapse
|
18
|
Rubtsov YP, Rudensky AY. TGFbeta signalling in control of T-cell-mediated self-reactivity. Nat Rev Immunol 2007; 7:443-53. [PMID: 17525753 DOI: 10.1038/nri2095] [Citation(s) in RCA: 253] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In the immune system, transforming growth factor-beta (TGFbeta) affects multiple cell lineages by either promoting or opposing their differentiation, survival and proliferation. Understanding the cellular mechanisms of TGFbeta-mediated regulation is complicated due to a broad distribution of TGFbeta receptors on the surface of different immune-cell types. Recent studies using in vivo genetic approaches revealed a critical role for TGFbeta signalling in T cells in restraining fatal autoimmune lesions. Here, we review recent advances in our understanding of a role for TGFbeta signalling in the regulation of T-cell differentiation in the thymus and in the periphery, with a particular emphasis on TGFbeta-mediated control of self-reactive T cells.
Collapse
Affiliation(s)
- Yuri P Rubtsov
- Department of Immunology, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | | |
Collapse
|
19
|
Tzachanis D, Li L, Lafuente EM, Berezovskaya A, Freeman GJ, Boussiotis VA. Twisted gastrulation (Tsg) is regulated by Tob and enhances TGF-beta signaling in activated T lymphocytes. Blood 2007; 109:2944-52. [PMID: 17164348 PMCID: PMC1852213 DOI: 10.1182/blood-2006-03-006510] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Quiescent T cells express Tob, an APRO gene family member, which functions as a transcriptional regulator. Subtractive hybridization identified Twisted gastrulation (Tsg) as one of the genes suppressed by Tob. Tsg is a secreted protein that interacts with Drosophila decapentaplegic (Dpp) and its vertebrate orthologs BMP2/4 and regulates morphogenetic effects in embryos. Here, we report the expression and function of Tsg in human T cells. Tsg mRNA was almost undetectable in unstimulated T cells and was up-regulated after activation by TCR/CD3 and either CD28, IL-2, or PMA. Tsg protein had no effect on responses of primary T cells to TCR/CD3 stimulation but had a potent inhibitory effect on proliferation and cytokine production of primed alloreactive CD4+ cells. Surprisingly, Tsg did not affect phosphorylation of the BMP-specific Smad1 but induced phosphorylation of the TGF-beta-specific Smad2 and mediated DNA binding on Smad3/4 consensus-binding sites, suggesting that it acted downstream of TGF-beta. In vitro association assays revealed a direct interaction of Tsg and TGF-beta proteins. Thus, Tsg functions as an agonist synergizing with TGF-beta to inhibit T-cell activation. Modulation of Tsg signaling may represent a novel target for molecular intervention toward control of aberrant T-cell responses during ongoing graft-versus-host disease (GVHD) and autoimmune diseases.
Collapse
Affiliation(s)
- Dimitrios Tzachanis
- Department of Hematology and Oncology, Beth Israel-Deaconess Medical Center, Boston, MA, USA
| | | | | | | | | | | |
Collapse
|
20
|
Li MO, Wan YY, Flavell RA. T cell-produced transforming growth factor-beta1 controls T cell tolerance and regulates Th1- and Th17-cell differentiation. Immunity 2007; 26:579-91. [PMID: 17481928 DOI: 10.1016/j.immuni.2007.03.014] [Citation(s) in RCA: 568] [Impact Index Per Article: 31.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2007] [Revised: 03/05/2007] [Accepted: 03/19/2007] [Indexed: 12/21/2022]
Abstract
TGF-beta1 is a regulatory cytokine with a pleiotropic role in immune responses. TGF-beta1 is widely expressed in leukocytes and stromal cells. However, the functions of TGF-beta1 expressed by specific lineages of cells remain unknown in vivo. Here, we show that mice with a T cell-specific deletion of the Tgfb1 gene developed lethal immunopathology in multiple organs, and this development was associated with enhanced T cell proliferation, activation, and CD4+ T cell differentiation into T helper 1 (Th1) and Th2 cells. TGF-beta1 produced by Foxp3-expressing regulatory T cells was required to inhibit Th1-cell differentiation and inflammatory-bowel disease in a transfer model. In addition, T cell-produced TGF-beta1 promoted Th17-cell differentiation and was indispensable for the induction of experimental autoimmune encephalomyelitis. These findings reveal essential roles for T cell-produced TGF-beta1 in controlling differentiation of T helper cells and controlling inflammatory diseases.
Collapse
MESH Headings
- Animals
- Cell Differentiation/immunology
- Cells, Cultured
- Colitis/genetics
- Colitis/immunology
- Colitis/metabolism
- Colitis/pathology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Forkhead Transcription Factors/metabolism
- Gene Deletion
- Gene Expression Regulation
- Homeostasis
- Immune Tolerance/immunology
- Lymphocyte Activation/immunology
- Mice
- Mice, Knockout
- T-Lymphocytes/cytology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transforming Growth Factor beta1/biosynthesis
- Transforming Growth Factor beta1/genetics
Collapse
Affiliation(s)
- Ming O Li
- Section of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | | | | |
Collapse
|
21
|
Licona-Limón P, Soldevila G. The role of TGF-beta superfamily during T cell development: new insights. Immunol Lett 2007; 109:1-12. [PMID: 17287030 DOI: 10.1016/j.imlet.2006.12.010] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2006] [Revised: 12/21/2006] [Accepted: 12/23/2006] [Indexed: 10/23/2022]
Abstract
Members of the transforming growth factor beta (TGF-beta) superfamily are soluble factors that regulate a variety of functional responses including proliferation, differentiation, apoptosis and cell cycle, among others, depending not only on the cell type and its differentiation state, but also on the milieu of cytokines present. All three members of this superfamily: TGF-betas, bone morphogenetic proteins (BMPs) and Activins, have been shown to be expressed in the thymus suggesting their potential role as regulators of the T lymphocyte differentiation process. Although initial reports described the role of TGF-beta in controlling specific checkpoints during thymocyte development, recent data has provided new evidence on the role of BMPs and Activins in this process. This review provides new insights on the function of members of the TGF-beta superfamily at different stages of thymocyte development.
Collapse
Affiliation(s)
- P Licona-Limón
- Department of Immunology, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Circuito Escolar s/n, México DF-04510, Mexico
| | | |
Collapse
|
22
|
Woolf E, Brenner O, Goldenberg D, Levanon D, Groner Y. Runx3 regulates dendritic epidermal T cell development. Dev Biol 2006; 303:703-14. [PMID: 17222403 DOI: 10.1016/j.ydbio.2006.12.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2006] [Revised: 11/22/2006] [Accepted: 12/05/2006] [Indexed: 01/19/2023]
Abstract
The Runx3 transcription factor regulates development of T cells during thymopoiesis and TrkC sensory neurons during dorsal root ganglia neurogenesis. It also mediates transforming growth factor-beta signaling in dendritic cells and is essential for development of skin Langerhans cells. Here, we report that Runx3 is involved in the development of skin dendritic epidermal T cells (DETCs); an important component of tissue immunoregulation. In developing DETCs, Runx3 regulates expression of the alphaEbeta7 integrin CD103, known to affect migration and epithelial retention of DETCs. It also regulates expression of IL-2 receptor beta (IL-2Rbeta) that mediates cell proliferation in response to IL-2 or IL-15. In the absence of Runx3, the reduction in CD103 and IL-2Rbeta expression on Runx3(-/-) DETC precursors resulted in impaired cell proliferation and maturation, leading to complete lack of skin DETCs in Runx3(-/-) mice. The data demonstrate the requirement of Runx3 for DETCs development and underscore the importance of CD103 and IL-2Rbeta in this process. Of note, while Runx3(-/-) mice lack both DETCs and Langerhans cells, the two most important components of skin immune surveillance, the mice did not develop skin lesions under pathogen-free (SPF) conditions.
Collapse
Affiliation(s)
- Eilon Woolf
- Department of Molecular Genetics, The Weizmann Institute of Science, Rehovot 76100, Israel
| | | | | | | | | |
Collapse
|
23
|
Takahama Y. Journey through the thymus: stromal guides for T-cell development and selection. Nat Rev Immunol 2006; 6:127-35. [PMID: 16491137 DOI: 10.1038/nri1781] [Citation(s) in RCA: 495] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lympho-stromal interactions in multiple microenvironments within the thymus have a crucial role in the regulation of T-cell development and selection. Recent studies have implicated that chemokines that are produced by thymic stromal cells have a pivotal role in positioning developing T cells within the thymus. In this Review, I discuss the importance of stroma-derived chemokines in guiding the traffic of developing thymocytes, with an emphasis on the processes of cortex-to-medulla migration and T-cell-repertoire selection, including central tolerance.
Collapse
Affiliation(s)
- Yousuke Takahama
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, 3-18-15 Kuramoto, Tokushima 770-8503, Japan.
| |
Collapse
|
24
|
Kurobe H, Liu C, Ueno T, Saito F, Ohigashi I, Seach N, Arakaki R, Hayashi Y, Kitagawa T, Lipp M, Boyd RL, Takahama Y. CCR7-Dependent Cortex-to-Medulla Migration of Positively Selected Thymocytes Is Essential for Establishing Central Tolerance. Immunity 2006; 24:165-77. [PMID: 16473829 DOI: 10.1016/j.immuni.2005.12.011] [Citation(s) in RCA: 228] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2005] [Revised: 10/31/2005] [Accepted: 12/27/2005] [Indexed: 01/05/2023]
Abstract
Immature CD4+CD8+ thymocytes, which are generated in the thymic cortex, are induced upon positive selection to differentiate into mature T lymphocytes and relocate to the thymic medulla. It was recently shown that a chemokine signal via CCR7 is essential for the cortex-to-medulla migration of positively selected thymocytes in the thymus. However, the role of the cortex-to-medulla migration in T cell development and selection has remained unclear. The present study shows that the developmental kinetics and the thymic export of mature thymocytes were undisturbed in adult mice lacking CCR7 or its ligands (CCR7L). The inhibition of sphingosine-1-phosphate-mediated lymphocyte egress from the thymus led to the accumulation of mature thymocytes in the cortex of CCR7- or CCR7L-deficient mice, unlike the accumulation in the medulla of normal mice, thereby suggesting that mature thymocytes may be exported directly from the cortex in the absence of CCR7 signals. However, the thymocytes that were generated in the absence of CCR7 or CCR7L were potent in causing autoimmune dacryoadenitis and sialadenitis in mice and were thus incapable of establishing central tolerance to organ-specific antigens. These results indicate that CCR7-mediated cortex-to-medulla migration of thymocytes is essential for establishing central tolerance rather than for supporting the maturation or export of thymocytes.
Collapse
Affiliation(s)
- Hirotsugu Kurobe
- Division of Experimental Immunology, Institute for Genome Research, University of Tokushima, Tokushima 770-8503, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Konrad MAP, Zúñiga-Pflücker JC. The BTG/TOB family protein TIS21 regulates stage-specific proliferation of developing thymocytes. Eur J Immunol 2005; 35:3030-42. [PMID: 16163674 DOI: 10.1002/eji.200526345] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
As thymocytes undergo differentiation in the thymus, they progress through distinct phases of quiescence and proliferation. Identifying cellular mechanisms that maintain thymocytes in a non-dividing state is critical to fully understand T cell development. A member of the B cell translocation gene/transducer of ErbB-2 (BTG/TOB) family of anti-proliferative proteins was identified as a key mediator of the quiescent state in peripheral anergic and unstimulated T cells. Here, we demonstrate that the BTG/TOB family member TPA-inducible sequence 21 (TIS21) is expressed in quiescent CD44+ CD25- early progenitor thymocytes and CD44- CD25+ cells prior to TCR beta-selection. However, TIS21 expression is decreased in proliferating CD25+ CD44+ progenitor thymocytes and CD25(low) CD44- beta-selected cells, suggesting that its regulated expression may enable thymocytes to remain quiescent in the absence of mitogenic signals. We addressed the role of TIS21 in regulating thymocyte stage-specific expansion by ectopically expressing TIS21 in developing thymocytes and hematopoietic progenitors. Dysregulated expression of TIS21 inhibited the expansion of thymocytes even in the presence of endogenous mitogenic signals, while thymocyte differentiation was unimpeded. These findings imply that the intracellular mechanisms regulating thymocyte differentiation and proliferation, which are induced downstream of developmental cues, function independently during early T cell development.
Collapse
Affiliation(s)
- Mark A P Konrad
- Department of Immunology, University of Toronto, Sunnybrook and Women's Research Institute, Toronto, Ontario, Canada
| | | |
Collapse
|
26
|
Abstract
Transforming growth factor-beta (TGF-beta) plays an essential role in regulating the homeostasis of cells in the lymphoid lineage. TGF-beta signaling is not required for normal thymopoiesis, but is essential for regulating the expansion, activation, and effector function of the mature CD4+ and CD8+ T cells in the peripheral lymphoid organs and target tissues. Recent studies in both mice and humans have elucidated an important and complex role for TGF-beta in regulatory T-cell biology. Disruption of TGF-beta signaling in T cells impairs the maintenance of regulatory T cells, results in the expansion of activated effector T cells, and is associated with the production of cytokines that have major effects on cells in their environment. While autoimmunity and inflammation are the principal phenotypes associated with the abrogation of TGF-beta signaling in T cells in mice, emerging evidence now also directly links Smad-dependent TGF-beta signaling in T cells to the suppression of epithelial neoplasia. The TGF-beta receptor-activated Smad3 plays a critical role in mediating many of the inhibitory effects of TGF-beta signaling in T cells, and has now been established as an important suppressor of leukemogenesis. These studies are increasing our awareness of the many complex mechanisms through which TGF-beta signaling controls the pathogenesis of cancer.
Collapse
Affiliation(s)
- John J Letterio
- The Laboratory of Cell Regulation and Carcinogenesis, The Center for Cancer Research, The National Cancer Institute, National Institutes of Health, Bethesda, MD 20892-5055, USA.
| |
Collapse
|
27
|
Sutherland JS, Goldberg GL, Hammett MV, Uldrich AP, Berzins SP, Heng TS, Blazar BR, Millar JL, Malin MA, Chidgey AP, Boyd RL. Activation of Thymic Regeneration in Mice and Humans following Androgen Blockade. THE JOURNAL OF IMMUNOLOGY 2005; 175:2741-53. [PMID: 16081852 DOI: 10.4049/jimmunol.175.4.2741] [Citation(s) in RCA: 343] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The thymus undergoes age-related atrophy, coincident with increased circulating sex steroids from puberty. The impact of thymic atrophy is most profound in clinical conditions that cause a severe loss in peripheral T cells with the ability to regenerate adequate numbers of naive CD4+ T cells indirectly correlating with patient age. The present study demonstrates that androgen ablation results in the complete regeneration of the aged male mouse thymus, restoration of peripheral T cell phenotype and function and enhanced thymus regeneration following bone marrow transplantation. Importantly, this technique is also applicable to humans, with analysis of elderly males undergoing sex steroid ablation therapy for prostatic carcinoma, demonstrating an increase in circulating T cell numbers, particularly naive (TREC+) T cells. Collectively these studies represent a fundamentally new approach to treating immunodeficiency states in humans.
Collapse
Affiliation(s)
- Jayne S Sutherland
- Department of Immunology, Monash University Medical School, Prahran, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
García-Suárez O, Pérez-Pérez M, Germanà A, Esteban I, Germanà G. Involvement of growth factors in thymic involution. Microsc Res Tech 2003; 62:514-23. [PMID: 14635145 DOI: 10.1002/jemt.10413] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The thymus undergoes an age-dependent degenerative process which is mainly characterized by a progressive loss of lymphoid tissue. Thymic involution is particularly important in relation to immunosenescence and its various associated diseases; this fact has prompted many studies aimed at understanding the causes and mechanisms of thymic degeneration which may, ultimately, lead to the possibility of manipulating it. In this sense, one of the aspects which has deserved most attention is the thymic microenvironment, and more precisely, the many growth factors to which the cells present in the organ are exposed. Thus, the levels of several of such factors have been reported to undergo age-dependent changes in the thymus, which may point at an influence on the regression of the organ. In this article we consider which growth factors and growth factor receptors occur in the vertebrate thymus. Then, focusing on those whose influences are better documented, i.e., neurotrophins, cytokines and IGFs, we discuss their potential role in the organ and the possibility of their being involved in thymic involution.
Collapse
|
29
|
Hager-Theodorides AL, Outram SV, Shah DK, Sacedon R, Shrimpton RE, Vicente A, Varas A, Crompton T. Bone morphogenetic protein 2/4 signaling regulates early thymocyte differentiation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:5496-504. [PMID: 12421925 DOI: 10.4049/jimmunol.169.10.5496] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Bone morphogenetic protein (BMP)2 and BMP4 are involved in the development of many tissues. In this study, we show that BMP2/4 signaling is involved in thymocyte development. Our data suggest that termination of BMP2/4 signaling is necessary for differentiation of CD44(+)CD25(-)CD4(-)CD8(-) double negative (DN) cells along the T cell lineage. BMP2 and BMP4 are produced by the thymic stroma and the requisite BMP receptor molecules (BMPR-1A, BMPR-1B, BMPR-II), and signal transduction molecules (Smad-1, -5, -8, and -4) are expressed by DN thymocytes. BMP4 inhibits thymocyte proliferation, enhances thymocyte survival, and arrests thymocyte differentiation at the CD44(+)CD25(-) DN stage, before T cell lineage commitment. Neutralization of endogenous BMP2 and BMP4 by treatment with the antagonist Noggin promotes and accelerates thymocyte differentiation, increasing the expression of CD2 and the proportion of CD44(-)CD25(-) DN cells and CD4(+)CD8(+) double-positive cells. Our study suggests that the BMP2/4 pathway may function in thymic homeostasis by regulating T cell lineage commitment and differentiation.
Collapse
MESH Headings
- Activin Receptors, Type I/biosynthesis
- Activin Receptors, Type I/genetics
- Adjuvants, Immunologic/pharmacology
- Adjuvants, Immunologic/physiology
- Animals
- Bone Morphogenetic Protein 2
- Bone Morphogenetic Protein 4
- Bone Morphogenetic Protein Receptors, Type I
- Bone Morphogenetic Proteins/antagonists & inhibitors
- Bone Morphogenetic Proteins/physiology
- Carrier Proteins
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Survival/genetics
- Cell Survival/immunology
- Cells, Cultured
- Dose-Response Relationship, Immunologic
- Fetus
- Gene Expression Regulation/immunology
- Genes, T-Cell Receptor beta/genetics
- Genes, T-Cell Receptor delta/genetics
- Growth Inhibitors/physiology
- Hyaluronan Receptors/biosynthesis
- Hyaluronan Receptors/metabolism
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Organ Culture Techniques
- Protein Serine-Threonine Kinases
- Proteins/pharmacology
- Receptors, Growth Factor
- Receptors, Interleukin-2/biosynthesis
- Receptors, Interleukin-2/metabolism
- Recombinant Fusion Proteins/pharmacology
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocytes/cytology
- T-Lymphocytes/metabolism
- Thymus Gland/cytology
- Thymus Gland/metabolism
- Transforming Growth Factor beta
Collapse
|
30
|
Prockop SE, Palencia S, Ryan CM, Gordon K, Gray D, Petrie HT. Stromal cells provide the matrix for migration of early lymphoid progenitors through the thymic cortex. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:4354-61. [PMID: 12370368 DOI: 10.4049/jimmunol.169.8.4354] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
During steady state lymphopoiesis in the postnatal thymus, migration of precursors outward from the deep cortex toward the capsule is required for normal differentiation. Such migration requires, at a minimum, expression of adhesive receptors on the migrating lymphoid cells, as well as a stable matrix of their ligands persisting throughout the region of migration. In this study, we address the nature of this adhesive matrix. Although some precursor stages bound efficiently to extracellular matrix ligands, a specific requirement for the cell surface ligand VCAM-1 was also found. In situ analysis revealed that early precursors are found in intimate contact with a matrix formed by stromal cells in the cortex, a proportion of which expresses VCAM-1. In vivo administration of an anti-VCAM-1 Ab resulted in decreased thymic size and altered distribution of early precursors within the cortex. These results indicate that precursors migrating outward through the cortex may use a cellular, rather than extracellular, matrix for adhesion, and suggest that the VCAM-1(+) subset of cortical stroma may play a crucial role in supporting the migration of early precursors in the steady state thymus.
Collapse
Affiliation(s)
- Susan E Prockop
- Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, NY 10021, USA
| | | | | | | | | | | |
Collapse
|
31
|
Su DM, Manley NR. Stage-specific changes in fetal thymocyte proliferation during the CD4-8- to CD4+8+ transition in wild type, Rag1-/-, and Hoxa3,Pax1 mutant mice. BMC Immunol 2002; 3:12. [PMID: 12241558 PMCID: PMC130029 DOI: 10.1186/1471-2172-3-12] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2002] [Accepted: 09/19/2002] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The function of the thymic microenvironment is to promote thymocyte maturation, in part via regulation of thymocyte proliferation and cell death. Defects in fetal thymic epithelial cell (TEC) development and function, and therefore in the formation of a functional microenvironment, can be caused either directly by TEC differentiation defects or indirectly by defective thymocyte maturation. In this paper we studied fetal thymocyte proliferation during the early transition from the CD3-4-8- (triple negative, TN) to CD4+8+ (double positive, DP) stages. We compared wild type mice with Rag1-/- mice and with Hoxa3+/-Pax1-/- compound mutant mice, which have blocks at different stages of thymocyte development. RESULTS Wild type fetal and adult thymus showed stage-specific differences in the proliferation profiles of developing thymocytes, with fetal stages showing generally higher levels of proliferation. The proliferation profile of fetal thymocytes from Rag1-/- mutants also had stage-specific increases in proliferation compared to wild type fetal thymocytes, in contrast to the lower proliferation previously reported for thymocytes from adult Rag1-/- mutants. We have previously shown that Hoxa3+/-Pax1-/- mice have abnormal fetal TEC development, resulting in increased apoptosis at the TN to DP transition and decreased DP cell numbers. Fetal thymocytes from Hoxa3+/-Pax1-/- compound mutants had increased proliferation, but fewer proliferating cells, at the DP stage. We also observed a decrease in the level of the cytokines IL-7 and SCF produced by Hoxa3+/-Pax1-/-TECs. CONCLUSION Our results indicate complex and stage-specific effects of abnormal TEC development on thymocyte proliferation.
Collapse
Affiliation(s)
- Dong-ming Su
- Department of Genetics, University of Georgia, Athens, Georgia 30602 USA.
| | | |
Collapse
|
32
|
Vaillant F, Blyth K, Andrew L, Neil JC, Cameron ER. Enforced expression of Runx2 perturbs T cell development at a stage coincident with beta-selection. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:2866-74. [PMID: 12218099 DOI: 10.4049/jimmunol.169.6.2866] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The development of T cells in the thymus is regulated by a series of stage-specific transcription factors. Deregulated expression of these factors can lead to alterations in thymocyte development with the production of aberrant cell subsets and predispose to tumor formation. The three genes of the Runx family are multilineage regulators of differentiation that have been reported to be expressed in the T cell lineage. However, their roles in thymocyte development and T cell function are largely unknown. While the Runx2/Cbfa1/AML3/Pebp2alphaa gene plays a primary role in osteogenesis and regulates a number of key bone regulatory genes, we show here that Runx2 is also expressed during the earliest phase of thymic development, in the double-negative subset. Furthermore, enforced expression of Runx2 in transgenic mice under the CD2 promoter was found to affect T cell development at a stage coincident with beta-selection, resulting in an expansion of double-negative CD4 and CD8 immature single-positive cells. Unlike wild-type controls this preselection population (CD4-CD8+heat-stable Ag+TCR-) is in a nonproliferative state, but appears to be primed for further transformation events. Overall the data suggest that Runx2 accelerates development to the CD8 immature single-positive stage, but retards subsequent differentiation to the double-positive stage. Thus, Runx2 joins a small group of transcription factors that can interfere with early T cell development, cause an expansion of a specific subset, and predispose to lymphoma.
Collapse
Affiliation(s)
- François Vaillant
- Molecular Oncology Laboratory, Institute of Comparative Medicine, University of Glasgow Veterinary School, Glasgow, United Kingdom.
| | | | | | | | | |
Collapse
|
33
|
Graf D, Nethisinghe S, Palmer DB, Fisher AG, Merkenschlager M. The developmentally regulated expression of Twisted gastrulation reveals a role for bone morphogenetic proteins in the control of T cell development. J Exp Med 2002; 196:163-71. [PMID: 12119341 PMCID: PMC2193926 DOI: 10.1084/jem.20020276] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
The evolutionarily conserved, secreted protein Twisted gastrulation (Tsg) modulates morphogenetic effects of decapentaplegic (dpp) and its orthologs, the bone morphogenetic proteins 2 and 4 (BMP2/4), in early Drosophila and vertebrate embryos. We have uncovered a role for Tsg at a much later stage of mammalian development, during T cell differentiation in the thymus. BMP4 is expressed by thymic stroma and inhibits the proliferation of CD4(-)CD8(-) double-negative (DN) thymocytes and their differentiation to the CD4(+)CD8(+) double-positive (DP) stage in vitro. Tsg is expressed by thymocytes and up-regulated after T cell receptor signaling at two developmental checkpoints, the transition from the DN to the DP and from the DP to the CD4(+) or CD8(+) single-positive stage. Tsg can synergize with the BMP inhibitor chordin to block the BMP4-mediated inhibition of thymocyte proliferation and differentiation. These data suggest that the developmentally regulated expression of Tsg may allow thymocytes to temporarily withdraw from inhibitory BMP signals.
Collapse
Affiliation(s)
- Daniel Graf
- Lymphocyte Development Group, Medical Research Council Clinical Sciences Centre, Imperial College of Medicine, London W12 0NN, United Kingdom
| | | | | | | | | |
Collapse
|
34
|
Takagi T, Harada J, Ishii S. Murine Schnurri-2 is required for positive selection of thymocytes. Nat Immunol 2001; 2:1048-53. [PMID: 11668343 DOI: 10.1038/ni728] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A key step in T cell development involves the positive selection of cells that recognize antigen presented by self-major histocompatibility complex. Yet, the signals that are activated by T cell receptor engagement and lead to cell survival remain unclear. We show here that mice lacking the transcription factor Schnurri-2 (Shn-2), a large metal-finger protein, had severely defective positive selection of CD4+ and CD8+ cells. Drosophila Shn acts as a cofactor of Smad homolog and is required for Decapentaplegic signaling. Vertebrates have at least three Shn orthologs (Shn-1, Shn-2 and Shn-3), which are thought to act as nuclear targets in the bone morphogenetic protein-transforming growth factor-beta-activin signaling pathways. These data raised the possibility that the Smad-Shn-2 complex is involved in the thymic selection of T cells.
Collapse
Affiliation(s)
- T Takagi
- Laboratory of Molecular Genetics, RIKEN Tsukuba Institute, CREST Research Project of JST, 3-1-1 Koyadai, Tsukuba, Ibaraki 305-0074, Japan
| | | | | |
Collapse
|
35
|
Abstract
The human thymus is required for establishment of a normal T cell repertoire in fetal development, as children born without a thymus (DiGeorge Syndrome) lack thymus-derived (T) and T cell immunity. While the function of the thymus in children for production of new T cells is clear, it has not been obvious that the adult thymus can produce significant numbers of new T cells. Until recently, no assays were available to directly evaluate postnatal thymic function. This paper reviews work on human thymic aging at Duke University School of Medicine and discusses the relevance of this work to devising new strategies for T cell immune reconstitution in man.
Collapse
Affiliation(s)
- B F Haynes
- Department of Medicine, Duke University Arthritis Center, Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
| | | | | | | |
Collapse
|
36
|
Lind EF, Prockop SE, Porritt HE, Petrie HT. Mapping precursor movement through the postnatal thymus reveals specific microenvironments supporting defined stages of early lymphoid development. J Exp Med 2001; 194:127-34. [PMID: 11457887 PMCID: PMC2193450 DOI: 10.1084/jem.194.2.127] [Citation(s) in RCA: 273] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2001] [Accepted: 06/05/2001] [Indexed: 11/29/2022] Open
Abstract
Cellular differentiation is a complex process involving integrated signals for lineage specification, proliferation, endowment of functional capacity, and survival or cell death. During embryogenesis, spatially discrete environments regulating these processes are established during the growth of tissue mass, a process that also results in temporal separation of developmental events. In tissues that undergo steady-state postnatal differentiation, another means for inducing spatial and temporal separation of developmental cues must be established. Here we show that in the postnatal thymus, this is achieved by inducing blood-borne precursors to enter the organ in a narrow region of the perimedullary cortex, followed by outward migration across the cortex before accumulation in the subcapsular zone. Notably, blood precursors do not transmigrate the cortex in an undifferentiated state, but rather undergo progressive developmental changes during this process, such that defined precursor stages appear in distinct cortical regions. Identification of these cortical regions, together with existing knowledge regarding the genetic potential of the corresponding lymphoid precursors, sets operational boundaries for stromal environments that are likely to induce these differentiative events. We conclude that active cell migration between morphologically similar but functionally distinct stromal regions is an integral component regulating differentiation and homeostasis in the steady-state thymus.
Collapse
Affiliation(s)
- Evan F. Lind
- Memorial Sloan-Kettering Cancer Center, New York, NY 10021
| | | | | | - Howard T. Petrie
- Memorial Sloan-Kettering Cancer Center, New York, NY 10021
- Joan and Sanford Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021
| |
Collapse
|
37
|
Mukamoto M, Kodama H. Regulation of early chicken thymocyte proliferation by transforming growth factor-beta from thymic stromal cells and thymocytes. Vet Immunol Immunopathol 2000; 77:121-32. [PMID: 11068070 DOI: 10.1016/s0165-2427(00)00223-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We examined expression of TGF-betas in chicken thymic stromal cells and thymocytes and roles of TGF-betas in thymocyte development within the thymus. Thymic stromal cells expressed TGF-beta 2 and 3 genes but not TGF-beta 4 gene. Thymocytes showed expressions of TGF-beta 2, 3 and 4 genes and each TGF-beta gene was expressed more strongly in CD3- than CD3+ thymocytes. When anti-TGF-beta antibody was added with supernatants of stromal cells into thymocyte culture, only proliferative activity of CD3- thymocytes was enhanced and the cells in S and G2/M compartments of cell cycle increased. These results suggest that TGF-beta which is expressed in the thymus may regulate the ability of immature thymocytes to progress through the cell cycle and to differentiate to CD3+ thymocytes.
Collapse
Affiliation(s)
- M Mukamoto
- Laboratory of Veterinary Immunology, Department of Veterinary Science, College of Agriculture, Osaka Prefecture University, Sakai, 599-8531, Osaka, Japan
| | | |
Collapse
|
38
|
Petrie HT, Tourigny M, Burtrum DB, Livak F. Precursor thymocyte proliferation and differentiation are controlled by signals unrelated to the pre-TCR. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:3094-8. [PMID: 10975821 DOI: 10.4049/jimmunol.165.6.3094] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In-frame rearrangement of the TCR-beta locus and expression of the pre-TCR are compulsory for the production of CD4+8+ thymocytes from CD4-8- precursors. Signals delivered via the pre-TCR are thought to induce the differentiation process as well as the extensive proliferation that accompanies this transition. However, it is equally possible that pre-TCR expression is required for the success of this transition, but does not play a direct role in the inductive process. In the present manuscript we examine this possibility using a variety of normal and genetically modified mouse models. Our evidence shows that differentiation and mitogenesis can both occur independently of pre-TCR expression. However, these processes are absolutely dependent on the presence of normal thymic architecture and cellular composition. These findings are consistent with a checkpoint role for the pre-TCR in regulating the divergence of survival and cell death fates at the CD4-8- to CD4+8+ transition. Further, our data suggest that precursor thymocyte differentiation is induced by other, probably ubiquitous, mechanisms that require the presence of normal thymic cellularity, composition, and architecture.
Collapse
MESH Headings
- Animals
- Cell Death/genetics
- Cell Death/immunology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Cell Division/genetics
- Cell Division/immunology
- Cell Survival/genetics
- Cell Survival/immunology
- Gene Expression Regulation/immunology
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor
- Genes, T-Cell Receptor beta
- Lymphocyte Activation/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitosis/genetics
- Mitosis/immunology
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/deficiency
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/physiology
- Signal Transduction/genetics
- Signal Transduction/immunology
- Stem Cells/immunology
- Stem Cells/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
Collapse
Affiliation(s)
- H T Petrie
- Immunology Program, Memorial Sloan-Kettering Cancer Center, and Joan and Sanford Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021, USA.
| | | | | | | |
Collapse
|
39
|
Su DM, Manley NR. Hoxa3 and pax1 transcription factors regulate the ability of fetal thymic epithelial cells to promote thymocyte development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:5753-60. [PMID: 10820253 DOI: 10.4049/jimmunol.164.11.5753] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thymocyte maturation into T cells depends on interactions between thymocytes and thymic epithelial cells. In this study, we show that mutations in two transcription factors, Hoxa3 and Pax1, act synergistically to cause defective thymic epithelial cell development, resulting in thymic ectopia and hypoplasia. Hoxa3+/-Pax1-/- compound mutant mice exhibited more severe thymus defects than Pax1-/- single mutants. Fetal liver adoptive transfer experiments revealed that the defect resided in radio-resistant stromal cells and not in hematopoietic cells. Compound mutants have fewer MHC class II+ epithelial cells, and the level of MHC expression detected was lower. Thymic epithelial cells in these mutants have reduced ability to promote thymocyte development, causing a specific block in thymocyte maturation at an early stage that resulted in a dramatic reduction in the number of CD4+8+ thymocytes. This phenotype was accompanied by increased apoptosis of CD4+8+ thymocytes and their immediate precursors, CD44-25-(CD3-4-8-) cells. Our results identify a transcriptional regulatory pathway required for thymic epithelial cell development and define multiple roles for epithelial cell regulation of thymocyte maturation at the CD4-8- to CD4+8+ transition.
Collapse
MESH Headings
- Adjuvants, Immunologic/genetics
- Adjuvants, Immunologic/physiology
- Animals
- Antigens, CD/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- CD4-CD8 Ratio
- Cell Death/genetics
- Cell Death/immunology
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/physiology
- Embryonic and Fetal Development/genetics
- Embryonic and Fetal Development/immunology
- Epithelial Cells/immunology
- Epithelial Cells/metabolism
- Gene Deletion
- Histocompatibility Antigens Class II/biosynthesis
- Homeodomain Proteins/genetics
- Homeodomain Proteins/physiology
- Lectins, C-Type
- Lymphocyte Depletion
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Paired Box Transcription Factors
- Phenotype
- Receptors, Antigen, T-Cell/biosynthesis
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/cytology
- Thymus Gland/embryology
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Transcription Factors/genetics
- Transcription Factors/physiology
Collapse
Affiliation(s)
- D M Su
- Institute of Molecular Medicine and Genetics and Department of Pediatrics, Medical College of Georgia, Augusta, GA 30912, USA.
| | | |
Collapse
|
40
|
Lucas PJ, Kim SJ, Melby SJ, Gress RE. Disruption of T cell homeostasis in mice expressing a T cell-specific dominant negative transforming growth factor beta II receptor. J Exp Med 2000; 191:1187-96. [PMID: 10748236 PMCID: PMC2193176 DOI: 10.1084/jem.191.7.1187] [Citation(s) in RCA: 208] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The immune system, despite its complexity, is maintained at a relative steady state. Mechanisms involved in maintaining lymphocyte homeostasis are poorly understood; however, recent availability of transgenic (Tg) and knockout mouse models with altered balance of lymphocyte cell populations suggest that cytokines play a major role in maintaining lymphocyte homeostasis. We show here that transforming growth factor (TGF)-beta plays a critical role in maintaining CD8(+) T cell homeostasis in a Tg mouse model that specifically overexpresses a dominant negative TGF-beta II receptor (DNRII) on T cells. DNRII T cell Tg mice develop a CD8(+) T cell lymphoproliferative disorder resulting in the massive expansion of the lymphoid organs. These CD8(+) T cells are phenotypically "naive" except for the upregulation of the cell surface molecule CD44, a molecule usually associated with memory T cells. Despite their dominance in the peripheral lymphoid organs, CD8(+) T cells appear to develop normally in the thymus, suggesting that TGF-beta exerts its homeostatic control in the peripheral immune system.
Collapse
Affiliation(s)
- Philip J. Lucas
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Seong-Jin Kim
- Laboratory of Cell Regulation and Carcinogenesis, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Spencer J. Melby
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| | - Ronald E. Gress
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892
| |
Collapse
|
41
|
Letterio JJ. Murine models define the role of TGF-beta as a master regulator of immune cell function. Cytokine Growth Factor Rev 2000; 11:81-7. [PMID: 10708955 DOI: 10.1016/s1359-6101(99)00031-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Many members of transforming growth factor-beta (TGF-beta) superfamily, including not only TGF-beta, but also the activins, and bone morphogenetic proteins (BMPs), have been demonstrated to affect the development and function of immune cells. From the proliferation and differentiation of pluripotent stem cells, to the activation and migration of mature lymphoid and myeloid lineages, the TGF-betas have been recognized for their ability to modulate the manner in which such cells respond to stimuli in their environment. Recent studies involving disruption of this pathway in genetically engineered mice now emphasize the importance of this activity and validate functional models predicted by in vitro studies. Phenotypic differences between mice harboring mutations in the TGF-beta1 ligand and the TGF-beta receptor-activated signaling intermediate Smad3 are presented and serve to highlight the valuable role of these in vivo genetic tests of function.
Collapse
Affiliation(s)
- J J Letterio
- Laboratory of Cell Regulation and Carcinogenesis, The National Cancer Institute, NIH, Bethesda, MD 20892-5055, USA.
| |
Collapse
|
42
|
Sempowski GD, Hale LP, Sundy JS, Massey JM, Koup RA, Douek DC, Patel DD, Haynes BF. Leukemia inhibitory factor, oncostatin M, IL-6, and stem cell factor mRNA expression in human thymus increases with age and is associated with thymic atrophy. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:2180-7. [PMID: 10657672 DOI: 10.4049/jimmunol.164.4.2180] [Citation(s) in RCA: 207] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The roles that thymus cytokines might play in regulating thymic atrophy are not known. Reversing thymic atrophy is important for immune reconstitution in adults. We have studied cytokine mRNA steady-state levels in 45 normal human (aged 3 days to 78 years) and 34 myasthenia gravis thymuses (aged 4 to 75 years) during aging, and correlated cytokine mRNA levels with thymic signal joint (sj) TCR delta excision circle (TREC) levels, a molecular marker for active thymopoiesis. LIF, oncostatin M (OSM), IL-6, M-CSF, and stem cell factor (SCF) mRNA were elevated in normal and myasthenia gravis-aged thymuses, and correlated with decreased levels of thymopoiesis, as determined by either decreased keratin-positive thymic epithelial space or decreased thymic sjTRECs. IL-7 is a key cytokine required during the early stages of thymocyte development. Interestingly, IL-7 mRNA expression did not fall with aging in either normal or myasthenia gravis thymuses. In vivo administration of LIF, OSM, IL-6, or SCF, but not M-CSF, i.p. to mice over 3 days induced thymic atrophy with loss of CD4+, CD8+ cortical thymocytes. Taken together, these data suggest a role for thymic cytokines in the process of thymic atrophy.
Collapse
Affiliation(s)
- G D Sempowski
- Department of Medicine, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Prud'homme GJ, Piccirillo CA. The inhibitory effects of transforming growth factor-beta-1 (TGF-beta1) in autoimmune diseases. J Autoimmun 2000; 14:23-42. [PMID: 10648114 DOI: 10.1006/jaut.1999.0339] [Citation(s) in RCA: 216] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The importance of transforming growth factor-beta-1 (TGF-beta1) in immunoregulation and tolerance has been increasingly recognized. It is now proposed that there are populations of regulatory T cells (T-reg), some designated T-helper type 3 (Th3), that exert their action primarily by secreting this cytokine. Here, we emphasize the following concepts: (1) TGF-beta1 has multiple suppressive actions on T cells, B cells, macrophages, and other cells, and increased TGF-beta1 production correlates with protection and/or recovery from autoimmune diseases; (2) TGF-beta1 and CTLA-4 are molecules that work together to terminate immune responses; (3) Th0, Th1 and Th2 clones can all secrete TGF-beta1 upon cross-linking of CTLA-4 (the functional significance of this in autoimmune diseases has not been reported, but TGF-beta1-producing regulatory T-cell clones can produce type 1 inflammatory cytokines); (4) TGF-beta1 may play a role in the passage from effector to memory T cells; (5) TGF-beta1 acts with some other inhibitory molecules to maintain a state of tolerance, which is most evident in immunologically privileged sites, but may also be important in other organs; (6) TGF-beta1 is produced by many cell types, is always present in the plasma (in its latent form) and permeates all organs, binding to matrix components and creating a reservoir of this immunosuppressive molecule; and (7) TGF-beta1 downregulates adhesion molecules and inhibits adhesion of leukocytes to endothelial cells. We propose that rather than being passive targets of autoimmunity, tissues and organs actively suppress autoreactive lymphocytes. We review the beneficial effects of administering TGF-beta1 in several autoimmune diseases, and show that it can be effectively administered by a somatic gene therapy approach, which results in depressed inflammatory cytokine production and increased endogenous regulatory cytokine production.
Collapse
Affiliation(s)
- G J Prud'homme
- Department of Pathology, McGill University, 3775 University St., Room B13, Montreal, Quebéc, H3A 2B4, Canada.
| | | |
Collapse
|
44
|
Lee J, Desiderio S. Cyclin A/CDK2 regulates V(D)J recombination by coordinating RAG-2 accumulation and DNA repair. Immunity 1999; 11:771-81. [PMID: 10626899 DOI: 10.1016/s1074-7613(00)80151-x] [Citation(s) in RCA: 96] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Accumulation of the V(D)J recombinase protein RAG-2 is restricted to G0/G1 cells by phosphorylation-mediated degradation at the G1-S boundary. Here cyclin A/CDK2 is shown to oppose RAG-2 accumulation; conversely, RAG-2 is induced by p27Kip1 and related CDK inhibitors. Coinduction of RAG-2 and G1 delay by p27Kip1 is accompanied by strong stimulation of V(D)J recombination. Unexpectedly, induction of RAG-2 accumulation in the absence of G1 delay has no effect on recombination frequency. p27Kip1 may stimulate V(D)J recombination by coordinating accumulation of RAG-2 with prolongation of G1, when nonhomologous end joining is preferentially active. Consistent with this, enforced expression of RAG-2 throughout cell cycle is associated with accumulation of aberrant recombination products reminiscent of those formed in the absence of nonhomologous end joining.
Collapse
Affiliation(s)
- J Lee
- Department of Molecular Biology and Genetics and Howard Hughes Medical Institute, The Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | |
Collapse
|
45
|
Affiliation(s)
- S Desiderio
- Department of Molecular Biology and Genetics, Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | |
Collapse
|
46
|
Abstract
Thymus development and microenvironment organization require stage- and site-specific cross-talk between thymocyte and stroma. In this study we have used recombinase-activating gene-deficient (RAG-2(-/-)) mice to analyze regulated gene expression both in thymocytes and stromal cells following injection of anti-CD3 monoclonal antibodies as inducer of thymus development. We show that IFN-gamma, TNF-alpha and lymphotactin are transcriptionally regulated in thymocytes, whereas cytoskeletal keratin 14, IL-1alpha and TNF-alpha are regulated in the stroma, quantitatively reproducing the variations associated with beta selection of thymocytes. In addition, RAG-2(-/-) thymus development is associated with entry of epithelial cells into the cell cycle. The histochemical evidence that expanded RAG-2(-/-) thymus becomes undistinguishable from wild-type cortex further suggests that cross-talk phenomena occurring during beta selection of thymocyte are reproduced in this system.
Collapse
Affiliation(s)
- S Porcellini
- Dipartimento di Biologia e Genetica per le Scienze Mediche, Università di Milano at Department of Biological and Technological Research (DIBIT), San Raffaele Scientific Institute (HSR), Milano, Italy
| | | | | |
Collapse
|
47
|
Panigada M, Porcellini S, Sutti F, Doneda L, Pozzoli O, Consalez GG, Guttinger M, Grassi F. GKLF in thymus epithelium as a developmentally regulated element of thymocyte-stroma cross-talk. Mech Dev 1999; 81:103-13. [PMID: 10330488 DOI: 10.1016/s0925-4773(98)00237-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Gut-enriched Krüppel-like factor (GKLF) is a transcriptional regulator expressed in differentiated epithelia. We identified GKLF transcript as a regulated element in thymic epithelium of recombinase-deficient mice during thymus development induced by anti-CD3 antibody injection. This treatment recapitulates the organogenetic process depending on productive rearrangement of T cell receptor (TCR) beta gene with thymocytes expansion and acquisition of the CD4+8+ double positive phenotype. In wildtype mice, GKLF is expressed very early in embryogenesis and becomes intensely up-regulated in thymus epithelium at day 18 of gestation when TCR beta expressing cells have selectively expanded and express both CD4 and CD8. The results presented here suggest that thymocytes may regulate GKLF transcriptionally in the cortical epithelium at the developmental check-point controlled by TCR beta gene rearrangement. Furthermore, GKLF expression in hematopoietic stroma might suggest the thus far uncharacterised participation of this factor in hematopoiesis.
Collapse
Affiliation(s)
- M Panigada
- Department of Biological and Technological Research (DIBIT), San Raffaele Scientific Institute (HSR), Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Takahama Y, Ohishi K, Tokoro Y, Sugawara T, Yoshimura Y, Okabe M, Kinoshita T, Takeda J. Functional competence of T cells in the absence of glycosylphosphatidylinositol-anchored proteins caused by T cell-specific disruption of the Pig-a gene. Eur J Immunol 1998; 28:2159-66. [PMID: 9692885 DOI: 10.1002/(sici)1521-4141(199807)28:07<2159::aid-immu2159>3.0.co;2-b] [Citation(s) in RCA: 93] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
T lymphocytes express various glycosylphosphatidylinositol (GPI)-anchored surface proteins, such as Thy-1 and Ly-6A. However, functional contribution of GPI-anchored proteins in T cell activation is as yet poorly understood. Here we report the generation of mutant mice deficient in the expression of GPI-anchored molecules exclusively in their T cells. We established mice carrying three identically oriented lox-P sites within the Pig-a gene, which encodes a component essential for the initial step of GPI anchor biosynthesis. These mice were crossed with mice carrying the Cre recombinase gene driven by the T cell-specific p56lck proximal promoter. Offspring carrying both the lox-P-containing Pig-a gene and the Cre transgene exhibited almost complete loss of the surface expression of GPI-anchored molecules on peripheral T cells. Interestingly, those T cells deficient in GPI-anchored molecules were capable of responding to T cell receptor stimulation in vitro and in vivo. These results indicate that T cells lacking the expression of GPI-anchored molecules are functionally competent in exerting TCR-mediated immune responses.
Collapse
Affiliation(s)
- Y Takahama
- Department of Immunology, Institute of Basic Medical Sciences, University of Tsukuba, Ibaraki, Japan
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Sfikakis PP, Kostomitsopoulos N, Kittas C, Stathopoulos J, Karayannacos P, Dellia-Sfikakis A, Mitropoulos D. Tamoxifen exerts testosterone-dependent and independent effects on thymic involution. INTERNATIONAL JOURNAL OF IMMUNOPHARMACOLOGY 1998; 20:305-12. [PMID: 9754678 DOI: 10.1016/s0192-0561(98)00034-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Circulating testosterone concentrations and seminal vesicles weights, as well as thymus and spleen weights and histology were assessed in male Wistar rats from the infantile to post-pubertal period. The widely used anti-estrogenic agent tamoxifen was then administered in adult intact and castrated male rats and its long-term effects on thymic involution and splenic growth were examined. The results showed that: (1) age-related involution of the male thymus from the juvenile period through puberty to post-puberty depends on the rising testosterone levels and represents mainly a decrease of thymic lymphoid-cell elements; (2) tamoxifen administration reverses thymic involution in intact adult male rats and this effect is related to a dose-dependent, tamoxifen-induced castration and decrease of testosterone levels; (3) the changes of circulating testosterone levels, either resulting from maturity, or induced by tamoxifen or by castration, have a minimal effect on splenic growth and weight; and (4) in contrast to intact animals, administration of tamoxifen at pharmacological doses to adult castrated rats results in thymic regression. Underscoring the critical role of testosterone on thymic involution, these findings show that tamoxifen is able to reverse ageing changes in the thymus by suppressing testosterone production, while conversely, exerts thymolytic effects in the absence of androgens.
Collapse
Affiliation(s)
- P P Sfikakis
- First Department of Propaedeutic Medicine, Athens University Medical School, Greece.
| | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
The transforming growth factor beta (TGF-beta) family of proteins are a set of pleiotropic secreted signaling molecules with unique and potent immunoregulatory properties. TGF-beta 1 is produced by every leukocyte lineage, including lymphocytes, macrophages, and dendritic cells, and its expression serves in both autocrine and paracrine modes to control the differentiation, proliferation, and state of activation of these immune cells. TGF-beta can modulate expression of adhesion molecules, provide a chemotactic gradient for leukocytes and other cells participating in an inflammatory response, and inhibit them once they have become activated. Increased production and activation of latent TGF-beta have been linked to immune defects associated with malignancy and autoimmune disorders, to susceptibility to opportunistic infection, and to the fibrotic complications associated with chronic inflammatory conditions. In addition to these roles in disease pathogenesis, TGF-beta is now established as a principal mediator of oral tolerance and can be recognized as the sine qua non of a unique subset of effector cells that are induced in this process. The accumulated knowledge gained through extensive in vitro functional analyses and from in vivo animal models, including newly established TGF-beta gene knockout and transgenic mice, supports the concept that clinical therapies based on modulation of this cytokine represent an important new approach to the treatment of disorders of immune function.
Collapse
Affiliation(s)
- J J Letterio
- Laboratory of Chemoprevention, National Cancer Institute, Bethesda, Maryland, USA
| | | |
Collapse
|