1
|
Park CS, Guan J, Rhee P, Gonzalez F, Lee HS, Park JH, Coscoy L, Robey EA, Shastri N, Sadegh-Nasseri S. Fam49b dampens TCR signal strength to regulate survival of positively selected thymocytes and peripheral T cells. eLife 2024; 13:e76940. [PMID: 39158947 PMCID: PMC11333044 DOI: 10.7554/elife.76940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Accepted: 07/16/2024] [Indexed: 08/20/2024] Open
Abstract
The fate of developing T cells is determined by the strength of T cell receptor (TCR) signal they receive in the thymus. This process is finely regulated through the tuning of positive and negative regulators in thymocytes. The Family with sequence similarity 49 member B (Fam49b) protein is a newly discovered negative regulator of TCR signaling that has been shown to suppress Rac-1 activity in vitro in cultured T cell lines. However, the contribution of Fam49b to the thymic development of T cells is unknown. To investigate this important issue, we generated a novel mouse line deficient in Fam49b (Fam49b-KO). We observed that Fam49b-KO double positive (DP) thymocytes underwent excessive negative selection, whereas the positive selection stage was unaffected. Fam49b deficiency impaired the survival of single positive thymocytes and peripheral T cells. This altered development process resulted in significant reductions in CD4 and CD8 single-positive thymocytes as well as peripheral T cells. Interestingly, a large proportion of the TCRγδ+ and CD8αα+TCRαβ+ gut intraepithelial T lymphocytes were absent in Fam49b-KO mice. Our results demonstrate that Fam49b dampens thymocytes TCR signaling in order to escape negative selection during development, uncovering the function of Fam49b as a critical regulator of the selection process to ensure normal thymocyte development and peripheral T cells survival.
Collapse
Affiliation(s)
- Chan-Su Park
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
- Department of Pharmaceutics, College of Pharmacy, Chungbuk National UniversityCheongjuRepublic of Korea
| | - Jian Guan
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Peter Rhee
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | - Federico Gonzalez
- Department of Nutritional Sciences and Toxicology, University of California,BerkeleyBerkeleyUnited States
| | - Hee-sung Lee
- Department of Pharmaceutics, College of Pharmacy, Chungbuk National UniversityCheongjuRepublic of Korea
| | - Ji-hyun Park
- Department of Pharmaceutics, College of Pharmacy, Chungbuk National UniversityCheongjuRepublic of Korea
| | - Laurent Coscoy
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Ellen A Robey
- Department of Molecular and Cell Biology, University of California, BerkeleyBerkeleyUnited States
| | - Nilabh Shastri
- Department of Pathology, The Johns Hopkins University School of MedicineBaltimoreUnited States
| | | |
Collapse
|
2
|
Choi SM, Park HJ, Choi EA, Jung KC, Lee JI. CD1b glycoprotein, a crucial marker of thymocyte development during T cell maturation in cynomolgus monkeys. Sci Rep 2023; 13:14388. [PMID: 37658106 PMCID: PMC10474046 DOI: 10.1038/s41598-023-41708-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/30/2023] [Indexed: 09/03/2023] Open
Abstract
Phenotypic markers that denote different developmental stages of thymocytes are important for understanding T cell development in the thymus. Here, we show that CD1b is a critical discriminator of thymocyte maturation stage in cynomolgus monkeys. CD1b was expressed by immature thymocytes prior to β-selection, and its expression decreased as cells became fully mature in the thymus. MHC-I expression was lowest at the CD3loCD1b+ immature double-positive (DP) stage, while the ratio of CD1d:MHC-I expression was significantly higher at this stage than at other developmental stages. PLZF was expressed by < 0.2% of thymocytes; most PLZF+ thymocytes were CD3-/loCD1b+ immature DP thymocytes with the potential to produce IL-4. EOMES+ thymocytes, which accounted for > 2% of total thymocytes, were mostly CD3+CD1b- mature thymocytes and predominantly of the CD8 single-positive (SP) lineage. An unconventional CD8+ T cell subset expressing the NKG2AC+CXCR3+ innate-like T cell marker was identified within the EOMES+ CD8 SP lineage; these cells exhibited a memory phenotype. Taken together, these findings show that CD1b is a valuable discriminatory marker of thymocyte development. The data presented herein can be used to characterize the features of PLZF- and EOMES-associated unconventional T cells in the thymus.
Collapse
Affiliation(s)
- Sung Min Choi
- Graduate Course of Translational Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Hi Jung Park
- Graduate Course of Translational Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Eun A Choi
- Graduate Course of Translational Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
| | - Kyeong Cheon Jung
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Department of Pathology, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, 03080, Republic of Korea
| | - Jae Il Lee
- Transplantation Research Institute, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
- Department of Medicine, Seoul National University College of Medicine, Seoul, 03080, Republic of Korea.
| |
Collapse
|
3
|
Yang X, Wang X, Lei L, Su Y, Zou Y, Liu H, Jiao A, Zhang C, Liu J, Li W, Ding R, Zhou X, Shi L, Zhang D, Sun C, Zhang B. Arid1a promotes thymocyte development through β-selection-dependent and β-selection-independent mechanisms. Immunology 2021; 165:402-413. [PMID: 34921692 DOI: 10.1111/imm.13440] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/15/2021] [Accepted: 12/15/2021] [Indexed: 01/16/2023] Open
Abstract
Early T-cell development from CD4- CD8- double-negative (DN) stage to CD4+ CD8+ double-positive (DP) stage in the thymus is regulated through multiple steps involving a batch of sequentially expressed factors. Our preliminary data and a recent report showed that AT-rich interaction domain 1A (Arid1a) is required for the transition from DN to DP stages, but the mechanism is not fully understood. In this study, we consolidated that conditional deletion of Arid1a in T-cell lineage intrinsically caused developmental blocks from DN3 to DN4 stages, as well as from DN4 to DP stages using both in vivo adoptive T-cell transfer model and in vitro culture system. The expression of intracellular TCRβ is significantly decreased in Arid1a-deficient DN4 cells compared with WT cells. OT1 transgenic TCR can rescue the defect in the transition from DN3 to DN4 stages, but not from DN to DP stages. Furthermore, we observed a comparable or stronger proliferation capacity accompanied by a significant increase in cell death in Arid1a-/- DP cells compared with that in WT controls. RNA-Seq analysis shows a significant enrichment of apoptotic pathway within differentially expressed genes between Arid1a-/- and WT DP cells, including the upregulation of Bim, Casp3 and Trp53 and the downregulation of Rorc, Bcl-XL and Mcl1. Therefore, our study reveals a novel mechanism that Arid1a controls early T-cell development by maintaining intracellular TCRβ expression-mediated β-selection and activating parallel cell survival pathways.
Collapse
Affiliation(s)
- Xiaofeng Yang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, China.,Xi'an Key Laboratory of Immune Related Diseases, Xi'an, China
| | - Xin Wang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Lei Lei
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, China.,Xi'an Key Laboratory of Immune Related Diseases, Xi'an, China
| | - Yanhong Su
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Yujing Zou
- Duke University Medical Center, Durham, North Carolina, USA
| | - Haiyan Liu
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Anjun Jiao
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Jun Liu
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Wenhua Li
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Renyi Ding
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Xiaobo Zhou
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Lin Shi
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Dan Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China
| | - Chenming Sun
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Xi'an Key Laboratory of Immune Related Diseases, Xi'an, China
| | - Baojun Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences; Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, China.,Institute of Infection and Immunity, Translational Medicine Institute, Xi'an Jiaotong University Health Science Center, Xi'an, China.,Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University), Ministry of Education, Xi'an, China.,Xi'an Key Laboratory of Immune Related Diseases, Xi'an, China
| |
Collapse
|
4
|
Han J, Zúñiga-Pflücker JC. A 2020 View of Thymus Stromal Cells in T Cell Development. THE JOURNAL OF IMMUNOLOGY 2021; 206:249-256. [PMID: 33397738 DOI: 10.4049/jimmunol.2000889] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022]
Abstract
The thymus is an intricate primary lymphoid organ, wherein bone marrow-derived lymphoid progenitor cells are induced to develop into functionally competent T cells that express a diverse TCR repertoire, which is selected to allow for the recognition of foreign Ags while avoiding self-reactivity or autoimmunity. Thymus stromal cells, which can include all non-T lineage cells, such as thymic epithelial cells, endothelial cells, mesenchymal/fibroblast cells, dendritic cells, and B cells, provide signals that are essential for thymocyte development as well as for the homeostasis of the thymic stroma itself. In this brief review, we focus on the key roles played by thymic stromal cells during early stages of T cell development, such as promoting the homing of thymic-seeding progenitors, inducing T lineage differentiation, and supporting thymocyte survival and proliferation. We also discuss recent advances on the transcriptional regulation that govern thymic epithelial cell function as well as the cellular and molecular changes that are associated with thymic involution and regeneration.
Collapse
Affiliation(s)
- Jianxun Han
- Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada; and
| | - Juan Carlos Zúñiga-Pflücker
- Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada; and.,Department of Immunology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
5
|
Kim M, Kim SD, Kim KI, Jeon EH, Kim MG, Lim YR, Lkhagva-Yondon E, Oh Y, Na K, Chung YC, Jin BK, Song YS, Jeon MS. Dynamics of T Lymphocyte between the Periphery and the Brain from the Acute to the Chronic Phase Following Ischemic Stroke in Mice. Exp Neurobiol 2021; 30:155-169. [PMID: 33707347 PMCID: PMC8118758 DOI: 10.5607/en20062] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 03/03/2021] [Accepted: 03/03/2021] [Indexed: 11/25/2022] Open
Abstract
Stroke causes systemic immunosuppression. T lymphocytes are involved in infarct size in the early stages of stroke. However, the phenotypes of T lymphocytes and their functions in peripheral immune organs and the brain have not been well analyzed in the acute and chronic phases of stroke. Here, we investigated pathological phenotypic alterations in the systemic immune response, especially changes in T lymphocytes, from one day to six months after ischemic stroke in mice. Impairment in thymocyte numbers, development, proliferation, and apoptosis were observed for up to two weeks. The number of mature T cells in the spleen and blood decreased and showed reduced interferon-γ production. Increased numbers of CD4-CD8-CD3+ double-negative T cells were observed in the mouse brain during the early stages of stroke, whereas interleukin (IL)-10+Foxp3+ regulatory T lymphocytes increased from two weeks during the chronic phase. These phenotypes correlated with body weight and neurological severity scores. The recovery of T lymphocyte numbers and increases in IL-10+Foxp3+ regulatory T lymphocytes may be important for long-term neurological outcomes. Dynamic changes in T lymphocytes between the acute and chronic phases may play different roles in pathogenesis and recovery. This study provides fundamental information regarding the T lymphocyte alterations from the brain to the peripheral immune organs following stroke.
Collapse
Affiliation(s)
- Minha Kim
- Translational Research Center, Department of Molecular Biomedicine, IRIMS, and College of Medicine, Inha University, Incheon 22332, Korea
| | - So-Dam Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea
| | - Kyoung In Kim
- Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Eun Hae Jeon
- Translational Research Center, Department of Molecular Biomedicine, IRIMS, and College of Medicine, Inha University, Incheon 22332, Korea.,Program in Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22332, Korea
| | - Min Gee Kim
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea
| | - Yu-Ree Lim
- Translational Research Center, Department of Molecular Biomedicine, IRIMS, and College of Medicine, Inha University, Incheon 22332, Korea
| | - Enkhmaa Lkhagva-Yondon
- Translational Research Center, Department of Molecular Biomedicine, IRIMS, and College of Medicine, Inha University, Incheon 22332, Korea.,Program in Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22332, Korea
| | - Yena Oh
- Translational Research Center, Department of Molecular Biomedicine, IRIMS, and College of Medicine, Inha University, Incheon 22332, Korea
| | - Kwangmin Na
- Translational Research Center, Department of Molecular Biomedicine, IRIMS, and College of Medicine, Inha University, Incheon 22332, Korea
| | - Young Cheul Chung
- Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Byung Kwan Jin
- Department of Biochemistry & Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Yun Seon Song
- College of Pharmacy, Sookmyung Women's University, Seoul 04310, Korea
| | - Myung-Shin Jeon
- Translational Research Center, Department of Molecular Biomedicine, IRIMS, and College of Medicine, Inha University, Incheon 22332, Korea.,Program in Biomedical Science and Engineering, Graduate School, Inha University, Incheon 22332, Korea.,Convergent Research Center for Metabolism and Immunoregulation, Inha University, Incheon 22212, Korea
| |
Collapse
|
6
|
Paganelli F, Lonetti A, Anselmi L, Martelli AM, Evangelisti C, Chiarini F. New advances in targeting aberrant signaling pathways in T-cell acute lymphoblastic leukemia. Adv Biol Regul 2019; 74:100649. [PMID: 31523031 DOI: 10.1016/j.jbior.2019.100649] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 08/24/2019] [Accepted: 09/03/2019] [Indexed: 10/26/2022]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive disorder characterized by malignant transformation of immature progenitors primed towards T-cell development. Over the past 15 years, advances in the molecular characterization of T-ALL have uncovered oncogenic key drivers and crucial signaling pathways of this disease, opening new chances for the development of novel therapeutic strategies. Currently, T-ALL patients are still treated with aggressive therapies, consisting of high dose multiagent chemotherapy. To minimize and overcome the unfavorable effects of these regimens, it is critical to identify innovative targets and test selective inhibitors of such targets. Major efforts are being made to develop small molecules against deregulated signaling pathways, which sustain T-ALL cell growth, survival, metabolism, and drug-resistance. This review will focus on recent improvements in the understanding of the signaling pathways involved in the pathogenesis of T-ALL and on the challenging opportunities for T-ALL targeted therapies.
Collapse
Affiliation(s)
- Francesca Paganelli
- Institute of Molecular Genetics, Luigi Luca Cavalli-Sforza-CNR National Research Council of Italy, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Annalisa Lonetti
- "Giorgio Prodi" Cancer Research Center, University of Bologna, Bologna, Italy
| | - Laura Anselmi
- Department of Biomedical, Metabolic, and Neural Sciences, Section of Morphology, Signal Transduction Unit, University of Modena and Reggio Emilia, Modena, Italy
| | - Alberto M Martelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Camilla Evangelisti
- Institute of Molecular Genetics, Luigi Luca Cavalli-Sforza-CNR National Research Council of Italy, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Francesca Chiarini
- Institute of Molecular Genetics, Luigi Luca Cavalli-Sforza-CNR National Research Council of Italy, Bologna, Italy; IRCCS Istituto Ortopedico Rizzoli, Bologna, Italy.
| |
Collapse
|
7
|
Joseph C, Klibi J, Amable L, Comba L, Cascioferro A, Delord M, Parietti V, Lenoir C, Latour S, Lucas B, Viret C, Toubert A, Benlagha K. TCR density in early iNKT cell precursors regulates agonist selection and subset differentiation in mice. Eur J Immunol 2019; 49:894-910. [DOI: 10.1002/eji.201848010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/27/2019] [Accepted: 03/21/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Claudine Joseph
- INSERM, UMR‐1160Institut Universitaire d'Hématologie Paris France
- Université Paris DiderotSorbonne Paris Cité Paris France
| | - Jihene Klibi
- INSERM, UMR‐1160Institut Universitaire d'Hématologie Paris France
- Université Paris DiderotSorbonne Paris Cité Paris France
| | - Ludivine Amable
- INSERM, UMR‐1160Institut Universitaire d'Hématologie Paris France
- Université Paris DiderotSorbonne Paris Cité Paris France
| | - Lorenzo Comba
- INSERM, UMR‐1160Institut Universitaire d'Hématologie Paris France
- Université Paris DiderotSorbonne Paris Cité Paris France
| | | | - Marc Delord
- Plateforme de Bio‐informatique et Bio statistiqueInstitut Universitaire d'HématologieUniversité Paris Diderot Sorbonne Paris Cité Paris France
| | - Veronique Parietti
- Département d'Expérimentation AnimaleInstitut Universitaire d'Hématologie Paris France
- Université Paris Diderot Sorbonne Paris Cité Paris France
| | - Christelle Lenoir
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection Paris France
- Imagine InstitutUniversité Paris Diderot Sorbonne Paris Cité Paris France
| | - Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection Paris France
- Imagine InstitutUniversité Paris Diderot Sorbonne Paris Cité Paris France
| | - Bruno Lucas
- Institut Cochin, Centre National de la Recherche Scientifique UMR8104, INSERM U1016Université Paris Descartes Paris France
| | - Christophe Viret
- CIRI, International Center for Infectiology ResearchUniversité de Lyon Lyon France
- INSERM U1111 Lyon France
- CNRS UMR5308 Lyon France
| | - Antoine Toubert
- INSERM, UMR‐1160Institut Universitaire d'Hématologie Paris France
- Université Paris DiderotSorbonne Paris Cité Paris France
| | - Kamel Benlagha
- INSERM, UMR‐1160Institut Universitaire d'Hématologie Paris France
- Université Paris DiderotSorbonne Paris Cité Paris France
| |
Collapse
|
8
|
Nacka-Aleksić M, Stojanović M, Pilipović I, Stojić-Vukanić Z, Kosec D, Leposavić G. Strain differences in thymic atrophy in rats immunized for EAE correlate with the clinical outcome of immunization. PLoS One 2018; 13:e0201848. [PMID: 30086167 PMCID: PMC6080797 DOI: 10.1371/journal.pone.0201848] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Accepted: 07/22/2018] [Indexed: 01/03/2023] Open
Abstract
An accumulating body of evidence suggests that development of autoimmune pathologies leads to thymic dysfunction and changes in peripheral T-cell compartment, which, in turn, perpetuate their pathogenesis. To test this hypothesis, thymocyte differentiation/maturation in rats susceptible (Dark Agouti, DA) and relatively resistant (Albino Oxford, AO) to experimental autoimmune encephalomyelitis (EAE) induction was examined. Irrespective of strain, immunization for EAE (i) increased the circulating levels of IL-6, a cytokine causally linked with thymic atrophy, and (ii) led to thymic atrophy reflecting partly enhanced thymocyte apoptosis associated with downregulated thymic IL-7 expression. Additionally, immunization diminished the expression of Thy-1, a negative regulator of TCRαβ-mediated signaling and activation thresholds, on CD4+CD8+ TCRαβlo/hi thymocytes undergoing selection and thereby impaired thymocyte selection/survival. This diminished the generation of mature CD4+ and CD8+ single positive TCRαβhi thymocytes and, consequently, CD4+ and CD8+ recent thymic emigrants. In immunized rats, thymic differentiation of natural regulatory CD4+Foxp3+CD25+ T cells (nTregs) was particularly affected reflecting a diminished expression of IL-7, IL-2 and IL-15. The decline in the overall thymic T-cell output and nTreg generation was more pronounced in DA than AO rats. Additionally, differently from immunized AO rats, in DA ones the frequency of CD28- cells secreting cytolytic enzymes within peripheral blood CD4+ T lymphocytes increased, as a consequence of thymic atrophy-related replicative stress (mirrored in CD4+ cell memory pool expansion and p16INK4a accumulation). The higher circulating level of TNF-α in DA compared with AO rats could also contribute to this difference. Consistently, higher frequency of cytolytic CD4+ granzyme B+ cells (associated with greater tissue damage) was found in spinal cord of immunized DA rats compared with their AO counterparts. In conclusion, the study indicated that strain differences in immunization-induced changes in thymopoiesis and peripheral CD4+CD28- T-cell generation could contribute to rat strain-specific clinical outcomes of immunization for EAE.
Collapse
Affiliation(s)
- Mirjana Nacka-Aleksić
- Department of Physiology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| | - Marija Stojanović
- Department of Physiology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| | - Ivan Pilipović
- Immunology Research Centre “Branislav Janković”, Institute of Virology, Vaccines and Sera “Torlak”, Belgrade, Serbia
| | - Zorica Stojić-Vukanić
- Department of Microbiology and Immunology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
| | - Duško Kosec
- Immunology Research Centre “Branislav Janković”, Institute of Virology, Vaccines and Sera “Torlak”, Belgrade, Serbia
| | - Gordana Leposavić
- Department of Physiology, University of Belgrade - Faculty of Pharmacy, Belgrade, Serbia
- * E-mail:
| |
Collapse
|
9
|
Wiede F, Dudakov JA, Lu KH, Dodd GT, Butt T, Godfrey DI, Strasser A, Boyd RL, Tiganis T. PTPN2 regulates T cell lineage commitment and αβ versus γδ specification. J Exp Med 2017; 214:2733-2758. [PMID: 28798028 PMCID: PMC5584121 DOI: 10.1084/jem.20161903] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 05/26/2017] [Accepted: 06/28/2017] [Indexed: 01/18/2023] Open
Abstract
During early thymocyte development, coordinated JAK/STAT5 and SFK/pre-TCR signaling is critical for T cell lineage commitment and αβ versus γδ specification. Wiede et al. show a role for the tyrosine phosphatase PTPN2 in attenuating SRC family kinase LCK and STAT5 signaling to regulate αβ and γδ T cell development. In the thymus, hematopoietic progenitors commit to the T cell lineage and undergo sequential differentiation to generate diverse T cell subsets, including major histocompatibility complex (MHC)–restricted αβ T cell receptor (TCR) T cells and non–MHC-restricted γδ TCR T cells. The factors controlling precursor commitment and their subsequent maturation and specification into αβ TCR versus γδ TCR T cells remain unclear. Here, we show that the tyrosine phosphatase PTPN2 attenuates STAT5 (signal transducer and activator of transcription 5) signaling to regulate T cell lineage commitment and SRC family kinase LCK and STAT5 signaling to regulate αβ TCR versus γδ TCR T cell development. Our findings identify PTPN2 as an important regulator of critical checkpoints that dictate the commitment of multipotent precursors to the T cell lineage and their subsequent maturation into αβ TCR or γδ TCR T cells.
Collapse
Affiliation(s)
- Florian Wiede
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia .,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Jarrod A Dudakov
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Kun-Hui Lu
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Garron T Dodd
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Tariq Butt
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia.,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Dale I Godfrey
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia.,Department of Microbiology and Immunology and Peter Doherty Institute for Infection and Immunity, University of Melbourne, Parkville, Victoria, Australia
| | - Andreas Strasser
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.,The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Richard L Boyd
- Department of Anatomy and Developmental Biology, Monash University, Clayton, Victoria, Australia
| | - Tony Tiganis
- Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia .,Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.,Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| |
Collapse
|
10
|
Lucas B, White AJ, Parnell SM, Henley PM, Jenkinson WE, Anderson G. Progressive Changes in CXCR4 Expression That Define Thymocyte Positive Selection Are Dispensable For Both Innate and Conventional αβT-cell Development. Sci Rep 2017; 7:5068. [PMID: 28698642 PMCID: PMC5505955 DOI: 10.1038/s41598-017-05182-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 05/24/2017] [Indexed: 11/09/2022] Open
Abstract
The ordered migration of immature thymocytes through thymic microenvironments generates both adaptive MHC restricted αβT-cells and innate CD1d-restricted iNKT-cells. While several chemokine receptors and ligands control multiple stages of this process, their involvement during early thymocyte development often precludes direct analysis of potential roles during later developmental stages. For example, because of early lethality of CXCR4-/- mice, and stage-specific requirements for CXCR4 in thymus colonisation and pre-TCR mediated selection, its role in thymic positive selection is unclear. Here we have examined CXCR4-CXCL12 interactions during the maturation of CD4+CD8+ thymocytes, including downstream stages of iNKT and αβT-cell development. We show CXCL12 expression is a common feature of cortical thymic epithelial cells, indicating widespread availability throughout the cortex. Moreover, CXCR4 expression by CD4+CD8+ pre-selection thymocytes is progressively downregulated following both MHC and CD1d-restricted thymic selection events. However, using CD4Cre-mediated deletion to bypass its involvement in CD4-CD8- thymocyte development, we show CXCR4 is dispensable for the maintenance and intrathymic positioning of CD4+CD8+ thymocytes, and their ability to generate mature αβT-cells and CD1d-restricted iNKT-cells. Collectively, our data define dynamic changes in CXCR4 expression as a marker for intrathymic selection events, and show its role in T-cell development is restricted to pre-CD4+CD8+ stages.
Collapse
Affiliation(s)
- Beth Lucas
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, B15 2TT, England
| | - Andrea J White
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, B15 2TT, England
| | - Sonia M Parnell
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, B15 2TT, England
| | - Peter M Henley
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, B15 2TT, England
| | - William E Jenkinson
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, B15 2TT, England
| | - Graham Anderson
- Institute for Immunology and Immunotherapy, College of Medical and Dental Sciences, Medical School, University of Birmingham, Birmingham, B15 2TT, England.
| |
Collapse
|
11
|
Oliveira ML, Akkapeddi P, Alcobia I, Almeida AR, Cardoso BA, Fragoso R, Serafim TL, Barata JT. From the outside, from within: Biological and therapeutic relevance of signal transduction in T-cell acute lymphoblastic leukemia. Cell Signal 2017. [PMID: 28645565 DOI: 10.1016/j.cellsig.2017.06.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematological cancer that arises from clonal expansion of transformed T-cell precursors. In this review we summarize the current knowledge on the external stimuli and cell-intrinsic lesions that drive aberrant activation of pivotal, pro-tumoral intracellular signaling pathways in T-cell precursors, driving transformation, leukemia expansion, spread or resistance to therapy. In addition to their pathophysiological relevance, receptors and kinases involved in signal transduction are often attractive candidates for targeted drug development. As such, we discuss also the potential of T-ALL signaling players as targets for therapeutic intervention.
Collapse
Affiliation(s)
- Mariana L Oliveira
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Padma Akkapeddi
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Isabel Alcobia
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal; Instituto de Histologia e Biologia do Desenvolvimento, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Afonso R Almeida
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Bruno A Cardoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Rita Fragoso
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - Teresa L Serafim
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal
| | - João T Barata
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Portugal.
| |
Collapse
|
12
|
Soluble γc cytokine receptor suppresses IL-15 signaling and impairs iNKT cell development in the thymus. Sci Rep 2016; 6:36962. [PMID: 27833166 PMCID: PMC5105068 DOI: 10.1038/srep36962] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 10/21/2016] [Indexed: 12/13/2022] Open
Abstract
The soluble γc protein (sγc) is a naturally occurring splice isoform of the γc cytokine receptor that is produced by activated T cells and inhibits γc cytokine signaling. Here we show that sγc expression is also highly upregulated in immature CD4+CD8+ thymocytes but then downregulated in mature thymocytes. These results indicate a developmentally controlled mechanism for sγc expression and suggest a potential role for sγc in regulating T cell development in the thymus. Indeed, sγc overexpression resulted in significantly reduced thymocyte numbers and diminished expansion of immature thymocytes, concordant to its role in suppressing signaling by IL-7, a critical γc cytokine in early thymopoiesis. Notably, sγc overexpression also impaired generation of iNKT cells, resulting in reduced iNKT cell percentages and numbers in the thymus. iNKT cell development requires IL-15, and we found that sγc interfered with IL-15 signaling to suppress iNKT cell generation in the thymus. Thus, sγc represents a new mechanism to control cytokine availability during T cell development that constrains mature T cell production and specifically iNKT cell generation in the thymus.
Collapse
|
13
|
Kimura MY, Thomas J, Tai X, Guinter TI, Shinzawa M, Etzensperger R, Li Z, Love P, Nakayama T, Singer A. Timing and duration of MHC I positive selection signals are adjusted in the thymus to prevent lineage errors. Nat Immunol 2016; 17:1415-1423. [PMID: 27668801 DOI: 10.1038/ni.3560] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 08/17/2016] [Indexed: 12/12/2022]
Abstract
Major histocompatibility complex class I (MHC I) positive selection of CD8+ T cells in the thymus requires that T cell antigen receptor (TCR) signaling end in time for cytokines to induce Runx3d, the CD8-lineage transcription factor. We examined the time required for these events and found that the overall duration of positive selection was similar for all CD8+ thymocytes in mice, despite markedly different TCR signaling times. Notably, prolonged TCR signaling times were counter-balanced by accelerated Runx3d induction by cytokines and accelerated differentiation into CD8+ T cells. Consequently, lineage errors did not occur except when MHC I-TCR signaling was so prolonged that the CD4-lineage-specifying transcription factor ThPOK was expressed, preventing Runx3d induction. Thus, our results identify a compensatory signaling mechanism that prevents lineage-fate errors by dynamically modulating Runx3d induction rates during MHC I positive selection.
Collapse
Affiliation(s)
- Motoko Y Kimura
- Experimental Immunology Branch, National Cancer Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Julien Thomas
- Experimental Immunology Branch, National Cancer Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Xuguang Tai
- Experimental Immunology Branch, National Cancer Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Terry I Guinter
- Experimental Immunology Branch, National Cancer Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Miho Shinzawa
- Experimental Immunology Branch, National Cancer Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Ruth Etzensperger
- Experimental Immunology Branch, National Cancer Institute, US National Institutes of Health, Bethesda, Maryland, USA
| | - Zhenhu Li
- Laboratory of Mammalian Genes and Development, Eunice Kennedy Schriver National Institute of Child Health and Human Development, US National Institutes of Health, Bethesda, Maryland, USA
| | - Paul Love
- Laboratory of Mammalian Genes and Development, Eunice Kennedy Schriver National Institute of Child Health and Human Development, US National Institutes of Health, Bethesda, Maryland, USA
| | | | - Alfred Singer
- Experimental Immunology Branch, National Cancer Institute, US National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
14
|
Lins MP, de Araújo Vieira LF, Rosa AAM, Smaniotto S. Growth hormone in the presence of laminin modulates interaction of human thymic epithelial cells and thymocytes in vitro. Biol Res 2016; 49:37. [PMID: 27590178 PMCID: PMC5010746 DOI: 10.1186/s40659-016-0097-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Accepted: 08/12/2016] [Indexed: 12/05/2022] Open
Abstract
Background Several evidences indicate that hormones and neuropeptides function as immunomodulators. Among these, growth hormone (GH) is known to act on the thymic microenvironment, supporting its role in thymocyte differentiation. The aim of this study was to evaluate the effect of GH on human thymocytes and thymic epithelial cells (TEC) in the presence of laminin. Results GH increased thymocyte adhesion on BSA-coated and further on laminin-coated surfaces. The number of migrating cells in laminin-coated membrane was higher in GH-treated thymocyte group. In both results, VLA-6 expression on thymocytes was constant. Also, treatment with GH enhanced laminin production by TEC after 24 h in culture. However, VLA-6 integrin expression on TEC remained unchanged. Finally, TEC/thymocyte co-culture model demonstrated that GH elevated absolute number of double-negative (CD4−CD8−) and single-positive CD4+ and CD8+ thymocytes. A decrease in cell number was noted in double-positive (CD4+CD8+) thymocytes. Conclusions The results of this study demonstrate that GH is capable of enhancing the migratory capacity of human thymocytes in the presence of laminin and promotes modulation of thymocyte subsets after co-culture with TEC.
Collapse
Affiliation(s)
- Marvin Paulo Lins
- Laboratory of Cell Biology, Institute of Biological Sciences and Health, Federal University of Alagoas, Maceió, Alagoas, Brazil
| | | | - Alfredo Aurélio Marinho Rosa
- Laboratory of Cell Biology, Institute of Biological Sciences and Health, Federal University of Alagoas, Maceió, Alagoas, Brazil
| | - Salete Smaniotto
- Laboratory of Cell Biology, Institute of Biological Sciences and Health, Federal University of Alagoas, Maceió, Alagoas, Brazil.
| |
Collapse
|
15
|
Tuulasvaara A, Vanhanen R, Baldauf HM, Puntila J, Arstila TP. Interleukin-7 promotes human regulatory T cell development at the CD4+CD8+ double-positive thymocyte stage. J Leukoc Biol 2016; 100:491-8. [PMID: 26965634 DOI: 10.1189/jlb.1a0415-164r] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Accepted: 02/20/2016] [Indexed: 12/19/2022] Open
Abstract
Although mature human FOXP3(+) regulatory T cells are CD127 (IL-7Rα) negative, CD4(+)CD8(+) FOXP3(+) thymocytes express relatively high levels of CD127 and are responsive to IL-7. However, the role of IL-7 in human regulatory T cell development is poorly known. We show that at the CD4(+)CD8(+) stage, FOXP3(+) thymocytes are highly susceptible to apoptosis, and IL-7 selectively rescues them from death, leading to an increased frequency of FOXP3(+) cells. IL-7 also promotes the development of regulatory T cell phenotype by inducing up-regulation of FOXP3(+) and CTLA-4 expression. In contrast, IL-7 does not enhance proliferation of FOXP3(+)thymocytes or induce demethylation of FOXP3(+) regulatory T cell-specific demethylated region. After the CD4(+)CD8(+) stage, the FOXP3(+) thymocytes down-regulate CD127 expression but despite very low levels of CD127, remain responsive to IL-7. These results suggest that IL-7 affects human regulatory T cell development in the thymus by at least 2 distinct mechanisms: suppression of apoptosis and up-regulation of FOXP3(+) expression.
Collapse
Affiliation(s)
- Anni Tuulasvaara
- Haartman Institute, Department of Bacteriology and Immunology, and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland; and
| | - Reetta Vanhanen
- Haartman Institute, Department of Bacteriology and Immunology, and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland; and
| | - Hanna-Mari Baldauf
- Haartman Institute, Department of Bacteriology and Immunology, and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland; and
| | - Juha Puntila
- Department of Surgery, Hospital for Children and Adolescents, Helsinki University Hospital, Helsinki, Finland
| | - T Petteri Arstila
- Haartman Institute, Department of Bacteriology and Immunology, and Research Programs Unit, Immunobiology, University of Helsinki, Helsinki, Finland; and
| |
Collapse
|
16
|
Abe A, Tani-ichi S, Shitara S, Cui G, Yamada H, Miyachi H, Kitano S, Hara T, Abe R, Yoshikai Y, Ikuta K. An Enhancer of the IL-7 Receptor α-Chain Locus Controls IL-7 Receptor Expression and Maintenance of Peripheral T Cells. THE JOURNAL OF IMMUNOLOGY 2015; 195:3129-38. [PMID: 26336149 DOI: 10.4049/jimmunol.1302447] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Accepted: 07/28/2015] [Indexed: 12/24/2022]
Abstract
The IL-7R plays critical roles in lymphocyte development and homeostasis. Although IL-7R expression is strictly regulated during lymphocyte differentiation and the immune response, little is known regarding its in vivo regulation. To address this issue, we established a mouse line with targeted deletion of the conserved non-coding sequence 1 (CNS1) element found 3.6 kb upstream of the IL-7Rα promoter. We report that IL-7Rα is expressed normally on T and B cells in thymus and bone marrow of CNS1(-/-) mice except for in regulatory T cells. In contrast, these mice show reduced IL-7Rα expression in conventional CD4 and CD8 T cells as well as regulatory T, NKT, and γδ T cells in the periphery. CD4 T cells of CNS1(-/-) mice showed IL-7Rα upregulation in the absence of growth factors and IL-7Rα downregulation by IL-7 or TCR stimulation, although the expression levels were lower than those in control mice. Naive CD4 and CD8 T cells of CNS1(-/-) mice show attenuated survival by culture with IL-7 and reduced homeostatic proliferation after transfer into lymphopenic hosts. CNS1(-/-) mice exhibit impaired maintenance of Ag-stimulated T cells. Furthermore, IL-7Rα upregulation by glucocorticoids and TNF-α was abrogated in CNS1(-/-) mice. This work demonstrates that the CNS1 element controls IL-7Rα expression and maintenance of peripheral T cells, suggesting differential regulation of IL-7Rα expression between central and peripheral lymphoid organs.
Collapse
Affiliation(s)
- Akifumi Abe
- Laboratory of Biological Protection, Department of Biological Responses, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Shizue Tani-ichi
- Laboratory of Biological Protection, Department of Biological Responses, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Soichiro Shitara
- Laboratory of Biological Protection, Department of Biological Responses, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan; Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan
| | - Guangwei Cui
- Laboratory of Biological Protection, Department of Biological Responses, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan; Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Hisataka Yamada
- Division of Host Defense, Network Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Hitoshi Miyachi
- Reproductive Engineering Team, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan; and
| | - Satsuki Kitano
- Reproductive Engineering Team, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan; and
| | - Takahiro Hara
- Laboratory of Biological Protection, Department of Biological Responses, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan
| | - Ryo Abe
- Division of Immunobiology, Research Institute for Biomedical Sciences, Tokyo University of Science, Chiba 278-0022, Japan
| | - Yasunobu Yoshikai
- Division of Host Defense, Network Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Koichi Ikuta
- Laboratory of Biological Protection, Department of Biological Responses, Institute for Virus Research, Kyoto University, Kyoto 606-8507, Japan;
| |
Collapse
|
17
|
Inghirami G, Chan WC, Pileri S. Peripheral T-cell and NK cell lymphoproliferative disorders: cell of origin, clinical and pathological implications. Immunol Rev 2015; 263:124-59. [PMID: 25510275 DOI: 10.1111/imr.12248] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
T-cell lymphoproliferative disorders are a heterogeneous group of neoplasms with distinct clinical-biological properties. The normal cellular counterpart of these processes has been postulated based on functional and immunophenotypic analyses. However, T lymphocytes have been proven to be remarkably capable of modulating their properties, adapting their function in relationship with multiple stimuli and to the microenvironment. This impressive plasticity is determined by the equilibrium among a pool of transcription factors and by DNA chromatin regulators. It is now proven that the acquisition of specific genomic defects leads to the enforcement/activation of distinct pathways, which ultimately alter the preferential activation of defined regulators, forcing the neoplastic cells to acquire features and phenotypes distant from their original fate. Thus, dissecting the landscape of the genetic defects and their functional consequences in T-cell neoplasms is critical not only to pinpoint the origin of these tumors but also to define innovative mechanisms to re-adjust an unbalanced state to which the tumor cells have become addicted and make them vulnerable to therapies and targetable by the immune system. In our review, we briefly describe the pathological and clinical aspects of the T-cell lymphoma subtypes as well as NK-cell lymphomas and then focus on the current understanding of their pathogenesis and the implications on diagnosis and treatment.
Collapse
Affiliation(s)
- Giorgio Inghirami
- Department of Molecular Biotechnology and Health Science and Center for Experimental Research and Medical Studies (CeRMS), University of Torino, Torino, Italy; Department of Pathology, and NYU Cancer Center, New York University School of Medicine, New York, NY, USA; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY, USA
| | | | | | | |
Collapse
|
18
|
Maude SL, Dolai S, Delgado-Martin C, Vincent T, Robbins A, Selvanathan A, Ryan T, Hall J, Wood AC, Tasian SK, Hunger SP, Loh ML, Mullighan CG, Wood BL, Hermiston ML, Grupp SA, Lock RB, Teachey DT. Efficacy of JAK/STAT pathway inhibition in murine xenograft models of early T-cell precursor (ETP) acute lymphoblastic leukemia. Blood 2015; 125:1759-67. [PMID: 25645356 PMCID: PMC4357583 DOI: 10.1182/blood-2014-06-580480] [Citation(s) in RCA: 172] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 01/08/2015] [Indexed: 12/30/2022] Open
Abstract
Early T-cell precursor (ETP) acute lymphoblastic leukemia (ALL) is a recently described subtype of T-ALL characterized by a unique immunophenotype and genomic profile, as well as a high rate of induction failure. Frequent mutations in cytokine receptor and Janus kinase (JAK)/signal transducer and activator of transcription (STAT) signaling pathways led us to hypothesize that ETP-ALL is dependent on JAK/STAT signaling. Here we demonstrate aberrant activation of the JAK/STAT pathway in ETP-ALL blasts relative to non-ETP T-ALL. Moreover, ETP-ALL showed hyperactivation of STAT5 in response to interleukin-7, an effect that was abrogated by the JAK1/2 inhibitor ruxolitinib. In vivo, ruxolitinib displayed activity in 6 of 6 patient-derived murine xenograft models of ETP-ALL, with profound single-agent efficacy in 5 models. Ruxolitinib treatment decreased peripheral blast counts relative to pretreatment levels and compared with control (P < .01) in 5 of 6 ETP-ALL xenografts, with marked reduction in mean splenic blast counts (P < .01) in 6 of 6 samples. Surprisingly, both JAK/STAT pathway activation and ruxolitinib efficacy were independent of the presence of JAK/STAT pathway mutations, raising the possibility that the therapeutic potential of ruxolitinib in ETP-ALL extends beyond those cases with JAK mutations. These findings establish the preclinical in vivo efficacy of ruxolitinib in ETP-ALL, a biologically distinct subtype for which novel therapies are needed.
Collapse
Affiliation(s)
- Shannon L Maude
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sibasish Dolai
- Leukaemia Biology, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Cristina Delgado-Martin
- Division of Hematology/Oncology, University of California, San Francisco Benioff Children's Hospital, San Francisco, CA
| | - Tiffaney Vincent
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Alissa Robbins
- Leukaemia Biology, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Arthavan Selvanathan
- Leukaemia Biology, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - Theresa Ryan
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Junior Hall
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA
| | - Andrew C Wood
- Molecular Medicine and Pathology, University of Auckland, Auckland, New Zealand
| | - Sarah K Tasian
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Stephen P Hunger
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Mignon L Loh
- Division of Hematology/Oncology, University of California, San Francisco Benioff Children's Hospital, San Francisco, CA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, TN; and
| | - Brent L Wood
- Division of Hematopathology, University of Washington and Seattle Cancer Care Alliance, Seattle, WA
| | - Michelle L Hermiston
- Division of Hematology/Oncology, University of California, San Francisco Benioff Children's Hospital, San Francisco, CA
| | - Stephan A Grupp
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Richard B Lock
- Leukaemia Biology, Children's Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, Sydney, Australia
| | - David T Teachey
- Division of Oncology, Children's Hospital of Philadelphia, Philadelphia, PA; Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| |
Collapse
|
19
|
Boudil A, Matei IR, Shih HY, Bogdanoski G, Yuan JS, Chang SG, Montpellier B, Kowalski PE, Voisin V, Bashir S, Bader GD, Krangel MS, Guidos CJ. IL-7 coordinates proliferation, differentiation and Tcra recombination during thymocyte β-selection. Nat Immunol 2015; 16:397-405. [PMID: 25729925 PMCID: PMC4368453 DOI: 10.1038/ni.3122] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/10/2015] [Indexed: 12/15/2022]
Abstract
Signaling via the pre-T cell antigen receptor (pre-TCR) and the receptor Notch1 induces transient self-renewal (β-selection) of TCRβ(+) CD4(-)CD8(-) double-negative stage 3 (DN3) and DN4 progenitor cells that differentiate into CD4(+)CD8(+) double-positive (DP) thymocytes, which then rearrange the locus encoding the TCR α-chain (Tcra). Interleukin 7 (IL-7) promotes the survival of TCRβ(-) DN thymocytes by inducing expression of the pro-survival molecule Bcl-2, but the functions of IL-7 during β-selection have remained unclear. Here we found that IL-7 signaled TCRβ(+) DN3 and DN4 thymocytes to upregulate genes encoding molecules involved in cell growth and repressed the gene encoding the transcriptional repressor Bcl-6. Accordingly, IL-7-deficient DN4 cells lacked trophic receptors and did not proliferate but rearranged Tcra prematurely and differentiated rapidly. Deletion of Bcl6 partially restored the self-renewal of DN4 cells in the absence of IL-7, but overexpression of BCL2 did not. Thus, IL-7 critically acts cooperatively with signaling via the pre-TCR and Notch1 to coordinate proliferation, differentiation and Tcra recombination during β-selection.
Collapse
Affiliation(s)
- Amine Boudil
- 1] Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Canada. [2] Department of Immunology, University of Toronto, Toronto, Canada
| | - Irina R Matei
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Canada
| | - Han-Yu Shih
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Goce Bogdanoski
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Canada
| | - Julie S Yuan
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Canada
| | - Stephen G Chang
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Canada
| | - Bertrand Montpellier
- 1] Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Canada. [2] Department of Immunology, University of Toronto, Toronto, Canada
| | - Paul E Kowalski
- Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Canada
| | | | | | - Gary D Bader
- 1] The Donnelly Centre, University of Toronto, Toronto, Canada. [2] Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Michael S Krangel
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, USA
| | - Cynthia J Guidos
- 1] Program in Developmental and Stem Cell Biology, Hospital for Sick Children Research Institute, Toronto, Canada. [2] Department of Immunology, University of Toronto, Toronto, Canada
| |
Collapse
|
20
|
Phee H, Au-Yeung BB, Pryshchep O, O'Hagan KL, Fairbairn SG, Radu M, Kosoff R, Mollenauer M, Cheng D, Chernoff J, Weiss A. Pak2 is required for actin cytoskeleton remodeling, TCR signaling, and normal thymocyte development and maturation. eLife 2014; 3:e02270. [PMID: 24843022 PMCID: PMC4017645 DOI: 10.7554/elife.02270] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The molecular mechanisms that govern thymocyte development and maturation are incompletely understood. The P21-activated kinase 2 (Pak2) is an effector for the Rho family GTPases Rac and Cdc42 that regulate actin cytoskeletal remodeling, but its role in the immune system remains poorly understood. In this study, we show that T-cell specific deletion of Pak2 gene in mice resulted in severe T cell lymphopenia accompanied by marked defects in development, maturation, and egress of thymocytes. Pak2 was required for pre-TCR β-selection and positive selection. Surprisingly, Pak2 deficiency in CD4 single positive thymocytes prevented functional maturation and reduced expression of S1P1 and KLF2. Mechanistically, Pak2 is required for actin cytoskeletal remodeling triggered by TCR. Failure to induce proper actin cytoskeletal remodeling impaired PLCγ1 and Erk1/2 signaling in the absence of Pak2, uncovering the critical function of Pak2 as an essential regulator that governs the actin cytoskeleton-dependent signaling to ensure normal thymocyte development and maturation. DOI:http://dx.doi.org/10.7554/eLife.02270.001 T cells are a key element of the immune system. There are many different types of T cells, and they all have their origins in hematopoietic stem cells that are found in the bone marrow. These stem cells leave the bone marrow and circulate in the body until they reach an organ called the thymus, where they become early thymic progenitor cells. These progenitor cells then undergo a process called differentiation to become specific types of T cells, which mature in the thymus before moving to the blood. Although various molecules and mechanisms are known to be involved in the development of T cells, many details of this process are not understood. One group of molecules that has been implicated in the differentiation of T cells is the p21-activated kinases. Kinases are proteins that activate or deactivate other proteins by adding phosphate groups to specific amino acids. Pak2 adds phosphorylate groups to various proteins that are involved in the reorganization of an important structure inside the cell called the cytoskeleton. A kinase called Pak2 has an important role in the reorganization of the cytoskeleton, and since this reorganization is involved in almost all aspects of T cell biology, it seems plausible that Pak2 is also involved in the development of T cells. However, it has not been possible to test this idea because deleting the gene for Pak2 in mice results in their death. Now, Phee et al. have overcome this problem by performing experiments in which the gene for Pak2 was only deleted in T cells. These mice had significantly fewer mature T cells than healthy mice. In particular, the absence of Pak2 in thymocytes (the cells that become T cells) prevented them from maturing into T cells, and also prevented them from producing a receptor protein that is needed for mature T cells to leave the thymus. This work implies that disruption of the Pak2-mediated signaling pathway that regulates the cytoskeleton may weaken the immune system in humans. DOI:http://dx.doi.org/10.7554/eLife.02270.002
Collapse
Affiliation(s)
- Hyewon Phee
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Byron B Au-Yeung
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, United States
| | - Olga Pryshchep
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Kyle Leonard O'Hagan
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Stephanie Grace Fairbairn
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, United States
| | - Maria Radu
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, United States
| | - Rachelle Kosoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, United States
| | - Marianne Mollenauer
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, United States
| | - Debra Cheng
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, United States
| | - Jonathan Chernoff
- Cancer Biology Program, Fox Chase Cancer Center, Philadelphia, United States
| | - Arthur Weiss
- Department of Medicine, Division of Rheumatology, University of California, San Francisco, San Francisco, United States Rosalind Russell Medical Research Center for Arthritis, Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, United States
| |
Collapse
|
21
|
Dooms H. Interleukin-7: Fuel for the autoimmune attack. J Autoimmun 2013; 45:40-8. [PMID: 23831438 DOI: 10.1016/j.jaut.2013.06.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 06/12/2013] [Indexed: 01/19/2023]
Abstract
Interleukin-7 (IL-7) is a critical survival factor for lymphocytes and recent studies suggest targeting the IL-7/IL-7Rα pathway holds promise for the treatment of autoimmune diseases. Several lines of evidence, genetic as well as functional, indicate an important role for this cytokine in autoimmune inflammation: polymorphisms in the IL-7Rα have been associated with increased risk for autoimmune disease and blocking IL-7/IL-7Rα with antibodies showed therapeutic efficacy in several autoimmune mouse models. Insights are starting to emerge about the mechanisms underlying IL-7's role in autoimmunity and tolerance, revealing surprising novel functions beyond its traditional activity as a T cell survival factor. In the first part of this review, the functions of IL-7 in the immune system are concisely described, providing a basis for understanding their potential role in promoting autoimmune responses. In the second part, current knowledge about the role of IL-7 in various autoimmune conditions is reviewed.
Collapse
Affiliation(s)
- Hans Dooms
- Department of Medicine, Arthritis Center/Rheumatology Section, Boston University School of Medicine, 72 East Concord Street, E519, Boston, MA 02118, USA.
| |
Collapse
|
22
|
Xiong J, Parker BL, Dalheimer SL, Yankee TM. Interleukin-7 supports survival of T-cell receptor-β-expressing CD4(-) CD8(-) double-negative thymocytes. Immunology 2013; 138:382-91. [PMID: 23215679 DOI: 10.1111/imm.12050] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 11/30/2012] [Accepted: 12/05/2012] [Indexed: 01/17/2023] Open
Abstract
Among the milestones that occur during T-cell development in the thymus is the expression of T-cell receptor-β (TCR-β) and the formation of the pre-TCR complex. Signals emanating from the pre-TCR trigger survival, proliferation and differentiation of T-cell precursors. Although the pre-TCR is essential for these cell outcomes, other receptors, such as Notch and CXCR4, also contribute. Whether interleukin-7 (IL-7) participates in promoting the survival or proliferation of pre-TCR-expressing cells is controversial. We used in vitro and in vivo models of T-cell development to examine the function of IL-7 in TCR-β-expressing thymocytes. Culturing TCR-β-expressing CD4(-) CD8(-) double-negative thymocytes in an in vitro model of T-cell development revealed that IL-7 reduced the frequency of CD4(+) CD8(+) double-positive thymocytes at the time of harvest. The mechanism for this change in the percentage of double-positive cells was that IL-7 promoted the survival of thymocytes that had not yet differentiated. By preserving the double-negative population, IL-7 reduced the frequency of double-positive thymocytes. Interleukin-7 was not required for proliferation in the in vitro system. To follow this observation, we examined mice lacking CD127 (IL-7Rα). In addition to the known effect of CD127 deficiency on T-cell development before TCR-β expression, CD127 deficiency also impaired the development of TCR-β-expressing double-negative thymocytes. Specifically, we found that Bcl-2 expression and cell cycle progression were reduced in TCR-β-expressing double-negative thymocytes in mice lacking CD127. We conclude that IL-7 continues to function after TCR-β is expressed by promoting the survival of TCR-β-expressing double-negative thymocytes.
Collapse
Affiliation(s)
- Juan Xiong
- Department of Microbiology, Molecular Genetics, and Immunology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | | | | | | |
Collapse
|
23
|
|
24
|
Interleukin-7 receptor controls development and maturation of late stages of thymocyte subpopulations. Proc Natl Acad Sci U S A 2012; 110:612-7. [PMID: 23267098 DOI: 10.1073/pnas.1219242110] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Interleukin (IL)-7 is a cytokine essential for T lymphocyte development and homeostasis. However, little is known about the roles of IL-7 receptor α-chain (IL-7Rα) in late stages of T-cell development. To address this question, we established IL-7Rα-floxed mice and crossed them with CD4-Cre transgenic mice. Resultant IL-7R conditional knockout (IL-7RcKO) mice exhibited marked reduction in CD8 single positive (SP) T cells, regulatory T cells (Tregs), and natural killer T (NKT) cells in thymus. The proportion and proliferation of both mature CD4SP and CD8SP thymocytes were decreased without affecting Runx expression. In addition, expression of the glucocorticoid-induced TNF receptor was reduced in CD4SP and CD8SP thymocytes, and expression of CD5 was decreased in CD8SP thymocytes. IL-7RcKO mice also showed impaired Treg and NKT cell proliferation and inhibition of NKT cell maturation. Bcl-2 expression was reduced in CD4SP and CD8SP thymocytes but not in Tregs and NKT cells, and introduction of a Bcl-2 transgene rescued frequency and CD5 expression of CD8SP thymocytes. Furthermore, IL-7RcKO mice exhibited greatly increased numbers of B cells and, to a lesser extent, γδ T and dendritic cells in thymus. Overall, this study demonstrates that IL-7Rα differentially controls development and maturation of thymocyte subpopulations in late developmental stages and suggests that IL-7R expression on αβ T cells suppresses development of other cell lineages in thymus.
Collapse
|
25
|
Hu Q, Nicol SA, Suen AYW, Baldwin TA. Examination of thymic positive and negative selection by flow cytometry. J Vis Exp 2012:4269. [PMID: 23093039 DOI: 10.3791/4269] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A healthy immune system requires that T cells respond to foreign antigens while remaining tolerant to self-antigens. Random rearrangement of the T cell receptor (TCR) α and β loci generates a T cell repertoire with vast diversity in antigen specificity, both to self and foreign. Selection of the repertoire during development in the thymus is critical for generating safe and useful T cells. Defects in thymic selection contribute to the development of autoimmune and immunodeficiency disorders(1-4). T cell progenitors enter the thymus as double negative (DN) thymocytes that do not express CD4 or CD8 co-receptors. Expression of the αβTCR and both co-receptors occurs at the double positive (DP) stage. Interaction of the αβTCR with self-peptide-MHC (pMHC) presented by thymic cells determines the fate of the DP thymocyte. High affinity interactions lead to negative selection and elimination of self-reactive thymocytes. Low affinity interactions result in positive selection and development of CD4 or CD8 single positive (SP) T cells capable of recognizing foreign antigens presented by self-MHC(5). Positive selection can be studied in mice with a polyclonal (wildtype) TCR repertoire by observing the generation of mature T cells. However, they are not ideal for the study of negative selection, which involves deletion of small antigen-specific populations. Many model systems have been used to study negative selection but vary in their ability to recapitulate physiological events(6). For example, in vitro stimulation of thymocytes lacks the thymic environment that is intimately involved in selection, while administration of exogenous antigen can lead to non-specific deletion of thymocytes(7-9). Currently, the best tools for studying in vivo negative selection are mice that express a transgenic TCR specific for endogenous self-antigen. However, many classical TCR transgenic models are characterized by premature expression of the transgenic TCRα chain at the DN stage, resulting in premature negative selection. Our lab has developed the HY(cd4) model, in which the transgenic HY TCRα is conditionally expressed at the DP stage, allowing negative selection to occur during the DP to SP transition as occurs in wildtype mice(10). Here, we describe a flow cytometry-based protocol to examine thymic positive and negative selection in the HY(cd4) mouse model. While negative selection in HY(cd4) mice is highly physiological, these methods can also be applied to other TCR transgenic models. We will also present general strategies for analyzing positive selection in a polyclonal repertoire applicable to any genetically manipulated mice.
Collapse
Affiliation(s)
- Qian Hu
- Department of Medical Microbiology and Immunology, University of Alberta, AB, Canada
| | | | | | | |
Collapse
|
26
|
Hong C, Luckey M, Park JH. Intrathymic IL-7: the where, when, and why of IL-7 signaling during T cell development. Semin Immunol 2012; 24:151-8. [PMID: 22421571 PMCID: PMC3358706 DOI: 10.1016/j.smim.2012.02.002] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2011] [Accepted: 02/15/2012] [Indexed: 12/26/2022]
Abstract
The thymus is the birthplace of all T lineage cells. But the thymus is also a cradle as it provides the environment for further maturation and differentiation of immature thymocytes. While many factors contribute to make the thymus a unique place for T cell development, here we review the essential role of intrathymic interleukin-7 (IL-7). In the absence of IL-7 signaling, survival, proliferation and differentiation of immature thymocytes are all severely impaired. Consequently, IL-7 is critical to nurture and guide T precursor cells through the diverse steps of thymic maturation. Interestingly, even as IL-7 signaling is such a critical factor, IL-7 signaling must be also actively suppressed during specific stages of T cell differentiation. These contradictory observations are puzzling but can be satisfactorily explained when understanding the developmental context of IL-7 signaling. In this regard, here we will discuss the spatiotemporal expression of intrathymic IL-7 and address the stage-specific effects of IL-7 signaling in developing thymocytes. Specifically, we will review other facets of intrathymic IL-7 beyond its role as a pro-survival factor and so clarify and reaffirm the unique role of IL-7 as a prime factor in T cell development and differentiation.
Collapse
Affiliation(s)
- Changwan Hong
- Exp. Immunol. Branch, National Cancer Inst., NIH, Bethesda, MD 20892-1360
| | - Megan Luckey
- Exp. Immunol. Branch, National Cancer Inst., NIH, Bethesda, MD 20892-1360
| | - Jung-Hyun Park
- Exp. Immunol. Branch, National Cancer Inst., NIH, Bethesda, MD 20892-1360
| |
Collapse
|
27
|
HES1 opposes a PTEN-dependent check on survival, differentiation, and proliferation of TCRβ-selected mouse thymocytes. Blood 2012; 120:1439-48. [PMID: 22649105 DOI: 10.1182/blood-2011-12-395319] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The developmental progression of immature thymocytes requires cooperative input from several pathways, with Notch signals playing an indispensable role at the T-cell receptor (TCR)-β selection checkpoint. Notch signals affect the activation of the PI3K/Akt pathway, which is required for pTα/TCRβ (pre-TCR)-induced survival, differentiation, and proliferation of developing αβ-lineage thymocytes. However, the molecular players responsible for the interaction between the Notch and PI3K pathways at this critical developmental stage are unknown. Here, we show that Notch induction of Hes1 is necessary to repress the PI3K/Akt pathway inhibitor, PTEN (phosphatase and tensin homolog), which in turn facilitates pre-TCR-induced differentiation. In support of this mechanism, deletion or down-regulation of Pten overcomes the Notch signaling requirement for survival and differentiation during β-selection. In addition, c-Myc is a critical target of Notch at this stage, as c-Myc expression overcomes the Notch signaling requirement for proliferation during β-selection. Collectively, our results point to HES1, via repression of PTEN, and c-Myc as critical mediators of Notch function at the β-selection checkpoint.
Collapse
|
28
|
González-García S, García-Peydró M, Alcain J, Toribio ML. Notch1 and IL-7 receptor signalling in early T-cell development and leukaemia. Curr Top Microbiol Immunol 2012; 360:47-73. [PMID: 22695916 DOI: 10.1007/82_2012_231] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Notch receptors are master regulators of many aspects of development and tissue renewal in metazoans. Notch1 activation is essential for T-cell specification of bone marrow-derived multipotent progenitors that seed the thymus, and for proliferation and further progression of early thymocytes along the T-cell lineage. Deregulated activation of Notch1 significantly contributes to the generation of T-cell acute lymphoblastic leukaemia (T-ALL). In addition to Notch1 signals, survival and proliferation signals provided by the IL-7 receptor (IL-7R) are also required during thymopoiesis. Our understanding of the molecular mechanisms controlling stage-specific survival and proliferation signals provided by Notch1 and IL-7R has recently been improved by the discovery that the IL-7R is a transcriptional target of Notch1. Thus, Notch1 controls T-cell development, in part by regulating the stage- and lineage-specific expression of IL-7R. The finding that induction of IL-7R expression downstream of Notch1 also occurs in T-ALL highlights the important contribution that deregulated IL-7R expression and function may have in this pathology. Confirming this notion, oncogenic IL7R gain-of-function mutations have recently been identified in childhood T-ALL. Here we discuss the fundamental role of Notch1 and IL-7R signalling pathways in physiological and pathological T-cell development in mice and men, highlighting their close molecular underpinnings.
Collapse
Affiliation(s)
- Sara González-García
- Centro de Biología Molecular, Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | | | | | | |
Collapse
|
29
|
Grigoriadis G, Vasanthakumar A, Banerjee A, Grumont R, Overall S, Gleeson P, Shannon F, Gerondakis S. c-Rel controls multiple discrete steps in the thymic development of Foxp3+ CD4 regulatory T cells. PLoS One 2011; 6:e26851. [PMID: 22066012 PMCID: PMC3204987 DOI: 10.1371/journal.pone.0026851] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Accepted: 10/05/2011] [Indexed: 12/21/2022] Open
Abstract
The development of natural Foxp3+ CD4 regulatory T cells (nTregs) proceeds via two steps that involve the initial antigen dependent generation of CD25+GITRhiFoxp3−CD4+ nTreg precursors followed by the cytokine induction of Foxp3. Using mutant mouse models that lack c-Rel, the critical NF-κB transcription factor required for nTreg differentiation, we establish that c-Rel regulates both of these developmental steps. c-Rel controls the generation of nTreg precursors via a haplo-insufficient mechanism, indicating that this step is highly sensitive to c-Rel levels. However, maintenance of c-Rel in an inactive state in nTreg precursors demonstrates that it is not required for a constitutive function in these cells. While the subsequent IL-2 induction of Foxp3 in nTreg precursors requires c-Rel, this developmental transition does not coincide with the nuclear expression of c-Rel. Collectively, our results support a model of nTreg differentiation in which c-Rel generates a permissive state for foxp3 transcription during the development of nTreg precursors that influences the subsequent IL-2 dependent induction of Foxp3 without a need for c-Rel reactivation.
Collapse
Affiliation(s)
- George Grigoriadis
- Centre for Immunology, Burnet Institute, Melbourne, Australia
- Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Australia
| | | | - Ashish Banerjee
- Centre for Immunology, Burnet Institute, Melbourne, Australia
| | - Raelene Grumont
- Centre for Immunology, Burnet Institute, Melbourne, Australia
| | - Sarah Overall
- Bio21, University of Melbourne, Parkville, Australia
| | - Paul Gleeson
- Bio21, University of Melbourne, Parkville, Australia
| | - Frances Shannon
- The John Curtin School of Medical Research, Australian National University, Canberra City, Australia
| | - Steve Gerondakis
- Centre for Immunology, Burnet Institute, Melbourne, Australia
- Australian Centre for Blood Diseases and Department of Clinical Hematology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Australia
- Department of Immunology, Monash University, Alfred Medical Research and Education Precinct, Melbourne, Australia
- * E-mail:
| |
Collapse
|
30
|
A Human Recombinant IL-7/HGFα Hybrid Cytokine Enhances T-Cell Reconstitution in Mice After Syngeneic Bone Marrow Transplantation. Transplantation 2011; 92:516-22. [DOI: 10.1097/tp.0b013e318228d766] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
31
|
Tani-Ichi S, Satake M, Ikuta K. The pre-TCR signal induces transcriptional silencing of the TCRγ locus by reducing the recruitment of STAT5 and Runx to transcriptional enhancers. Int Immunol 2011; 23:553-63. [PMID: 21750145 DOI: 10.1093/intimm/dxr055] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The mouse TCRγ locus is positively regulated by the transcription factors STAT5 and Runx. While the locus undergoes frequent rearrangements in T lymphocytes, TCRγ transcription is repressed in αβ T cells. This phenomenon, known as TCRγ silencing, depends on pre-TCR-induced thymocyte proliferation. The molecular basis for TCRγ silencing, however, is largely unknown. Here, we show that pre-TCR signaling reduces transcription and histone acetylation of the TCRγ locus irrespective of V-J rearrangements. We also demonstrate that Runx is recruited to Eγ and HsA enhancer elements of the TCRγ locus, primarily at the CD4(-)CD8(-) double-negative stage and that Runx binding to these elements decreases at later stages of thymocyte development. Importantly, anti-CD3 antibody treatment decreased IL-7R expression levels, STAT5 phosphorylation and recruitment of STAT5 and Runx to Eγ and HsA elements in RAG2-deficient thymocytes, suggesting that pre-TCR signaling triggers reduced binding of STAT5 and Runx to the enhancer elements. Furthermore, we observed that misexpression of STAT5 or Runx in the CD4(+)CD8(+) double-positive cell line DPK induces TCRγ gene transcription. Finally, we showed that TCRγ transcription is induced in αβ T cells from Runx3 transgenic mice, suggesting that Runx3 counteracts TCRγ silencing in αβ T cells in vivo. Our results suggest that pre-TCR signaling indirectly inactivates TCRγ enhancers by reducing recruitment of STAT5 and Runx and imply that this effect is an important step for TCRγ silencing in αβ T cells.
Collapse
Affiliation(s)
- Shizue Tani-Ichi
- Laboratory of Biological Protection, Department of Biological Responses, Institute for Virus Research, Kyoto University, Sakyo-ku, Japan
| | | | | |
Collapse
|
32
|
Ten-color flow cytometry reveals distinct patterns of expression of CD124 and CD126 by developing thymocytes. BMC Immunol 2011; 12:36. [PMID: 21689450 PMCID: PMC3130696 DOI: 10.1186/1471-2172-12-36] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2010] [Accepted: 06/20/2011] [Indexed: 11/10/2022] Open
Abstract
Background We have developed a 12-parameter/10-color flow cytometric staining method for the simultaneous detection and characterization of 21 mouse thymocyte subpopulations that represent discreet stages of T cell development. To demonstrate the utility of this method, we assessed cytokine receptor expression on mouse thymocyte subsets. These experiments revealed distinct patterns of surface expression of receptors for the cytokines IL-4 and IL-6. Results The IL-4 receptor α chain (CD124) was highly expressed on the earliest thymocyte subsets, then downregulated prior to T cell receptor β-selection and finally upregulated in the CD4/CD8 double positive cells prior to positive selection. The IL-6 receptor α chain (CD126) showed a different pattern of expression. It was expressed on the most mature subsets within the CD4 and CD8 single positive (SP) compartments and was absent on all other thymocytes with the exception of a very small cKit-CD4-CD8- population. Intracellular staining of SP thymocytes for phosphorylated STAT-1 demonstrated that IL-6 signaling was confined to the most mature SP subsets. Conclusions This 12-parameter staining methodology uses only commercially available fluorochrome-coupled monoclonal antibodies and therefore could be employed by any investigator with access to a 4-laser flow cytometer. This novel staining scheme allowed us to easily phenotype thymocyte subpopulations that span across development, from the early thymic progenitors (ETPs) to the most mature subsets of the CD4 and CD8 single positive populations.
Collapse
|
33
|
Jin J, Goldschneider I, Lai L. In Vivo Administration of the Recombinant IL-7/Hepatocyte Growth Factor β Hybrid Cytokine Efficiently Restores Thymopoiesis and Naive T Cell Generation in Lethally Irradiated Mice after Syngeneic Bone Marrow Transplantation. THE JOURNAL OF IMMUNOLOGY 2011; 186:1915-22. [DOI: 10.4049/jimmunol.1001238] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
34
|
Weinreich MA, Jameson SC, Hogquist KA. Postselection thymocyte maturation and emigration are independent of IL-7 and ERK5. THE JOURNAL OF IMMUNOLOGY 2010; 186:1343-7. [PMID: 21187442 DOI: 10.4049/jimmunol.1002238] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The transcription factor Krüppel-like factor 2 (KLF2) controls the emigration of conventional T cells from the thymus through its regulation of the cell surface receptor S1P1. Prior to KLF2 expression, developing T cells require a positive selection signal through the TCR. However, following positive selection there are time, spatial, and maturational events that occur before KLF2 is finally upregulated and emigration occurs. We are interested in determining the signals that upregulate KLF2 and allow thymocytes to emigrate into circulation and whether they are linked to functional maturation. In endothelial cells KLF2 expression has been shown to be dependent on the mitogen-activated protein kinase ERK5. Furthermore, it has been reported that IL-7 signaling leads to the phosphorylation of ERK5. Thus, we hypothesized that IL-7R signaling through ERK5 could drive the expression of KLF2. In this study, we provide evidence that this hypothesis is incorrect. We also found that CD8 lineage specification occurred normally in the absence of IL-7R signaling, in contrast to a recently proposed model. We showed that both CD4 and CD8 T cells complete maturation and express KLF2 independently of ERK5 and IL-7.
Collapse
Affiliation(s)
- Michael A Weinreich
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA
| | | | | |
Collapse
|
35
|
Declining lymphoid progenitor fitness promotes aging-associated leukemogenesis. Proc Natl Acad Sci U S A 2010; 107:21713-8. [PMID: 21098275 DOI: 10.1073/pnas.1005486107] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Aging is associated with the functional decline of cells, tissues, and organs. At the same time, age is the single most important prognostic factor in the development of most human cancers, including chronic myelogenous and acute lymphoblastic leukemias initiated by Bcr-Abl oncogenic translocations. Prevailing paradigms attribute the association between aging and cancers to the accumulation of oncogenic mutations over time, because the accrual of oncogenic events is thought to be the rate-limiting step in initiation and progression of cancers. Conversely, aging-associated functional decline caused by both cell-autonomous and non-cell-autonomous mechanisms is likely to reduce the fitness of stem and progenitor cell populations. This reduction in fitness should be conducive for increased selection of oncogenic mutations that can at least partially alleviate fitness defects, thereby promoting the initiation of cancers. We tested this hypothesis using mouse hematopoietic models. Our studies indicate that the dramatic decline in the fitness of aged B-lymphopoiesis coincides with altered receptor-associated kinase signaling. We further show that Bcr-Abl provides a much greater competitive advantage to old B-lymphoid progenitors compared with young progenitors, coinciding with restored kinase signaling pathways, and that this enhanced competitive advantage translates into increased promotion of Bcr-Abl-driven leukemias. Moreover, impairing IL-7-mediated signaling is sufficient to promote selection for Bcr-Abl-expressing B progenitors. These studies support an unappreciated causative link between aging and cancer: increased selection of oncogenic mutations as a result of age-dependent alterations of the fitness landscape.
Collapse
|
36
|
Cytokine polymorphisms in Th1/Th2 pathway genes, body mass index, and risk of non-Hodgkin lymphoma. Blood 2010; 117:585-90. [PMID: 20952689 DOI: 10.1182/blood-2010-07-295097] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
We conducted a population-based, case-control study in Connecticut women to test the hypothesis that genetic variations in Th1 and Th2 cytokine genes modify the relationship between body mass index (BMI) and risk of non-Hodgkin lymphoma (NHL). Compared with those with BMI less than 25 kg/m(2), women with BMI more than or equal to 25 kg/m(2) had 50% to 90% increased risk of NHL among women who carried IFNGR2 (rs9808753) AA, IL5 (rs2069812) CT/TT, IL7R (rs1494555) AA, and TNF (rs1799724) CC genotypes, but no increased risk among women with IFNGR2 AG/GG, IL5 CC, IL7R AG/GG, and TNF CT/TT genotypes. A significant interaction with BMI was only observed for IFNGR2 (rs9808753 P(forinteraction) = .034) and IL7R (rs1494555 P(forinteraction) = .016) for NHL overall; IL7R (rs1494555 P(forinteraction) = .016) and TNF (1799724 P(forinteraction) = .031) for B-cell lymphoma; and IL5 (rs2069812 P(forinteraction) = .034) for T-cell lymphoma. After stratification by common B-cell lymphoma subtypes, a significant interaction was observed for IFNGR2 (rs9808753 P(forinteraction) = .006), IL13 (rs20541 P(forinteraction) = .019), and IL7R (rs1494555 P(forinteraction) = .012) for marginal zone B-cell lymphoma; IL7R (rs1494555 P(forinteraction) = .017) for small lymphocytic lymphoma/chronic lymphocytic leukemia; and IL12A (rs568408 P(forinteraction) = .013) and TNF (1799724 P(forinteraction) = .04) for follicular lymphoma. The results suggest that common genetic variation in Th1/Th2 pathway genes may modify the association between BMI and NHL risk.
Collapse
|
37
|
Cdc25A-driven proliferation regulates CD62L levels and lymphocyte movement in response to interleukin-7. Exp Hematol 2010; 38:1143-56. [PMID: 20831893 DOI: 10.1016/j.exphem.2010.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2010] [Revised: 08/16/2010] [Accepted: 08/27/2010] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Interleukin-7 (IL-7) is a multifunctional cytokine and a promising immunotherapeutic agent. However, because transient T-cell depletion is an immediate outcome of IL-7 administration at supraphysiological doses, we investigated the mechanism by which the IL-7 proliferative signal transduced through Cdc25A, a key activator of cyclin-dependent kinases, could modulate lymphocyte movement. MATERIALS AND METHODS Employing novel methods of manipulating Cdc25A gene expression, combined with in vitro and in vivo evaluation of IL-7 application, we assessed the expression of activation and homing markers and identified the mechanism by which IL-7 could induce T-cell expansion and alter lymphocyte motility. RESULTS Constitutively active Cdc25A drove T-cell proliferation independently of IL-7 and resulted in an activated phenotype (CD69(hi), CD44(hi)). Conversely, inhibition of Cdc25A resulted in decreased proliferation, reduced expression of activation markers, and upregulation of the lymph node homing molecule, CD62L, which promoted cell adhesion when engaged by ligand. We found that IL-7 prevented the nuclear translocation of the transcription factor, Foxo1, in a manner dependent on the activity of Cdc25A, resulting in decreased levels of CD62L. In vivo administration of IL-7 decreased lymph node cellularity, while treatment with IL-7, premixed with a neutralizing IL-7 antibody (M25), increased total lymph node cells--with more nuclear Foxo1 detected in cells from mice receiving IL-7 + M25. CONCLUSIONS These results are consistent with the model that IL-7 drives Cdc25A-mediated T-cell proliferation, which prevents the nuclear translocation of Foxo1, leading to reduced expression of CD62L and the migration of T cells into circulation.
Collapse
|
38
|
Vicente R, Swainson L, Marty-Grès S, De Barros SC, Kinet S, Zimmermann VS, Taylor N. Molecular and cellular basis of T cell lineage commitment. Semin Immunol 2010; 22:270-5. [PMID: 20630771 DOI: 10.1016/j.smim.2010.04.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2010] [Accepted: 04/23/2010] [Indexed: 12/16/2022]
Abstract
The thymus forms as an alymphoid thymic primordium with T cell differentiation requiring the seeding of this anlage. This review will focus on the characteristics of the hematopoietic progenitors which colonize the thymus and their subsequent commitment/differentiation, both in mice and men. Within the thymus, the interplay between Notch1 and IL-7 signals is crucial for the orchestration of T cell development, but the precise requirements for these factors in murine and human thympoeisis are not synonymous. Recent advances in our understanding of the mechanisms regulating precursor entry and their maintenance in the thymus will also be presented.
Collapse
Affiliation(s)
- Rita Vicente
- Institut de Génétique Moléculaire de Montpellier, CNRS UMR 5535/IFR 122, 34293 Montpellier Cedex 5, France
| | | | | | | | | | | | | |
Collapse
|
39
|
Kurtulus S, Tripathi P, Opferman JT, Hildeman DA. Contracting the 'mus cells'--does down-sizing suit us for diving into the memory pool? Immunol Rev 2010; 236:54-67. [PMID: 20636808 PMCID: PMC2907539 DOI: 10.1111/j.1600-065x.2010.00920.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Maintenance of T-cell homeostasis is critical for normal functioning of the immune system. After thymocyte selection, T cells enter the peripheral lymphoid organs, where they are maintained as naive cells. Transient disruption of homeostasis occurs when naive T cells undergo antigen-driven expansion and acquire effector functions. Effector T cells then either undergo apoptosis (i.e. contraction at the population level) or survive to become memory cells. This apoptotic process is crucial: it resets T-cell homeostasis, promotes protective immunity, and limits autoimmunity. Although initial studies using in vitro models supported a role for death receptor signaling, more recent in vivo studies have implicated Bcl-2 family members as being critical for the culling of T-cell responses. While several Bcl-2 family members likely contribute to T-cell contraction, the pro-apoptotic molecule Bim and its anti-apoptotic antagonist Bcl-2 are essential regulators of the process. This review discusses the progress made in our understanding of the mechanisms underlying contraction of T-cell responses and how some cells avoid this cell death and become memory T cells.
Collapse
Affiliation(s)
- Sema Kurtulus
- Division of Immunobiology in the Department of Pediatrics at the University of Cincinnati and Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Pulak Tripathi
- Division of Immunobiology in the Department of Pediatrics at the University of Cincinnati and Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Joseph T. Opferman
- Department of Biochemistry at St. Jude Children's Research Hospital, Memphis, TN, USA
| | - David A. Hildeman
- Division of Immunobiology in the Department of Pediatrics at the University of Cincinnati and Children's Hospital Medical Center, Cincinnati, Ohio, USA
| |
Collapse
|
40
|
Orr SJ, Roessler S, Quigley L, Chan T, Ford JW, O'Connor GM, McVicar DW. Implications for gene therapy-limiting expression of IL-2R gamma c delineate differences in signaling thresholds required for lymphocyte development and maintenance. THE JOURNAL OF IMMUNOLOGY 2010; 185:1393-403. [PMID: 20592278 DOI: 10.4049/jimmunol.0903528] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
X-linked SCID patients are deficient in functional IL-2Rgamma(c) leading to the loss of IL-2/IL-4/IL-7/IL-9/IL-15/IL-21 signaling and a lack of NK and mature T cells. Patients treated with IL-2Rgamma(c) gene therapy have T cells develop; however, their NK cell numbers remain low, suggesting antiviral responses may be compromised. Similarly, IL-2Rgamma(c)(-/-) mice reconstituted with IL-2Rgamma(c) developed few NK cells, and reconstituted T cells exhibited defective proliferative responses suggesting incomplete recovery of IL-2Rgamma(c) signaling. Given the shift toward self-inactivating long terminal repeats with weaker promoters to control the risk of leukemia, we assessed NK and T cell numbers and function in IL-2Rgamma(c)(-/-) mice reconstituted with limiting amounts of IL-2Rgamma(c). Reconstitution resulted in lower IL-2/-15-mediated STAT5 phosphorylation and proliferation in NK and T cells. However, TCR costimulation restored cytokine-driven T cell proliferation to wild-type levels. Vector modifications that improved IL-2Rgamma(c) levels increased cytokine-induced STAT5 phosphorylation in both populations and increased NK cell proliferation demonstrating that IL-2Rgamma(c) levels are limiting. In addition, although the half-lives of both NK and T cells expressing intermediate levels of IL-2Rgamma(c) are reduced compared with wild-type cells, the reduction in NK cell half-live is much more severe than in T cells. Collectively, these data indicate different IL-2Rgamma(c) signaling thresholds for lymphocyte development and proliferation making functional monitoring imperative during gene therapy. Further, our findings suggest that IL-2Rgamma(c) reconstituted T cells may persist more efficiently than NK cells due to compensation for suboptimal IL-2Rgamma(c) signaling by the TCR.
Collapse
Affiliation(s)
- Selinda J Orr
- Cancer and Inflammation Program, National Cancer Institute-Frederick, Frederick, MD 21702, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
The interaction of LCK and the CD4 co-receptor alters the dose response of T-cells to interleukin-7. Immunol Lett 2010; 131:170-81. [PMID: 20433867 DOI: 10.1016/j.imlet.2010.04.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2009] [Revised: 03/31/2010] [Accepted: 04/20/2010] [Indexed: 11/20/2022]
Abstract
CD8 and CD4 T-cells grow optimally under different concentrations of the cytokine, interleukin-7 (IL-7). While CD8 T-cells expand at high doses of IL-7, CD4 T-cells favor low doses. To examine the reason for the preference of CD4 T-cells for lower doses of the cytokine, we used IL-7 dependent T-cells to study signal transduction upon a range of IL-7 concentrations. We found that the high dose responsiveness of CD8 T-cells to IL-7 could be altered if these cells also expressed CD4. Using the phosphorylation of STAT5 as an indicator of growth, we found that the co-receptor associated kinase, LCK, contributed to phospho-STAT5 levels. Phospho-STAT5 was elevated at high dose IL-7 for CD8 T-cells and at low dose IL-7 for CD4 T-cells, which was reversed upon LCK inhibition. Examining the direct association of LCK with CD4 using a T- cell line that over-expresses CD4, we determined that CD4 could directly sequester LCK. Non-CD4 T-cells were not restricted in this manner and levels of phospho-STAT5 increased proportionally to the IL-7 dose. Our studies, therefore, show that the response of a T-cell to IL-7 can be modulated by the availability of LCK.
Collapse
|
42
|
Marino JH, Tan C, Taylor AA, Bentley C, Van De Wiele CJ, Ranne R, Paliotta M, Broughan TA, Teague TK. Differential IL-7 responses in developing human thymocytes. Hum Immunol 2010; 71:329-33. [PMID: 20074604 PMCID: PMC2896073 DOI: 10.1016/j.humimm.2010.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Revised: 12/23/2009] [Accepted: 01/07/2010] [Indexed: 01/17/2023]
Abstract
Interleukin (IL)-7 is a factor essential for mouse and human thymopoiesis. Mouse thymocytes have altered sensitivities to IL-7 at different developmental stages. CD4/CD8 double positive (DP) mouse thymocytes are shielded from the influence of IL-7 because of loss of CD127 (IL-7Ralpha). In this study, we assessed IL-7 receptor expression and IL-7 signaling in human thymocytes. We found human DP cells to be severely limited in their ability to phosphorylate STAT-5 in response to IL-7. The relative expression levels of the IL-7-inducible proteins Bcl-2 and Mcl-1 were also lower in human DP cells, consistent with a stage-specific decrease in IL-7 responsiveness. IL-7 responses were restored in a subset of cells that matured past the DP stage. Unlike the regulation of IL-7 signaling in mouse thymocytes, loss of IL-7 signaling in human DP cells was not due to absence of CD127, but instead correlated with downregulation of CD132 (common gamma chain).
Collapse
MESH Headings
- Animals
- CD4 Antigens/biosynthesis
- CD8 Antigens/biosynthesis
- Cell Differentiation
- Cells, Cultured
- Child, Preschool
- Humans
- Infant
- Infant, Newborn
- Interleukin Receptor Common gamma Subunit/genetics
- Interleukin Receptor Common gamma Subunit/immunology
- Interleukin Receptor Common gamma Subunit/metabolism
- Interleukin-7/immunology
- Interleukin-7/pharmacology
- Mice
- Precursor Cells, T-Lymphoid/cytology
- Precursor Cells, T-Lymphoid/drug effects
- Precursor Cells, T-Lymphoid/immunology
- Precursor Cells, T-Lymphoid/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Receptors, Interleukin-7/genetics
- Receptors, Interleukin-7/immunology
- Receptors, Interleukin-7/metabolism
- STAT5 Transcription Factor/genetics
- STAT5 Transcription Factor/metabolism
- Signal Transduction/drug effects
- Signal Transduction/immunology
- Thymus Gland/cytology
- Thymus Gland/immunology
Collapse
Affiliation(s)
- Julie H Marino
- Department of Surgery, University of Oklahoma College of Medicine, Tulsa, OK, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Teague TK, Tan C, Marino JH, Davis BK, Taylor AA, Huey RW, Van De Wiele CJ. CD28 expression redefines thymocyte development during the pre-T to DP transition. Int Immunol 2010; 22:387-97. [DOI: 10.1093/intimm/dxq020] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
44
|
|
45
|
Park JH, Adoro S, Guinter T, Erman B, Alag AS, Catalfamo M, Kimura MY, Cui Y, Lucas PJ, Gress RE, Kubo M, Hennighausen L, Feigenbaum L, Singer A. Signaling by intrathymic cytokines, not T cell antigen receptors, specifies CD8 lineage choice and promotes the differentiation of cytotoxic-lineage T cells. Nat Immunol 2010; 11:257-64. [PMID: 20118929 PMCID: PMC3555225 DOI: 10.1038/ni.1840] [Citation(s) in RCA: 181] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Accepted: 12/29/2009] [Indexed: 12/11/2022]
Abstract
Immature CD4(+)CD8(+) (double-positive (DP)) thymocytes are signaled via T cell antigen receptors (TCRs) to undergo positive selection and become responsive to intrathymic cytokines such as interleukin 7 (IL-7). We report here that cytokine signaling is required for positively selected thymocytes to express the transcription factor Runx3, specify CD8 lineage choice and differentiate into cytotoxic-lineage T cells. In DP thymocytes genetically engineered to be cytokine responsive, IL-7 signaling induced TCR-unsignaled DP thymocytes to express Runx3 and to differentiate into mature CD8(+) T cells, completely circumventing positive selection. We conclude that TCR-mediated positive selection converts DP cells into cytokine-responsive thymocytes, but it is subsequent signaling by intrathymic cytokines that specifies CD8 lineage choice and promotes differentiation into cytotoxic-lineage T cells.
Collapse
Affiliation(s)
- Jung-Hyun Park
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Li LX, Goetz CA, Katerndahl CDS, Sakaguchi N, Farrar MA. A Flt3- and Ras-dependent pathway primes B cell development by inducing a state of IL-7 responsiveness. THE JOURNAL OF IMMUNOLOGY 2010; 184:1728-36. [PMID: 20065110 DOI: 10.4049/jimmunol.0903023] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ras plays an important role in B cell development. However, the stage at which Ras governs B cell development remains unclear. Moreover, the upstream receptors and downstream effectors of Ras that govern B cell differentiation remain undefined. Using mice that express a dominant-negative form of Ras, we demonstrate that Ras-mediated signaling plays a critical role in the development of common lymphoid progenitors. This developmental block parallels that found in flt3(-/-) mice, suggesting that Flt3 is an important upstream activator of Ras in early B cell progenitors. Ras inhibition impaired proliferation of common lymphoid progenitors and pre-pro-B cells but not pro-B cells. Rather, Ras promotes STAT5-dependent pro-B cell differentiation by enhancing IL-7Ralpha levels and suppressing socs2 and socs3 expression. Our results suggest a model in which Flt3/Ras-dependent signals play a critical role in B cell development by priming early B cell progenitors for subsequent STAT5-dependent B cell differentiation.
Collapse
Affiliation(s)
- Lin-Xi Li
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455, USA
| | | | | | | | | |
Collapse
|
47
|
Abstract
The transcription factor Signal Tranducer and Activator of Transcription 5 (STAT5) plays an important role in numerous biological processes including, but not limited to, (i) homeostasis of hematopoietic stem cells, (ii) development of essentially all blood cell lineages, (iii) growth hormone effects, (iv) differentiation of mammary epithelium, and (v) central nervous system control of metabolism. Two key tools for deciphering STAT5 biology have involved the use of mice in which the Stat5a and Stat5b genes can be conditionally deleted (Stat5(FL/FL) mice) and the development of systems in which STAT5a or STAT5b is rendered constitutively active. In this chapter, the distinct mechanisms that have been developed to render STAT5 constitutively active and their use in probing biological processes are discussed.
Collapse
Affiliation(s)
- Michael A Farrar
- Center for Immunology, Masonic Cancer Center, University of Minnesota, Minneapolis, USA
| |
Collapse
|
48
|
Abstract
Interleukin-7 (IL-7) is required for the development and survival of T cells and plays a critical role in modulating T-cell homeostasis. This review will address current understanding of IL-7 biology, review recent clinical experiences and discuss potential future clinical applications of IL-7, or IL-7 blockade, in the setting of disease.
Collapse
Affiliation(s)
- C M Capitini
- Immunology Section, Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | | | | |
Collapse
|
49
|
González-García S, García-Peydró M, Martín-Gayo E, Ballestar E, Esteller M, Bornstein R, de la Pompa JL, Ferrando AA, Toribio ML. CSL-MAML-dependent Notch1 signaling controls T lineage-specific IL-7R{alpha} gene expression in early human thymopoiesis and leukemia. ACTA ACUST UNITED AC 2009; 206:779-91. [PMID: 19349467 PMCID: PMC2715119 DOI: 10.1084/jem.20081922] [Citation(s) in RCA: 116] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Notch1 activation is essential for T-lineage specification of lymphomyeloid progenitors seeding the thymus. Progression along the T cell lineage further requires cooperative signaling provided by the interleukin 7 receptor (IL-7R), but the molecular mechanisms responsible for the dynamic and lineage-specific regulation of IL-7R during thymopoiesis are unknown. We show that active Notch1 binds to a conserved CSL-binding site in the human IL7R gene promoter and critically regulates IL7R transcription and IL-7R α chain (IL-7Rα) expression via the CSL–MAML complex. Defective Notch1 signaling selectively impaired IL-7Rα expression in T-lineage cells, but not B-lineage cells, and resulted in a compromised expansion of early human developing thymocytes, which was rescued upon ectopic IL-7Rα expression. The pathological implications of these findings are demonstrated by the regulation of IL-7Rα expression downstream of Notch1 in T cell leukemias. Thus, Notch1 controls early T cell development, in part by regulating the stage- and lineage-specific expression of IL-7Rα.
Collapse
Affiliation(s)
- Sara González-García
- Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Morley SC, Weber KS, Kao H, Allen PM. Protein kinase C-theta is required for efficient positive selection. THE JOURNAL OF IMMUNOLOGY 2008; 181:4696-708. [PMID: 18802072 DOI: 10.4049/jimmunol.181.7.4696] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Protein kinase C-theta (PKCtheta) is critical for TCR-initiated signaling in mature T cells, but initial reports found no requirement for PKCtheta in thymocyte development. Thymocytes and peripheral T cells utilize many of the same signaling components and, given the significant role of PKCtheta in peripheral T cells, it was surprising that it was not involved at all in TCR signaling in thymocytes. We decided to re-evaluate the role of PKCtheta in thymocyte development using the well-characterized class II-restricted n3.L2 TCR-transgenic TCR model. Analysis of n3.L2 PKCtheta(-/-) mice revealed a defect in thymocyte-positive selection, resulting in a 50% reduction in the generation of n3.L2 CD4 single-positive thymocytes and n3.L2 CD4 mature T cells. Competition between n3.L2 WT and n3.L2 PKCtheta(-/-) thymocytes in bone marrow chimeras revealed a more dramatic defect, with a >80% reduction in generation of n3.L2 CD4 single-positive thymocytes derived from PKCtheta(-/-) mice. Inefficient positive selection of n3.L2 PKCtheta(-/-) CD4 single-positive cells resulted from "weaker" signaling through the TCR and correlated with diminished ERK activation. The defect in positive selection was not complete in the PKCtheta(-/-) mice, most likely accounted for by compensation by other PKC isoforms not evident in peripheral cells. Similar decreased positive selection of both CD4 and CD8 single-positive thymocytes was also seen in nontransgenic PKCtheta(-/-) mice. These findings now place PKCtheta as a key signaling molecule in the positive selection of thymocytes as well as in the activation of mature T cells.
Collapse
Affiliation(s)
- Sharon Celeste Morley
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | | | | | |
Collapse
|