1
|
Steier Z, Kim EJY, Aylard DA, Robey EA. The CD4 Versus CD8 T Cell Fate Decision: A Multiomics-Informed Perspective. Annu Rev Immunol 2024; 42:235-258. [PMID: 38271641 DOI: 10.1146/annurev-immunol-083122-040929] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
The choice of developing thymocytes to become CD8+ cytotoxic or CD4+ helper T cells has been intensely studied, but many of the underlying mechanisms remain to be elucidated. Recent multiomics approaches have provided much higher resolution analysis of gene expression in developing thymocytes than was previously achievable, thereby offering a fresh perspective on this question. Focusing on our recent studies using CITE-seq (cellular indexing of transcriptomes and epitopes) analyses of mouse thymocytes, we present a detailed timeline of RNA and protein expression changes during CD8 versus CD4 T cell differentiation. We also revisit our current understanding of the links between T cell receptor signaling and expression of the lineage-defining transcription factors ThPOK and RUNX3. Finally, we propose a sequential selection model to explain the tight linkage between MHC-I versus MHC-II recognition and T cell lineage choice. This model incorporates key aspects of previously proposed kinetic signaling, instructive, and stochastic/selection models.
Collapse
Affiliation(s)
- Zoë Steier
- Department of Bioengineering and Center for Computational Biology, University of California, Berkeley, California, USA
- Graduate Program in Bioengineering, University of California, Berkeley, and University of California, San Francisco, Berkeley and San Francisco, California, USA
- Current affiliation: Institute for Medical Engineering and Science, Massachusetts Institute of Technology; Broad Institute of MIT and Harvard; and Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
| | - Esther Jeong Yoon Kim
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, California, USA;
| | - Dominik A Aylard
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, California, USA;
| | - Ellen A Robey
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, California, USA;
| |
Collapse
|
2
|
Orlova E, Loginova O, Loginova N, Shekhmametyev R, Shirshev S. The Hypoxia Affects the Main Thymocyte Subset Distributions in Congenital Heart Diseases. TURKISH JOURNAL OF IMMUNOLOGY 2022. [DOI: 10.4274/tji.galenos.2022.92485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
|
3
|
A Validation Study on Immunophenotypic Differences in T-lymphocyte Chromosomal Radiosensitivity between Newborns and Adults in South Africa. RADIATION 2021. [DOI: 10.3390/radiation2010001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Children have an increased risk of developing radiation-induced secondary malignancies compared to adults, due to their high radiosensitivity and longer life expectancy. In contrast to the epidemiological evidence, there is only a handful of radiobiology studies which investigate the difference in radiosensitivity between children and adults at a cellular level. In this study, the previous results on the potential age dependency in chromosomal radiosensitivity were validated again by means of the cytokinesis-block micronucleus (CBMN) assay in T-lymphocytes isolated from the umbilical cord and adult peripheral blood of a South African population. The isolated cells were irradiated with 60Co γ-rays at doses ranging from 0.5 Gy to 4 Gy. Increased radiosensitivities of 34%, 42%, 29%, 26% and 16% were observed for newborns compared to adults at 0.5, 1, 2, 3 and 4 Gy, respectively. An immunophenotypic evaluation with flow cytometry revealed a significant change in the fraction of naïve (CD45RA+) T-lymphocytes in CD4+ and CD8+ T-lymphocytes with age. Newborns co-expressed an average of 91.05% CD45RA+ (range: 80.80–98.40%) of their CD4+ cells, while this fraction decreased to an average of 39.08% (range: 12.70–58.90%) for adults. Similar observations were made for CD8+ cells. This agrees with previous published results that the observed differences in chromosomal radiosensitivity between newborn and adult T-lymphocytes could potentially be linked to their immunophenotypic profiles.
Collapse
|
4
|
Boulet S, Odagiu L, Dong M, Lebel MÈ, Daudelin JF, Melichar HJ, Labrecque N. NR4A3 Mediates Thymic Negative Selection. THE JOURNAL OF IMMUNOLOGY 2021; 207:1055-1064. [PMID: 34312259 DOI: 10.4049/jimmunol.1901228] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 06/16/2021] [Indexed: 11/19/2022]
Abstract
Central tolerance aims to limit the production of T lymphocytes bearing TCR with high affinity for self-peptide presented by MHC molecules. The accumulation of thymocytes with such receptors is limited by negative selection or by diversion into alternative differentiation, including T regulatory cell commitment. A role for the orphan nuclear receptor NR4A3 in negative selection has been suggested, but its function in this process has never been investigated. We find that Nr4a3 transcription is upregulated in postselection double-positive thymocytes, particularly those that have received a strong selecting signal and are destined for negative selection. Indeed, we found an accumulation of cells bearing a negative selection phenotype in NR4A3-deficient mice as compared with wild-type controls, suggesting that Nr4a3 transcriptional induction is necessary to limit accumulation of self-reactive thymocytes. This is consistent with a decrease of cleaved caspase-3+-signaled thymocytes and more T regulatory and CD4+Foxp3-HELIOS+ cells in the NR4A3-deficient thymus. We further tested the role for NR4A3 in negative selection by reconstituting transgenic mice expressing the OVA Ag under the control of the insulin promoter with bone marrow cells from OT-I Nr4a3 +/+ or OT-I Nr4a3 -/- mice. Accumulation of autoreactive CD8 thymocytes and autoimmune diabetes developed only in the absence of NR4A3. Overall, our results demonstrate an important role for NR4A3 in T cell development.
Collapse
Affiliation(s)
- Salix Boulet
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada
| | - Livia Odagiu
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada; and
| | - Mengqi Dong
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada.,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada; and
| | - Marie-Ève Lebel
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada
| | | | - Heather J Melichar
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada.,Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
| | - Nathalie Labrecque
- Maisonneuve-Rosemont Hospital Research Center, Montreal, Quebec, Canada; .,Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, Quebec, Canada; and.,Département de Médecine, Université de Montréal, Montreal, Quebec, Canada
| |
Collapse
|
5
|
Liu QM, Xu SS, Li L, Pan TM, Shi CL, Liu H, Cao MJ, Su WJ, Liu GM. In vitro and in vivo immunomodulatory activity of sulfated polysaccharide from Porphyra haitanensis. Carbohydr Polym 2017; 165:189-196. [PMID: 28363539 DOI: 10.1016/j.carbpol.2017.02.032] [Citation(s) in RCA: 85] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/08/2017] [Accepted: 02/09/2017] [Indexed: 11/16/2022]
Abstract
The immunoregulatory activity of sulfated polysaccharide from Porphyra haitanensis (PHPS) was investigated in a RAW264.7 macrophages cell model and a BALB/c murine model. The subpopulation of dendritic cells (DCs) and regulatory T cells (Tregs) from PHPS-treated mice splenocytes were also measured by flow cytometry. Consistent with previous reports, we showed that PHPS increased the phagocytosis of RAW264.7 macrophages, and enhanced the secretion of interleukin (IL)-6, IL-10 and tumor necrosis factor-α (TNF-α). Meanwhile, PHPS induced the production of nitric oxide via the Jun N-terminal kinase (JNK) and the Janus kinase (JAK2) signaling pathways in RAW264.7 macrophages. Furthermore, PHPS promoted the proliferation of mice lymphocytes, inducing the generation of TNF-α and IL-10 in vivo, as well as the subpopulation of CD4+ splenic T lymphocytes, DCs, and Tregs. These results indicated that PHPS plays key roles in immunoregulation and may be apply to develop new health foods.
Collapse
Affiliation(s)
- Qing-Mei Liu
- College of Food and Biological Engineering, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen 361021, Fujian, PR China
| | - Sha-Sha Xu
- College of Food and Biological Engineering, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen 361021, Fujian, PR China
| | - Long Li
- College of Food and Biological Engineering, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen 361021, Fujian, PR China
| | - Tzu-Ming Pan
- Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, No. 1, Sec. 4, Roosevelt Road, Taipei 10617, Taiwan
| | - Chao-Lan Shi
- College of Food and Biological Engineering, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen 361021, Fujian, PR China
| | - Hong Liu
- College of Food and Biological Engineering, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen 361021, Fujian, PR China
| | - Min-Jie Cao
- College of Food and Biological Engineering, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen 361021, Fujian, PR China
| | - Wen-Jin Su
- College of Food and Biological Engineering, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen 361021, Fujian, PR China
| | - Guang-Ming Liu
- College of Food and Biological Engineering, Fujian Collaborative Innovation Center for Exploitation and Utilization of Marine Biological Resources, Jimei University, 43 Yindou Road, Xiamen 361021, Fujian, PR China.
| |
Collapse
|
6
|
The catalytic activity of the mitogen-activated protein kinase extracellular signal-regulated kinase 3 is required to sustain CD4+ CD8+ thymocyte survival. Mol Cell Biol 2014; 34:3374-87. [PMID: 25002529 DOI: 10.1128/mcb.01701-13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Extracellular signal-regulated kinase 3 (ERK3) is an atypical member of the mitogen-activated protein kinase (MAPK) family whose function is largely unknown. Given the central role of MAPKs in T cell development, we hypothesized that ERK3 may regulate thymocyte development. Here we have shown that ERK3 deficiency leads to a 50% reduction in CD4(+) CD8(+) (DP) thymocyte number. Analysis of hematopoietic chimeras revealed that the reduction in DP thymocytes is intrinsic to hematopoietic cells. We found that early thymic progenitors seed the Erk3(-/-) thymus and can properly differentiate and proliferate to generate DP thymocytes. However, ERK3 deficiency results in a decrease in the DP thymocyte half-life, associated with a higher level of apoptosis. As a consequence, ERK3-deficient DP thymocytes are impaired in their ability to make successful secondary T cell receptor alpha (TCRα) gene rearrangement. Introduction of an already rearranged TCR transgene restores thymic cell number. We further show that knock-in of a catalytically inactive allele of Erk3 fails to rescue the loss of DP thymocytes. Our results uncover a unique role for ERK3, dependent on its kinase activity, during T cell development and show that this atypical MAPK is essential to sustain DP survival during RAG-mediated rearrangements.
Collapse
|
7
|
Marquis M, Boulet S, Mathien S, Rousseau J, Thébault P, Daudelin JF, Rooney J, Turgeon B, Beauchamp C, Meloche S, Labrecque N. The non-classical MAP kinase ERK3 controls T cell activation. PLoS One 2014; 9:e86681. [PMID: 24475167 PMCID: PMC3903551 DOI: 10.1371/journal.pone.0086681] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 12/13/2013] [Indexed: 11/19/2022] Open
Abstract
The classical mitogen-activated protein kinases (MAPKs) ERK1 and ERK2 are activated upon stimulation of cells with a broad range of extracellular signals (including antigens) allowing cellular responses to occur. ERK3 is an atypical member of the MAPK family with highest homology to ERK1/2. Therefore, we evaluated the role of ERK3 in mature T cell response. Mouse resting T cells do not transcribe ERK3 but its expression is induced in both CD4⁺ and CD8⁺ T cells following T cell receptor (TCR)-induced T cell activation. This induction of ERK3 expression in T lymphocytes requires activation of the classical MAPK ERK1 and ERK2. Moreover, ERK3 protein is phosphorylated and associates with MK5 in activated primary T cells. We show that ERK3-deficient T cells have a decreased proliferation rate and are impaired in cytokine secretion following in vitro stimulation with low dose of anti-CD3 antibodies. Our findings identify the atypical MAPK ERK3 as a new and important regulator of TCR-induced T cell activation.
Collapse
Affiliation(s)
- Miriam Marquis
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology and Immunology, University of Montreal, Quebec, Canada
| | - Salix Boulet
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
| | - Simon Mathien
- Institute of Research in Immunology and Cancer, University of Montreal, Quebec, Canada
| | - Justine Rousseau
- Department of Pharmacology and Molecular Biology, University of Montreal, Quebec, Canada
- Institute of Research in Immunology and Cancer, University of Montreal, Quebec, Canada
| | - Paméla Thébault
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
| | | | - Julie Rooney
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
| | - Benjamin Turgeon
- Department of Pharmacology and Molecular Biology, University of Montreal, Quebec, Canada
- Institute of Research in Immunology and Cancer, University of Montreal, Quebec, Canada
| | | | - Sylvain Meloche
- Department of Pharmacology and Molecular Biology, University of Montreal, Quebec, Canada
- Institute of Research in Immunology and Cancer, University of Montreal, Quebec, Canada
| | - Nathalie Labrecque
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, Quebec, Canada
- Department of Microbiology, Infectiology and Immunology, University of Montreal, Quebec, Canada
- Department of Medicine, University of Montreal, Quebec, Canada
| |
Collapse
|
8
|
Abstract
During alphabeta T cell development, cells diverge into alternate CD4 helper and CD8(+) cytotoxic T cell lineages. The precise correlation between a T cell's CD8 and CD4 choice and its TCR specificity to class I or class II MHC was noted more than 20 years ago, and establishing the underlying mechanism has remained a focus of intense study since then. This review deals with three formerly discrete topics that are gradually becoming interconnected: the role of TCR signaling in lineage commitment, the regulation of expression of the CD4 and CD8 genes, and transcriptional regulation of lineage commitment. It is widely accepted that TCR signaling exerts a decisive influence on lineage choice, although the underlying mechanism remains intensely debated. Current evidence suggests that both duration and intensity of TCR signaling may control lineage choice, as proposed by the kinetic signaling and quantitative instructive models, respectively. Alternate expression of the CD4 and CD8 genes is the most visible manifestation of lineage choice, and much progress has been made in defining the responsible cis elements and transcription factors. Finally, important clues to the molecular basis of lineage commitment have been provided by the recent identification of the transcription factor ThPOK as a key regulator of lineage choice. ThPOK is selectively expressed in class II-restricted cells at the CD4(+)8(lo) stage and is necessary and sufficient for development to the CD4 lineage. Given the central role of ThPOK in lineage commitment, understanding its upstream regulation and downstream gene targets is expected to reveal further important aspects of the molecular machinery underlying lineage commitment.
Collapse
Affiliation(s)
- Xi He
- Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111, USA
| | | | | |
Collapse
|
9
|
Komaniwa S, Hayashi H, Kawamoto H, Sato SB, Ikawa T, Katsura Y, Udaka K. Lipid-mediated presentation of MHC class II molecules guides thymocytes to the CD4 lineage. Eur J Immunol 2008; 39:96-112. [DOI: 10.1002/eji.200838796] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
10
|
Adoro S, Erman B, Sarafova SD, Van Laethem F, Park JH, Feigenbaum L, Singer A. Targeting CD4 coreceptor expression to postselection thymocytes reveals that CD4/CD8 lineage choice is neither error-prone nor stochastic. THE JOURNAL OF IMMUNOLOGY 2008; 181:6975-83. [PMID: 18981117 DOI: 10.4049/jimmunol.181.10.6975] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The mechanism by which CD4/CD8 lineage choice is coordinated with TCR specificity during positive selection remains an unresolved problem in immunology. The stochastic/selection model proposes that CD4/CD8 lineage choice in TCR-signaled CD4(+)CD8(+) thymocytes occurs randomly and therefore is highly error-prone. This perspective is strongly supported by "coreceptor rescue" experiments in which transgenic CD4 coreceptors were ectopically expressed on thymocytes throughout their development and caused significant numbers of cells bearing MHC-II-specific TCR to differentiate into mature, CD8 lineage T cells. However, it is not known if forced coreceptor expression actually rescued positively selected thymocytes making an incorrect lineage choice or if it influenced developing thymocytes into making an incorrect lineage choice. We have now reassessed coreceptor rescue and the concept that lineage choice is highly error-prone with a novel CD4 transgene (referred to as E8(I)-CD4) that targets expression of transgenic CD4 coreceptors specifically to thymocytes that have already undergone positive selection and adopted a CD8 lineage fate. Unlike previous CD4 transgenes, the E8(I)-CD4 transgene has no effect on early thymocyte development and cannot itself influence CD4/CD8 lineage choice. We report that the E8(I)-CD4 transgene did in fact induce expression of functional CD4 coreceptor proteins on newly arising CD8 lineage thymocytes precisely at the point in thymic development that transgenic CD4 coreceptors would putatively rescue MHC-II-specific thymocytes that incorrectly adopted the CD8 lineage. However, the E8(I)-CD4 transgene did not reveal any MHC-II-selected thymocytes that adopted the CD8 lineage fate. These results demonstrate that CD4/CD8 lineage choice is neither error-prone nor stochastic.
Collapse
Affiliation(s)
- Stanley Adoro
- Experimental Immunology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
11
|
Muroi S, Naoe Y, Miyamoto C, Akiyama K, Ikawa T, Masuda K, Kawamoto H, Taniuchi I. Cascading suppression of transcriptional silencers by ThPOK seals helper T cell fate. Nat Immunol 2008; 9:1113-21. [PMID: 18776907 DOI: 10.1038/ni.1650] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Accepted: 07/31/2008] [Indexed: 11/09/2022]
Abstract
CD4 and the transcription factor ThPOK are essential for the differentiation of major histocompatibility complex class II-restricted thymocytes into the helper T cell lineage; their genes (Cd4 and Zbtb7b (called 'ThPOK' here)) are repressed by transcriptional silencer elements in cytotoxic T cells. The molecular mechanisms regulating expression of these genes during helper T cell lineage differentiation remain unknown. Here we showed that inefficient upregulation of ThPOK, induced by removal of the proximal enhancer from the ThPOK locus, resulted in the transdifferentiation of helper lineage-specified cells into the cytotoxic T cell lineage. Furthermore, direct antagonism by ThPOK of the Cd4 and ThPOK silencers generated two regulatory loops that initially inhibited Cd4 downregulation and later stabilized ThPOK expression. Our results show how an initial lineage-specification signal can be amplified and stabilized during the lineage-commitment process.
Collapse
Affiliation(s)
- Sawako Muroi
- Laboratory for Transcriptional Regulation, RIKEN Research Center for Allergy and Immunology, Yokohama, Japan
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Egawa T, Littman DR. ThPOK acts late in specification of the helper T cell lineage and suppresses Runx-mediated commitment to the cytotoxic T cell lineage. Nat Immunol 2008; 9:1131-9. [PMID: 18776905 PMCID: PMC2666788 DOI: 10.1038/ni.1652] [Citation(s) in RCA: 161] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2008] [Accepted: 08/06/2008] [Indexed: 11/09/2022]
Abstract
The transcription factor ThPOK has been shown to be required and sufficient for CD4+CD8− thymocyte generation, yet the mechanism through which ThPOK orchestrates CD4 helper T cell lineage differentiation remains unclear. Here we utilized reporter mice to track expression of transcription factors in developing thymocytes. Distal promoter-driven Runx3 (Runx3d) expression was restricted to MHC class I-selected thymocytes. In ThPOK-deficient mice, Runx3d expression was de-repressed in MHCII-selected thymocytes, contributing to their redirection to the CD8 T cell lineage. In the absence of both ThPOK and Runx, redirection was prevented and cells potentially belonging to the CD4 lineage, presumably specified independently of ThPOK, were generated. Our results suggest that MHCII-selected thymocytes are directed towards the CD4 lineage independently of ThPOK, but require ThPOK to prevent Runx-dependent differentiation towards the CD8 lineage.
Collapse
Affiliation(s)
- Takeshi Egawa
- Molecular Pathogenesis Program, The Helen and Martin Kimmel Center for Biology and Medicine at the Skirball Institute of Biomolecular Medicine, New York University School of Medicine, New York, NY 10016, USA
| | | |
Collapse
|
13
|
Nepomnaschy I, Lombardi G, Bekinschtein P, Berguer P, Francisco V, De Almeida J, Buggiano V, Pasqualini C, Piazzon I. Alterations during Positive Selection in the Thymus of nackt CD4-Deficient Mice. Scand J Immunol 2008. [DOI: 10.1111/j.1365-3083.2000.00818.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
14
|
He X, Park K, Wang H, He X, Zhang Y, Hua X, Li Y, Kappes DJ. CD4-CD8 lineage commitment is regulated by a silencer element at the ThPOK transcription-factor locus. Immunity 2008; 28:346-58. [PMID: 18342007 DOI: 10.1016/j.immuni.2008.02.006] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2007] [Revised: 01/28/2008] [Accepted: 02/07/2008] [Indexed: 10/22/2022]
Abstract
The transcription factor ThPOK is necessary and sufficient to trigger adoption of the CD4 lymphocyte fate. Here we investigate the regulation of ThPOK expression and its subsequent control of CD4+ T cell commitment. Treatment of immature thymocytes with anti-TCR (T cell receptor) showed that TCR signals were important in ThPOK induction and that the CD4+8lo stage was the likely target of the inductive TCR signal. We identified at the ThPOK locus a key distal regulatory element (DRE) that mediated its differential expression in class I- versus II-restricted CD4+8lo thymocytes. The DRE was both necessary for suppression of ThPOK expression in class I-restricted thymocytes and sufficient for its induction in class II-restricted thymocytes. Mutagenesis analysis defined an essential 80bp core DRE sequence and its potential regulatory motifs. We propose a silencer-dependent model of lineage choice, whereby inactivation of the DRE silencer by a strong TCR signal leads to CD4 commitment, whereas continued silencer activity leads to CD8 commitment.
Collapse
Affiliation(s)
- Xi He
- Fox Chase Cancer Center, 7701 Burholme Ave., Philadelphia, PA 19111, USA
| | | | | | | | | | | | | | | |
Collapse
|
15
|
He X, Kappes DJ. CD4/CD8 lineage commitment: light at the end of the tunnel? Curr Opin Immunol 2006; 18:135-42. [PMID: 16480861 DOI: 10.1016/j.coi.2006.02.003] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Accepted: 02/01/2006] [Indexed: 11/17/2022]
Abstract
Two surprisingly clear results have emerged in the past year that suggest that the seemingly intractable problem of CD4/CD8 lineage commitment might eventually be resolved. Manipulating expression of the CD4 and CD8 coreceptors has long been a favorite method to examine the influence of T-cell receptor signalling on lineage commitment. An elegant new twist on this approach now shows that it is all a matter of timing. Thus, termination of CD4 expression after the initiation of positive selection is sufficient to cause complete redirection of class II-restricted thymocytes to the CD8 lineage, which strongly supports quantitative instructive models of lineage commitment. Progress in the field has been significantly hampered by ignorance of the underlying intracellular pathways. Two independent groups, which employed old-fashioned genetics versus new-fangled microarray technology, have now identified the same transcription factor, Th-POK, as a key regulator of alternate lineage commitment. The presence of this factor directs positively selected thymocytes to the CD4 lineage, whereas its absence causes default development to the CD8 lineage.
Collapse
Affiliation(s)
- Xiao He
- Fox Chase Cancer Center, 7701 Burholme Avenue, Philadelphia, PA 19111, USA
| | | |
Collapse
|
16
|
Abstract
The molecular basis of CD4:CD8 lineage commitment, in particular the mechanism by which the precise correlation between lineage choice and T-cell receptor (TCR) specificity toward class I or II major histocompatibility complex is achieved, remains controversial. Both stochastic/selective and instructive models in various forms have been proposed to explain this correlation. The two main experimental approaches previously employed to elucidate this process have focused on the beginning and end of the process, i.e. the influence of TCR signaling and the alternate transcriptional control of the CD4 and CD8 loci during commitment. The recent finding that the transcription factor Th-POK is necessary and sufficient for CD4 commitment has now provided a direct entry point for studying the intracellular pathways that govern lineage commitment. Here, we review data leading to the identification and characterization of this factor and discuss the implications of these studies in the context of current models of lineage commitment.
Collapse
|
17
|
Marrero I, Benvenutti LA, Kalil J, Coelho V. Autoreactivity to self H-2Kb peptides in TAP1 mice. Intravenous administration of H-2Kb class I-derived peptides induces long-term survival of grafts from C57BL/6 donors. Immunology 2005; 115:484-94. [PMID: 16011517 PMCID: PMC1782177 DOI: 10.1111/j.1365-2567.2005.02182.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
We and others have previously shown that TAP1-/- mice (H-2b) reject grafts from donors without major histocompatibility complex (MHC) disparity that express wild-type levels of H-2b class I molecules (C57BL/6, TAP1+/+ mice). In this same model, we also showed that subcutaneous priming of TAP1-/- mice with synthetic peptides derived from the H-2Kb molecule accelerated graft rejection and that in vivo depletion of CD4+ T cells induced a significant prolongation of graft survival, suggesting an important role for CD4 T cells. We hypothesize that, in this model, rejection is triggered by the recognition of class I molecules or derived peptides, in an inflammatory microenvironment, by a functionally altered autoreactive T-cell repertoire that escapes the control of peripheral regulatory mechanisms. In the present study, we analysed the cellular autoreactivity induced by synthetic peptides derived from the H-2Kb sequence in naive and TAP1-/- mice transplanted with C57BL/6 grafts, and investigated whether intravenous modulation of autoreactivity to these peptides induced transplantation tolerance. We showed that TAP1-/- mice have peripheral autoreactive T cells that recognize H-2Kb peptides. A significant amplification of proliferation against these peptides was detected in TAP1-/- mice that rejected grafts, indicating that the inflammatory context of transplantation induced peripheral expansion of these autoreactive T cells. Furthermore, intravenous injection of H-2Kb-derived peptides significantly prolonged graft survival in some animals. In these mice (> 100 days graft survival), we observed intragraft inhibition of interferon-gamma and interleukin-10 expression, suggesting that these cytokines have an active role during the rejection. In conclusion, our present data indicate that inflammatory autoreactive T cells directed against H-2Kb peptides can be inhibited in the periphery to prolong graft survival in TAP1-/- mice.
Collapse
Affiliation(s)
- Idania Marrero
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| | - Luiz Alberto Benvenutti
- Department of Pathology, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
| | - Jorge Kalil
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| | - Verônica Coelho
- Immunology Laboratory, Heart Institute (InCor), São Paulo University School of MedicineSão Paulo, Brazil
- Institute for Investigation in Immunology (iii), Millennium InstituteSão Paulo, Brazil
| |
Collapse
|
18
|
Abstract
The mechanism of CD4-CD8 lineage commitment, which ensures the correlation between T cell receptor specificity and adoption of the T killer or T helper phenotype, has long been the subject of intense debate. Various approaches are slowly elucidating the underlying molecular pathways. Analysis of the function of T cell receptor signaling (the 'top-down' approach) supports the view that differences in signal strength and/or duration 'instruct' alternative commitment. Analysis of the transcriptional regulation of the genes encoding CD4 and CD8 (the 'bottom-up' approach) has identified critical cis-acting elements and their interacting factors. Finally, identification of the transcription factor Th-POK as a central component of the CD4 lineage-determining pathway has provided a new starting point from which to unravel this intriguing process 'from the inside out'.
Collapse
|
19
|
Bosselut R, Guinter TI, Sharrow SO, Singer A. Unraveling a revealing paradox: Why major histocompatibility complex I-signaled thymocytes "paradoxically" appear as CD4+8lo transitional cells during positive selection of CD8+ T cells. J Exp Med 2003; 197:1709-19. [PMID: 12810689 PMCID: PMC2193957 DOI: 10.1084/jem.20030170] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The mechanism by which T cell receptor specificity determines the outcome of the CD4/CD8 lineage decision in the thymus is not known. An important clue is the fact that major histocompatibility complex (MHC)-I-signaled thymocytes paradoxically appear as CD4+8lo transitional cells during their differentiation into CD8+ T cells. Lineage commitment is generally thought to occur at the CD4+8+ (double positive) stage of differentiation and to result in silencing of the opposite coreceptor gene. From this perspective, the appearance of MHC-I-signaled thymocytes as CD4+8lo cells would be due to effects on CD8 surface protein expression, not CD8 gene expression. But contrary to this perspective, this study demonstrates that MHC-I-signaled thymocytes appear as CD4+8lo cells because of transient down-regulation of CD8 gene expression, not because of changes in CD8 surface protein expression or distribution. This study also demonstrates that initial cessation of CD8 gene expression in MHC-I-signaled thymocytes is not necessarily indicative of commitment to the CD4+ T cell lineage, as such thymocytes retain the potential to differentiate into CD8+ T cells. These results challenge classical concepts of lineage commitment but fulfill predictions of the kinetic signaling model.
Collapse
Affiliation(s)
- Remy Bosselut
- Laboratory of Immune Cell Biology, National Cancer Institute, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
20
|
Abstract
The Ikaros gene encodes a zinc finger transcription factor that is selectively expressed by all hematopoietic cells. Although Ikaros is required for lymphocyte differentiation, its role in the myeloid lineage is unclear. We show here that Ikaros expression is temporally regulated during neutrophil differentiation: Ikaros is primarily expressed at immature stages and significantly less so in mature neutrophils. Furthermore Ik(L/L) mice, harboring a hypomorphic mutation at the Ikaros locus, exhibit several defects during neutrophil differentiation. (1) Ik(L/L) fetal livers contain high numbers of neutrophil lineage cells, and this increase is reflected in the number of GM-CSF-dependent progenitor cells. (2) The migratory potential and survival of neutrophil progenitors is altered in vitro. (3) Expression of the Gr-1 marker is delayed and repressed. In contrast, neutrophil function appears normal. These data demonstrate that Ikaros regulates early neutrophil differentiation but is dispensable in mature neutrophils.
Collapse
Affiliation(s)
- Alexis Dumortier
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Centre National pour la Recherche Scientifique (CNRS), Université Louis Pasteur (ULP), Illkirch Cedex, France
| | | | | | | |
Collapse
|
21
|
Abstract
Cell-fate decisions are controlled typically by conserved receptors that interact with co-evolved ligands. Therefore, the lineage-specific differentiation of immature CD4+ CD8+ T cells into CD4+ or CD8+ mature T cells is unusual in that it is regulated by clonally expressed, somatically generated T-cell receptors (TCRs) of unpredictable fine specificity. Yet, each mature T cell generally retains expression of the co-receptor molecule (CD4 or CD8) that has an MHC-binding property that matches that of its TCR. Two models were proposed initially to explain this remarkable outcome--'instruction' of lineage choice by initial signalling events or 'selection' after a stochastic fate decision that limits further development to cells with coordinated TCR and co-receptor specificities. Aspects of both models now appear to be correct; mistake-prone instruction of lineage choice precedes a subsequent selection step that filters out most incorrect decisions.
Collapse
Affiliation(s)
- Ronald N Germain
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892-1892, USA.
| |
Collapse
|
22
|
Yassai M, Ammon K, Goverman J, Marrack P, Naumov Y, Gorski J. A molecular marker for thymocyte-positive selection: selection of CD4 single-positive thymocytes with shorter TCRB CDR3 during T cell development. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:3801-7. [PMID: 11937532 DOI: 10.4049/jimmunol.168.8.3801] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The generation of the naive T cell repertoire is a direct result of maturation and selection events in the thymus. Although maturation events are judged predominantly on the expression of surface markers, molecular markers, more intimately involved in the selection process, can be informative. We have identified a molecular marker for selection in later stages of maturation in humans. Thymocytes are selected for the expression of TCR beta-chains with shorter CDR3 at the double-positive to single-positive (SP) transition. Here we extend these studies to the mouse and show that the selection phenotype is not related to alpha-chain pairing but is a function of the MHC haplotype. Interestingly, the selection is much more apparent in CD4 SP thymocytes than in CD8 SP cells. This is in contrast to human thymocytes, where the selection is equally apparent in both lineages. The involvement of MHC in the process argues that this is a positive selection stage. The difference in the extent of this selection between the two SP lineages may indicate a class difference in the nature of the TCR-MHC interaction, the role of coreceptors in the selection process, or both.
Collapse
MESH Headings
- Animals
- Biomarkers/analysis
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Complementarity Determining Regions/analysis
- Complementarity Determining Regions/blood
- Complementarity Determining Regions/genetics
- Gene Rearrangement, beta-Chain T-Cell Antigen Receptor
- Genes, MHC Class II/physiology
- Immunophenotyping
- Mice
- Mice, Inbred C57BL
- Mice, Inbred DBA
- Mice, Knockout
- Mice, Transgenic
- Peptide Fragments/genetics
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/biosynthesis
- Receptors, Antigen, T-Cell, alpha-beta/blood
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- T-Lymphocyte Subsets/cytology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
Collapse
Affiliation(s)
- Maryam Yassai
- Blood Research Institute, Blood Center of Southeastern Wisconsin, Milwaukee, WI 53201, USA
| | | | | | | | | | | |
Collapse
|
23
|
Abstract
Cells of the central nervous system (CNS) and immune system communicate regularly. There is a constant surveillance of the intact, healthy CNS by activated T-cells, and massive infiltration of the CNS by immune cells under pathological conditions such as neurodegeneration or neuroinflammation. Labeling CNS-infiltrating T-cells is an essential tool to identify the signals and mechanisms, which mediate the interaction between immune cells and cells of the CNS. In this article, we will present an overview describing currently used cellular markers and demonstrate how these markers have contributed to our current knowledge of CNS inflammation and immune surveillance.
Collapse
Affiliation(s)
- A Flügel
- Max-Planck-Institute of Neurobiology, Munich, Germany.
| | | |
Collapse
|
24
|
Wang Q, Malherbe L, Zhang D, Zingler K, Glaichenhaus N, Killeen N. CD4 promotes breadth in the TCR repertoire. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:4311-20. [PMID: 11591754 DOI: 10.4049/jimmunol.167.8.4311] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
A diverse population of MHC class II-restricted CD4 lineage T cells develops in mice that lack expression of the CD4 molecule. In this study, we show that the TCR repertoire selected in the absence of CD4 is distinct, but still overlapping in its properties with that selected in the presence of CD4. Immunization of mice lacking CD4 caused the clonal expansion of T cells that showed less breadth in the range of Ag-binding properties exhibited by their TCRs. Specifically, the CD4-deficient Ag-specific TCR repertoire was depleted of TCRs that demonstrated low-affinity binding to their ligands. The data thus suggest a key role for CD4 in broadening the TCR repertoire by potentiating productive TCR signaling and clonal expansion in response to the engagement of low-affinity antigenic ligands.
Collapse
Affiliation(s)
- Q Wang
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143, USA
| | | | | | | | | | | |
Collapse
|
25
|
Abstract
As a new slant on T lymphocyte repertoire selection, we have examined batteries of TCR sequences in thymi from transgenic mice engineered to exhibit limited, focussed TCR diversity. We have tracked the fate of differentiating thymocytes expressing a set of particular TCR through the positive selection process. Subtly different TCR sequences can promote different maturation pathways and commitment choices. Two distinct routes are followed by CD8-lineage cells interacting with MHC class I molecules, via TCR(hi) CD4(+)CD8(+) or CD4(+)CD8(int) intermediates, while CD4-lineage cells mature exclusively via a CD4(+)CD8(int) stage. The CD8-lineage routes are partially exclusive, indicating that the latter cell type is not always preceded by the former. The distribution of sequences also indicates that CD4 / CD8-lineage commitment is not strictly correlated with the class of MHC molecule engaged, and that some mechanism prevents mismatched intermediates from achieving full maturity.
Collapse
Affiliation(s)
- M Correia-Neves
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS / INSERM / ULP), Illkirch, C.U. de Strasbourg, France
| | | | | |
Collapse
|
26
|
Strong J, Wang Q, Killeen N. Impaired survival of T helper cells in the absence of CD4. Proc Natl Acad Sci U S A 2001; 98:2566-71. [PMID: 11226279 PMCID: PMC30178 DOI: 10.1073/pnas.051329698] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Signal transduction in response to ligand recognition by T cell receptors regulates T cell fate within and beyond the thymus. Herein we examine the involvement of the CD4 molecule in the regulation of T helper cell survival. T helper cells that lack CD4 expression are prone to apoptosis and show diminished survival after adoptive transfer to irradiated recipients. The helper lineage in CD4(-/-) animals shows a higher than normal apparent rate of cell division and is also enriched for cells exhibiting a memory cell phenotype. Thus the data point to a necessary role for CD4 in the regulation of T helper cell survival and homeostasis.
Collapse
Affiliation(s)
- J Strong
- Department of Microbiology and Immunology, University of California, San Francisco, CA 94143-0414, USA
| | | | | |
Collapse
|
27
|
Nepomnaschy I, Lombardi G, Bekinschtein P, Berguer P, Francisco V, de Almeida J, Buggiano V, Pasqualini CD, Piazzon I. Alterations during positive selection in the thymus of nackt CD4-deficient mice. Scand J Immunol 2000; 52:555-62. [PMID: 11119260 DOI: 10.1046/j.1365-3083.2000.00818.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The T-cell repertoire is shaped by the positive and negative selection of immature CD4(+) CD8(+) double positive (DP) thymocytes. Positive selection of DP T cells to the CD4(+) CD8(-) and CD4(-) CD8(+) simple positive (SP) lineages is a multistep process which involves cellular interactions between thymocytes and stromal cells. Mutant nackt (nkt/nkt) mice have been shown to have a deficiency in the CD4(+) CD8(-) T-cell subset both in the thymus and in the periphery. The present report suggests that nkt/nkt mice present alterations in early steps of positive selection because they show decreases in the percentages of CD69(+) and CD5(+) cells within the DP subset. Experiments involving bone marrow transfer and thymic chimeras demonstrate that the thymic epithelium of nkt/nkt mice is involved in the alterations registered during positive selection and dictates the ultimate fate of CD4(+) SP cells.
Collapse
Affiliation(s)
- I Nepomnaschy
- ILEX-CONICET, División Medicina Experimental, Instituto de Investigaciones Hematológicas, Academia Nacional de Medicina, Buenos Aires, Argentina.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gao JX, Zhang J, Awaraji C, Bhatia M, Jevnikar A, Singh B, Bell D, Delovitch TL. Preferential proliferation and differentiation of double-positive thymocytes into CD8(+) single-positive thymocytes in a novel cell culture medium. Cell Immunol 2000; 202:41-53. [PMID: 10873305 DOI: 10.1006/cimm.2000.1656] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The identification of factors that regulate the proliferation and differentiation of double-positive (DP) into CD4(+) and CD8(+) single-positive (SP) thymocytes has proven difficult due to the inability of DP thymocytes to proliferate, expand, and differentiate into SP thymocytes in available cell culture media. Here we report on the ability of DP thymocytes to differentiate in a novel conditioned medium, termed XLCM, derived from the supernatant of mitogen activated human cord blood mononuclear cells. During a 5-day culture in XLCM in the absence of thymic stromal cells, DP thymocytes from normal mice and MHC double knockout mice (lack SP thymocytes) proliferate, expand, and differentiate into several (alphabetaTCR(+), NK1.1(+)alphabetaTCR(+), and gammadeltaTCR(+)) subsets of CD4(+) and predominantly CD8(+) SP thymocytes. These studies suggest that the use of XLCM may aid in the characterization of factors that regulate the differentiation of DP thymocytes into CD8(+) SP thymocytes.
Collapse
Affiliation(s)
- J X Gao
- Autoimmunity/Diabetes Group, The University of Western Ontario, London, Ontario, N6G 2V4, Canada
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Peña-Rossi C, Zuckerman LA, Strong J, Kwan J, Ferris W, Chan S, Tarakhovsky A, Beyers AD, Killeen N. Negative Regulation of CD4 Lineage Development and Responses by CD5. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.12.6494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
CD5 deficiency results in a hyper-responsive phenotype to Ag receptor stimulation. Here we show that the development and responses of CD4 lineage T cells are regulated by the function of CD5. Thymocytes expressing the I-Ad-restricted DO11.10 TCR undergo abnormal selection without CD5. In H-2d mice, the absence of CD5 causes deletion of double-positive thymocytes, but allows for efficient selection of cells expressing high levels of the DO11.10 clonotype. By contrast, there is enhanced negative selection against the DO11.10 clonotype in the presence of I-Ab. T cell hybridomas and DO11.10 T cells are more responsive to TCR stimulation in the absence of CD5. Such hypersensitivity can be eliminated by expression of wild-type CD5, but not by a form of CD5 that lacks the cytoplasmic tail. Finally, CD5 deficiency partially suppresses the block of CD4 lineage development in CD4-deficient mice. Taken together, the data support a general role for CD5 as a negative regulator of Ag receptor signaling in the development and immune responses of CD4 lineage T cells.
Collapse
Affiliation(s)
- Claudia Peña-Rossi
- *Department of Microbiology and Immunology, University of California, San Francisco, CA 94143
| | - Linda A. Zuckerman
- *Department of Microbiology and Immunology, University of California, San Francisco, CA 94143
| | - Julie Strong
- *Department of Microbiology and Immunology, University of California, San Francisco, CA 94143
| | - Joanne Kwan
- *Department of Microbiology and Immunology, University of California, San Francisco, CA 94143
| | - William Ferris
- †Department of Medical Biochemistry and Medical Research Council Center for Molecular and Cellular Biology, University of Stellenbosch, Tygerberg, South Africa
| | - Susan Chan
- ‡Institut de Génétique et de Biologie Moléculaire et Cellulaire, Strasbourg, Illkirch, France; and
| | | | - Albert D. Beyers
- †Department of Medical Biochemistry and Medical Research Council Center for Molecular and Cellular Biology, University of Stellenbosch, Tygerberg, South Africa
| | - Nigel Killeen
- *Department of Microbiology and Immunology, University of California, San Francisco, CA 94143
| |
Collapse
|
30
|
Salmon P, Mong M, Kang XJ, Cado D, Robey E. The Role of CD8α′ in the CD4 Versus CD8 Lineage Choice. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.10.5312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
During thymic development the recognition of MHC proteins by developing thymocytes influences their lineage commitment, such that recognition of class I MHC leads to CD8 T cell development, whereas recognition of class II MHC leads to CD4 T cell development. The coreceptors CD8 and CD4 may contribute to these different outcomes through interactions with class I and class II MHC, respectively, and through interactions with the tyrosine kinase p56lck (Lck) via their cytoplasmic domains. In this paper we provide evidence that an alternatively spliced form of CD8 that cannot interact with Lck (CD8α′) can influence the CD4 vs CD8 lineage decision. Constitutive expression of a CD8 minigene transgene that encodes both CD8α and CD8α′ restores CD8 T cell development in CD8α mutant mice, but fails to permit the development of mismatched CD4 T cells bearing class I-specific TCRs. These results indicate that CD8α′ favors the development of CD8-lineage T cells, perhaps by reducing Lck activity upon class I MHC recognition in the thymus.
Collapse
Affiliation(s)
- Patrick Salmon
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Mimi Mong
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Xiao-Jun Kang
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Dragana Cado
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| | - Ellen Robey
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720
| |
Collapse
|
31
|
Marx A, Müller-Hermelink HK. From basic immunobiology to the upcoming WHO-classification of tumors of the thymus. The Second Conference on Biological and Clinical Aspects of Thymic Epithelial Tumors and related recent developments. Pathol Res Pract 1999; 195:515-33. [PMID: 10483582 DOI: 10.1016/s0344-0338(99)80001-6] [Citation(s) in RCA: 45] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The Second Conference on Biological and Clinical Aspects of Thymic Epithelial Tumors in Leiden, The Netherlands, 1998, set the stage for an interdisciplinary meeting of immunologists, pathologists and members of various clinical disciplines to exchange their recent findings in the field of thymus-related biology, pathology, and medicine. The contributions covered such diverse subjects as the role of transcription factors and cytokines in the development of the thymic microenvironment, thymic T, B and NK cell development, the pathogenesis of myasthenia gravis and other thymoma-associated autoimmunities, the pathology of thymic epithelial tumors and germ cell neoplasms, and new approaches to their diagnosis and treatment. This editorial will briefly sum up the data presented at the Conference and will comment on related novel findings that have been reported since then. Because it was also at the Leiden Conference, that the proposal of the WHO committee for the classification of thymic tumors was discussed for the first time, a description of the upcoming WHO Classification of Tumors of the Thymus is given with emphasis on the diagnostic criteria of thymic epithelial tumors, that should now be termed as type A, AB, B1-3 and type C thymomas, to make pathological and clinical studies comparable in the future.
Collapse
|
32
|
Abstract
Combining CD5-null, MHC-deficient and lineage-specific reporter animals, we have investigated the influence of CD5 on positive selection and the choice of CD4- versus CD8-lineage commitment on broad populations of thymocytes. CD5 has no obvious quantitative effect in wild-type mice. In mice lacking MHC class II molecules, however, increased numbers of transitional, class I-selected CD4+ CD8(int) CD3(hi) cells were positively selected in the absence of CD5. Importantly, they were committed to the CD4 lineage. Our results indicate that CD5 negatively regulates the differentiation of CD4-committed cells in suboptimal conditions, thus perhaps serving to tighten the correlation between restriction of the TCR and lineage choice.
Collapse
Affiliation(s)
- S Chan
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (CNRS/INSERM/ULP) Illkirch, C.U. de Strasbourg, France
| | | | | | | | | |
Collapse
|
33
|
Wack A, Corbella P, Harker N, Roderick K, Norton T, Williams K, Williams O, Kioussis D. Th Cells and Th2 Responses Can Develop in the Absence of MHC Class II-CD4 Interactions. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.3.1162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Abstract
In this paper, we address the question whether CD4 and MHC class II expression are necessary for the development of the T helper lineage during thymocyte maturation and for activation-induced Th2 responses. To bypass the CD4-MHC class II interaction requirements for positive selection and activation, we used mice that are doubly transgenic for CD8 and for the MHC class I-restricted TCR F5. This transgene combination leads to MHC class I-dependent maturation of CD4 lineage cells. Upon activation, these CD4 lineage T cells secrete IL-4 and give help to B cells but show no cytotoxic activity. Remarkably, neither MHC class II nor CD4 expression are necessary for the generation and helper functions of these cells. This suggests that under normal conditions, coreceptor-MHC interactions are necessary to ensure the canonical combinations of coreceptor and function in developing thymocytes, but that they do not determine functional commitment. Our results also imply that expression of the CD4 gene does not influence, but is merely associated with the decision to establish the T helper program. In addition, we show that activation through TCR-MHC class I interactions can induce Th2 responses independently of CD4 and MHC class II expression.
Collapse
Affiliation(s)
- Andreas Wack
- Division of Molecular Immunology, National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Paola Corbella
- Division of Molecular Immunology, National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Nicola Harker
- Division of Molecular Immunology, National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Kathleen Roderick
- Division of Molecular Immunology, National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Trisha Norton
- Division of Molecular Immunology, National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Keith Williams
- Division of Molecular Immunology, National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Owen Williams
- Division of Molecular Immunology, National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Dimitris Kioussis
- Division of Molecular Immunology, National Institute for Medical Research, Mill Hill, London, United Kingdom
| |
Collapse
|
34
|
Abstract
Differentiation of T cells is a complicated affair and there has been a dearth of markers that faithfully reflect thymocyte phenotype. A new strategy based on T-cell receptor gene sequencing has revealed a marker that can be used to monitor thymocyte differentiation with fidelity and without perturbation.
Collapse
Affiliation(s)
- C Benoist
- Institut de Génétique et de, Biologie Moléculaire et Cellulaire, (CNRS/INSERM/ULP) BP, 163 67400, Illkirch C.U, de Strasbourg, France.
| | | |
Collapse
|