1
|
Matsumoto K, Okuyama K, Sidwell T, Yamashita M, Endo T, Satoh-Takayama N, Ohno H, Morio T, Rothenberg EV, Taniuchi I. A Bcl11b N797K variant isolated from an immunodeficient patient inhibits early thymocyte development in mice. Front Immunol 2024; 15:1363704. [PMID: 38495886 PMCID: PMC10940544 DOI: 10.3389/fimmu.2024.1363704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 02/20/2024] [Indexed: 03/19/2024] Open
Abstract
BCL11B is a transcription factor with six C2H2-type zinc-finger domains. Studies in mice have shown that Bcl11b plays essential roles in T cell development. Several germline heterozygous BCL11B variants have been identified in human patients with inborn errors of immunity (IEI) patients. Among these, two de novo mis-sense variants cause asparagine (N) to lysine (K) replacement in distinct zinc-finger domains, BCL11BN441K and BCL11BN807K. To elucidate the pathogenesis of the BCL11BN807K variant, we generated a mouse model of BCL11BN807K by inserting the corresponding mutation, Bcl11bN797K, into the mouse genome. In Bcl11b+/N797K mice, the proportion of immature CD4-CD8+ single-positive thymocytes was increased, and the development of invariant natural killer cells was severely inhibited in a T-cell-intrinsic manner. Under competitive conditions, γδT cell development was outcompeted by control cells. Bcl11bN797K/N797K mice died within one day of birth. Recipient mice reconstituted with Bcl11bN797K/N797K fetal liver cells nearly lacked CD4+CD8+ double-positive thymocytes, which was consistent with the lack of their emergence in culture from Bcl11bN797K/N797K fetal liver progenitors. Interestingly, Bcl11bN797K/N797K progenitors gave rise to aberrant c-Kit+ and CD44+ cells both in vivo and in vitro. The increase in the proportion of immature CD8 single-positive thymocytes in the Bcl11bN797K mutants is caused, in part, by the inefficient activation of the Cd4 gene due to the attenuated function of the two Cd4 enhancers via distinct mechanisms. Therefore, we conclude that immunodeficient patient-derived Bcl11bN797K mutant mice elucidated a novel role for Bcl11b in driving the appropriate transition of CD4-CD8- into CD4+CD8+ thymocytes.
Collapse
Affiliation(s)
- Kazuaki Matsumoto
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama Kanagawa, Japan
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kazuki Okuyama
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama Kanagawa, Japan
| | - Tom Sidwell
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Motoi Yamashita
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama Kanagawa, Japan
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takaho Endo
- Genome Platform, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama Kanagawa, Japan
| | - Naoko Satoh-Takayama
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama Kanagawa, Japan
| | - Hiroshi Ohno
- Laboratory for Intestinal Ecosystem, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama Kanagawa, Japan
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Ellen V. Rothenberg
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, United States
| | - Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama Kanagawa, Japan
| |
Collapse
|
2
|
Rahmberg AR, Markowitz TE, Mudd JC, Hirsch V, Brenchley JM. Epigenetic Reprogramming Leads to Downregulation of CD4 and Functional Changes in African Green Monkey Memory CD4 + T Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2022; 209:337-345. [PMID: 35750337 PMCID: PMC9283288 DOI: 10.4049/jimmunol.2200109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 05/05/2022] [Indexed: 05/12/2023]
Abstract
African green monkeys (AGMs), Chlorocebus pygerythrus, are a natural host for a lentivirus related to HIV, SIV. SIV-infected AGMs rarely progress to AIDS despite robust viral replication. Though multiple mechanisms are involved, a primary component is the animals' ability to downregulate CD4 expression on mature CD4+ Th cells, rendering these cells resistant to infection by SIV. These CD8αα+ T cells retain functional characteristics of CD4+ Th cells while simultaneously acquiring abilities of cytotoxic CD8αβ+ T cells. To determine mechanisms underlying functional differences between T cell subsets in AGMs, chromatin accessibility in purified populations was determined by assay for transposase-accessible chromatin sequencing. Differences in chromatin accessibility alone were sufficient to cluster cells by subtype, and accessibility at the CD4 locus reflected changes in CD4 expression. DNA methylation at the CD4 locus also correlated with inaccessible chromatin. By associating accessible regions with nearby genes, gene expression was found to correlate with accessibility changes. T cell and immune system activation pathways were identified when comparing regions that changed accessibility from CD4+ T cells to CD8αα+ T cells. Different transcription factor binding sites are revealed as chromatin accessibility changes, and these differences may elicit downstream changes in differentiation. This comprehensive description of the epigenetic landscape of AGM T cells identified genes and pathways that could have translational value in therapeutic approaches recapitulating the protective effects CD4 downregulation.
Collapse
Affiliation(s)
- Andrew R Rahmberg
- Barrier Immunity Section, Lab of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Tovah E Markowitz
- NIAID Collaborative Bioinformatics Resource, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
- Axle Informatics, Bethesda, MD
| | - Joseph C Mudd
- Tulane National Primate Research Center, Division of Immunology, Tulane University, New Orleans, LA; and
| | - Vanessa Hirsch
- Nonhuman Primate Virology Section, Lab of Molecular Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Jason M Brenchley
- Barrier Immunity Section, Lab of Viral Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD;
| |
Collapse
|
3
|
Frias AB, Boi SK, Lan X, Youngblood B. Epigenetic regulation of T cell adaptive immunity. Immunol Rev 2021; 300:9-21. [PMID: 33644866 DOI: 10.1111/imr.12943] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 12/24/2022]
Abstract
The conceptualization of adaptive immunity, founded on the observation of immunological memory, has served as the basis for modern vaccination and immunotherapy approaches. This fundamental concept has allowed immunologists to explore mechanisms that enable humoral and cellular lymphocytes to tailor immune response functions to a wide array of environmental insults and remain poised for future pathogenic encounters. Until recently, for T cells it has remained unclear how memory differentiation acquires and sustains a gene expression program that grants a cell with a capacity for a heightened recall response. Recent investigations into this critical question have identified epigenetic programs as a causal molecular mechanism governing T cell subset specification and immunological memory. Here, we outline the studies that have illustrated this concept and posit on how insights into T cell adaptive immunity can be applied to improve upon existing immunotherapies.
Collapse
Affiliation(s)
- Adolfo B Frias
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shannon K Boi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xin Lan
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.,College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ben Youngblood
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
4
|
Mudd JC, Lai S, Shah S, Rahmberg A, Flynn JK, Starke CE, Perkins MR, Ransier A, Darko S, Douek DC, Hirsch VM, Cameron M, Brenchley JM. Epigenetic silencing of CD4 expression in nonpathogenic SIV infection in African green monkeys. JCI Insight 2020; 5:139043. [PMID: 32841214 PMCID: PMC7526541 DOI: 10.1172/jci.insight.139043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 08/13/2020] [Indexed: 01/19/2023] Open
Abstract
African green monkeys (AGMs) are natural hosts of SIV that postthymically downregulate CD4 to maintain a large population of CD4-CD8aa+ virus-resistant cells with Th functionality, which can result in AGMs becoming apparently cured of SIVagm infection. To understand the mechanisms of this process, we performed genome-wide transcriptional analysis on T cells induced to downregulate CD4 in vitro from AGMs and closely related patas monkeys and T cells that maintain CD4 expression from rhesus macaques. In T cells that downregulated CD4, pathway analysis revealed an atypical regulation of the DNA methylation machinery, which was reversible when pharmacologically targeted with 5-aza-2 deoxycytidine. This signature was driven largely by the dioxygenase TET3, which became downregulated with loss of CD4 expression. CpG motifs within the AGM CD4 promoter region became methylated during CD4 downregulation in vitro and were stably imprinted in AGM CD4-CD8aa+ T cells sorted directly ex vivo. These results suggest that AGMs use epigenetic mechanisms to durably silence the CD4 gene. Manipulation of these mechanisms could provide avenues for modulating SIV and HIV-1 entry receptor expression in hosts that become progressively infected with SIV, which could lead to novel therapeutic interventions aimed to reduce HIV viremia in vivo.
Collapse
Affiliation(s)
- Joseph C Mudd
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research
| | - Stephen Lai
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research
| | - Sanjana Shah
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research
| | - Andrew Rahmberg
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research
| | - Jacob K Flynn
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research
| | - Carly E Starke
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research
| | - Molly R Perkins
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research
| | - Amy Ransier
- Human Immunology Section, Vaccine Research Center, and
| | - Sam Darko
- Human Immunology Section, Vaccine Research Center, and
| | | | - Vanessa M Hirsch
- Nonhuman Primate Virology Section, Laboratory of Molecular Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Maryland, USA
| | - Mark Cameron
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, Ohio, USA
| | - Jason M Brenchley
- Barrier Immunity Section, Laboratory of Viral Diseases, Division of Intramural Research
| |
Collapse
|
5
|
Hosoya T, D'Oliveira Albanus R, Hensley J, Myers G, Kyono Y, Kitzman J, Parker SCJ, Engel JD. Global dynamics of stage-specific transcription factor binding during thymocyte development. Sci Rep 2018; 8:5605. [PMID: 29618724 PMCID: PMC5884796 DOI: 10.1038/s41598-018-23774-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Accepted: 03/20/2018] [Indexed: 12/29/2022] Open
Abstract
In vertebrates, multiple transcription factors (TFs) bind to gene regulatory elements (promoters, enhancers, and silencers) to execute developmental expression changes. ChIP experiments are often used to identify where TFs bind to regulatory elements in the genome, but the requirement of TF-specific antibodies hampers analyses of tens of TFs at multiple loci. Here we tested whether TF binding predictions using ATAC-seq can be used to infer the identity of TFs that bind to functionally validated enhancers of the Cd4, Cd8, and Gata3 genes in thymocytes. We performed ATAC-seq at four distinct stages of development in mouse thymus, probing the chromatin accessibility landscape in double negative (DN), double positive (DP), CD4 single positive (SP4) and CD8 SP (SP8) thymocytes. Integration of chromatin accessibility with TF motifs genome-wide allowed us to infer stage-specific occupied TF binding sites within known and potentially novel regulatory elements. Our results provide genome-wide stage-specific T cell open chromatin profiles, and allow the identification of candidate TFs that drive thymocyte differentiation at each developmental stage.
Collapse
Affiliation(s)
- Tomonori Hosoya
- Department of Cell and Developmental Biology, Ann Arbor, USA
| | | | - John Hensley
- Department of Computational Medicine and Bioinformatics, Ann Arbor, USA
| | - Greggory Myers
- Department of Cell and Developmental Biology, Ann Arbor, USA
| | - Yasuhiro Kyono
- Department of Computational Medicine and Bioinformatics, Ann Arbor, USA.,Department of Human Genetics, University of Michigan, 3035 BSRB, 109 Zina Pitcher Place, Ann Arbor, Michigan, 48109-2200, USA
| | - Jacob Kitzman
- Department of Computational Medicine and Bioinformatics, Ann Arbor, USA.,Department of Human Genetics, University of Michigan, 3035 BSRB, 109 Zina Pitcher Place, Ann Arbor, Michigan, 48109-2200, USA
| | - Stephen C J Parker
- Department of Computational Medicine and Bioinformatics, Ann Arbor, USA.,Department of Human Genetics, University of Michigan, 3035 BSRB, 109 Zina Pitcher Place, Ann Arbor, Michigan, 48109-2200, USA
| | | |
Collapse
|
6
|
Vacchio MS, Bosselut R. What Happens in the Thymus Does Not Stay in the Thymus: How T Cells Recycle the CD4+-CD8+ Lineage Commitment Transcriptional Circuitry To Control Their Function. THE JOURNAL OF IMMUNOLOGY 2017; 196:4848-56. [PMID: 27260768 DOI: 10.4049/jimmunol.1600415] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Accepted: 04/06/2016] [Indexed: 12/24/2022]
Abstract
MHC-restricted CD4(+) and CD8(+) T cells are at the core of most adaptive immune responses. Although these cells carry distinct functions, they arise from a common precursor during thymic differentiation, in a developmental sequence that matches CD4 and CD8 expression and functional potential with MHC restriction. Although the transcriptional control of CD4(+)-CD8(+) lineage choice in the thymus is now better understood, less was known about what maintains the CD4(+) and CD8(+) lineage integrity of mature T cells. In this review, we discuss the mechanisms that establish in the thymus, and maintain in postthymic cells, the separation of these lineages. We focus on recent studies that address the mechanisms of epigenetic control of Cd4 expression and emphasize how maintaining a transcriptional circuitry nucleated around Thpok and Runx proteins, the key architects of CD4(+)-CD8(+) lineage commitment in the thymus, is critical for CD4(+) T cell helper functions.
Collapse
Affiliation(s)
- Melanie S Vacchio
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Rémy Bosselut
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| |
Collapse
|
7
|
Issuree PDA, Ng CP, Littman DR. Heritable Gene Regulation in the CD4:CD8 T Cell Lineage Choice. Front Immunol 2017; 8:291. [PMID: 28382035 PMCID: PMC5360760 DOI: 10.3389/fimmu.2017.00291] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/28/2017] [Indexed: 12/04/2022] Open
Abstract
The adaptive immune system is dependent on functionally distinct lineages of T cell antigen receptor αβ-expressing T cells that differentiate from a common progenitor in the thymus. CD4+CD8+ progenitor thymocytes undergo selection following interaction with MHC class I and class II molecules bearing peptide self-antigens, giving rise to CD8+ cytotoxic and CD4+ helper or regulatory T cell lineages, respectively. The strict correspondence of CD4 and CD8 expression with distinct cellular phenotypes has made their genes useful surrogates for investigating molecular mechanisms of lineage commitment. Studies of Cd4 and Cd8 transcriptional regulation have uncovered cis-regulatory elements that are critical for mediating epigenetic modifications at distinct stages of development to establish heritable transcriptional programs. In this review, we examine the epigenetic mechanisms involved in Cd4 and Cd8 gene regulation during T cell lineage specification and highlight the features that make this an attractive system for uncovering molecular mechanisms of heritability.
Collapse
Affiliation(s)
- Priya D A Issuree
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine , New York, NY , USA
| | - Charles P Ng
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine , New York, NY , USA
| | - Dan R Littman
- The Kimmel Center for Biology and Medicine of the Skirball Institute, New York University School of Medicine, New York, NY, USA; Howard Hughes Medical Institute, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
8
|
Abstract
There has been speculation as to how bi-potent CD4(+) CD8(+) double-positive precursor thymocytes choose their distinct developmental fate, becoming either CD4(+) helper or CD8(+) cytotoxic T cells. Based on the clear correlation of αβT cell receptor (TCR) specificity to major histocompatibility complex (MHC) classes with this lineage choice, various studies have attempted to resolve this question by examining the cellular signaling events initiated by TCR engagements, a strategy referred to as a 'top-down' approach. On the other hand, based on the other correlation of CD4/CD8 co-receptor expression with its selected fate, other studies have addressed this question by gradually unraveling the sequential mechanisms that control the phenotypic outcome of this fate decision, a method known as the 'bottom-up' approach. Bridging these two approaches will contribute to a more comprehensive understanding of how TCR signals are coupled with developmental programs in the nucleus. Advances made during the last two decades seemed to make these two approaches more closely linked. For instance, identification of two transcription factors, ThPOK and Runx3, which play central roles in the development of helper and cytotoxic lineages, respectively, provided significant insights into the transcriptional network that controls a CD4/CD8 lineage choice. This review summarizes achievements made using the 'bottom-up' approach, followed by a perspective on future pathways toward coupling TCR signaling with nuclear programs.
Collapse
Affiliation(s)
- Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, RIKEN Center for Integrative Medical Sciences, Kanagawa, Japan
| |
Collapse
|
9
|
Westendorf K, Durek P, Ayew S, Mashreghi MF, Radbruch A. Chromosomal localisation of the CD4cre transgene in B6·Cg-Tg(Cd4-cre)1Cwi mice. J Immunol Methods 2016; 436:54-7. [PMID: 27345256 DOI: 10.1016/j.jim.2016.06.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/22/2016] [Accepted: 06/22/2016] [Indexed: 11/26/2022]
Abstract
The B6·Cg-Tg(Cd4-cre)1Cwi line expresses CRE recombinase under the control of the promoter and regulatory elements of the Cd4 gene. Here we show that CRE recombinase expression reduces the number and frequencies of CD4 positive subsets in a dose-dependent manner and localize the integration site of the transgenic construct to position 60335693-60341285 (qD) of chromosome 3. The insert contains at least 15 complete sequential copies of the transgenic construct. Based on this information we describe a novel PCR assay for genetic typing of transgenic mice.
Collapse
Affiliation(s)
- Kerstin Westendorf
- Deutsches Rheuma-Forschungszentrum Berlin, Chariteplatz 1, 10117 Berlin, Germany
| | - Pawel Durek
- Deutsches Rheuma-Forschungszentrum Berlin, Chariteplatz 1, 10117 Berlin, Germany.
| | - Samia Ayew
- Deutsches Rheuma-Forschungszentrum Berlin, Chariteplatz 1, 10117 Berlin, Germany
| | - Mir-Farzin Mashreghi
- Deutsches Rheuma-Forschungszentrum Berlin, Chariteplatz 1, 10117 Berlin, Germany.
| | - Andreas Radbruch
- Deutsches Rheuma-Forschungszentrum Berlin, Chariteplatz 1, 10117 Berlin, Germany
| |
Collapse
|
10
|
Abstract
During blood cell development, hematopoietic stem cells generate diverse mature populations via several rounds of binary fate decisions. At each bifurcation, precursors adopt one fate and inactivate the alternative fate either stochastically or in response to extrinsic stimuli and stably maintain the selected fates. Studying of these processes would contribute to better understanding of etiology of immunodeficiency and leukemia, which are caused by abnormal gene regulation during the development of hematopoietic cells. The CD4(+) helper versus CD8(+) cytotoxic T-cell fate decision serves as an excellent model to study binary fate decision processes. These two cell types are derived from common precursors in the thymus. Positive selection of their TCRs by self-peptide presented on either MHC class I or class II triggers their fate decisions along with mutually exclusive retention and silencing of two coreceptors, CD4 and CD8. In the past few decades, extensive effort has been made to understand the T-cell fate decision processes by studying regulation of genes encoding the coreceptors and selection processes. These studies have identified several key transcription factors and gene regulatory networks. In this chapter, I will discuss recent advances in our understanding of the binary cell fate decision processes of T cells.
Collapse
Affiliation(s)
- Takeshi Egawa
- Department of Pathology and Immunology, School of Medicine, Washington University in St. Louis, St. Louis, Missouri, USA.
| |
Collapse
|
11
|
Rapid and unambiguous detection of DNase I hypersensitive site in rare population of cells. PLoS One 2014; 9:e85740. [PMID: 24465674 PMCID: PMC3897510 DOI: 10.1371/journal.pone.0085740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2013] [Accepted: 12/02/2013] [Indexed: 11/19/2022] Open
Abstract
DNase I hypersensitive (DHS) sites are important for understanding cis regulation of gene expression. However, existing methods for detecting DHS sites in small numbers of cells can lead to ambiguous results. Here we describe a simple new method, in which DNA fragments with ends generated by DNase I digestion are isolated and used as templates for two PCR reactions. In the first PCR, primers are derived from sequences up- and down-stream of the DHS site. If the DHS site exists in the cells, the first PCR will not produce PCR products due to the cuts of the templates by DNase I between the primer sequences. In the second PCR, one primer is derived from sequence outside the DHS site and the other from the adaptor. This will produce a smear of PCR products of different sizes due to cuts by DNase I at different positions at the DHS site. With this design, we detected a DHS site at the CD4 gene in two CD4 T cell populations using as few as 2×10(4) cells. We further validated this method by detecting a DHS site of the IL-4 gene that is specifically present in type 2 but not type 1 T helper cells. Overall, this method overcomes the interference by genomic DNA not cut by DNase I at the DHS site, thereby offering unambiguous detection of DHS sites in the cells.
Collapse
|
12
|
Transcriptional control of CD4 and CD8 coreceptor expression during T cell development. Cell Mol Life Sci 2013; 70:4537-53. [PMID: 23793512 PMCID: PMC3827898 DOI: 10.1007/s00018-013-1393-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2013] [Revised: 05/27/2013] [Accepted: 05/29/2013] [Indexed: 11/24/2022]
Abstract
The differentiation and function of peripheral helper and cytotoxic T cell lineages is coupled with the expression of CD4 and CD8 coreceptor molecules, respectively. This indicates that the control of coreceptor gene expression is closely linked with the regulation of CD4/CD8 lineage decision of DP thymocytes. Research performed during the last two decades revealed comprehensive mechanistic insight into the developmental stage- and subset/lineage-specific regulation of Cd4, Cd8a and Cd8b1 (Cd8) gene expression. These studies provided important insight into transcriptional control mechanisms during T cell development and into the regulation of cis-regulatory networks in general. Moreover, the identification of transcription factors involved in the regulation of CD4 and CD8 significantly advanced the knowledge of the transcription factor network regulating CD4/CD8 cell-fate choice of DP thymocytes. In this review, we provide an overview of the identification and characterization of CD4/CD8 cis-regulatory elements and present recent progress in our understanding of how these cis-regulatory elements control CD4/CD8 expression during T cell development and in peripheral T cells. In addition, we describe the transcription factors implicated in the regulation of coreceptor gene expression and discuss how these factors are integrated into the transcription factor network that regulates CD4/CD8 cell-fate choice of DP thymocytes.
Collapse
|
13
|
Koch F, Fenouil R, Gut M, Cauchy P, Albert TK, Zacarias-Cabeza J, Spicuglia S, de la Chapelle AL, Heidemann M, Hintermair C, Eick D, Gut I, Ferrier P, Andrau JC. Transcription initiation platforms and GTF recruitment at tissue-specific enhancers and promoters. Nat Struct Mol Biol 2011; 18:956-63. [PMID: 21765417 DOI: 10.1038/nsmb.2085] [Citation(s) in RCA: 254] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2010] [Accepted: 05/12/2011] [Indexed: 11/09/2022]
Abstract
Recent work has shown that RNA polymerase (Pol) II can be recruited to and transcribe distal regulatory regions. Here we analyzed transcription initiation and elongation through genome-wide localization of Pol II, general transcription factors (GTFs) and active chromatin in developing T cells. We show that Pol II and GTFs are recruited to known T cell-specific enhancers. We extend this observation to many new putative enhancers, a majority of which can be transcribed with or without polyadenylation. Importantly, we also identify genomic features called transcriptional initiation platforms (TIPs) that are characterized by large areas of Pol II and GTF recruitment at promoters, intergenic and intragenic regions. TIPs show variable widths (0.4-10 kb) and correlate with high CpG content and increased tissue specificity at promoters. Finally, we also report differential recruitment of TFIID and other GTFs at promoters and enhancers. Overall, we propose that TIPs represent important new regulatory hallmarks of the genome.
Collapse
Affiliation(s)
- Frederic Koch
- Centre d'Immunologie de Marseille-Luminy, Université Aix-Marseille, Campus de Luminy, Marseille, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
The epigenetic landscape of lineage choice: lessons from the heritability of CD4 and CD8 expression. Curr Top Microbiol Immunol 2011; 356:165-88. [PMID: 21989924 PMCID: PMC4417357 DOI: 10.1007/82_2011_175] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Developing αβ T cells choose between the helper and cytotoxic lineages, depending upon the specificity of their T cell receptors for MHC molecules. The expression of the CD4 co-receptor on helper cells and the CD8 co-receptor on cytotoxic cells is intimately linked to this decision, and their regulation at the transcriptional level has been the subject of intense study to better understand lineage choice. Indeed, as the fate of developing T cells is decided, the expression status of these genes is accordingly locked. Genetic models have revealed important transcriptional elements and the ability to manipulate these elements in the framework of development has added a new perspective on the temporal nature of their function and the epigenetic maintenance of gene expression. We examine here novel insights into epigenetic mechanisms that have arisen through the study of these genes.
Collapse
|
15
|
Abstract
The helper versus cytotoxic-lineage choice of CD4(+)CD8(+) DP thymocytes correlates with MHC restriction of their T cell receptors and the termination of either CD8 or CD4 coreceptor expression. It has been hypothesized that transcription factors regulating the expression of the Cd4/Cd8 coreceptor genes must play a role in regulating the lineage decision of DP thymocytes. Indeed, progress made during the past decade led to the identification of several transcription factors that regulate CD4/CD8 expression that are as well important regulators of helper/cytotoxic cell fate choice. These studies provided insight into the molecular link between the regulation of coreceptor expression and lineage decision. However, studies initiated by the identification of ThPOK, a central transcription factor for helper T cell development, have offered another perspective on the cross-regulation between these two processes. Here, we review advances in our understanding of regulatory circuits composed of transcription factors and their link to epigenetic mechanisms, which play essential roles in specifying and sealing cell lineage identity during the CD4/CD8 commitment process of DP thymocytes.
Collapse
Affiliation(s)
- Ichiro Taniuchi
- Laboratory for Transcriptional Regulation, Research Center for Allergy and Immunology, RIKEN, Suehiro-cho, Turumi-ku, Yokohama, Kanagawa, Japan
| | | |
Collapse
|
16
|
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
|
17
|
Kok K, Nock GE, Verrall EAG, Mitchell MP, Hommes DW, Peppelenbosch MP, Vanhaesebroeck B. Regulation of p110delta PI 3-kinase gene expression. PLoS One 2009; 4:e5145. [PMID: 19357769 PMCID: PMC2663053 DOI: 10.1371/journal.pone.0005145] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2008] [Accepted: 02/19/2009] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Despite an intense interest in the biological functions of the phosphoinositide 3-kinase (PI3K) signalling enzymes, little is known about the regulation of PI3K gene expression. This also applies to the leukocyte-enriched p110delta catalytic subunit of PI3K, an enzyme that has attracted widespread interest because of its role in immunity and allergy. PRINCIPAL FINDINGS We show that p110delta expression is mainly regulated at the transcriptional level. In fibroblasts, lymphocytes and myeloid cells, p110delta gene transcription appears to be constitutive and not subject to acute stimulation. 5'RACE experiments revealed that p110delta mRNA transcripts contain distinct upstream untranslated exons (named exon -1, -2a, -2b, -2c and -2d), which are located up to 81 kb upstream of the translational start codon in exon 1. The levels of all the different p110delta transcripts are higher in leukocytes compared to non-leukocytes, with the p110delta transcript containing exon -2a most abundantly expressed. We have identified a highly conserved transcription factor (TF) binding cluster in the p110delta gene which has enhanced promoter activity in leukocytes compared to non-leukocytes. In human, this TF cluster is located immediately upstream of exon -2a whilst in mouse, it is located within exon -2a. CONCLUSION This study identifies a conserved PIK3CD promoter region that may account for the predominant leukocyte expression of p110delta.
Collapse
Affiliation(s)
- Klaartje Kok
- Centre for Cell Signalling, Institute of Cancer, Queen Mary University of London, Charterhouse Square, London, United Kingdom
- Department of Cell Biology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Gemma E. Nock
- Centre for Cell Signalling, Institute of Cancer, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Elizabeth A. G. Verrall
- Centre for Cell Signalling, Institute of Cancer, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Michael P. Mitchell
- Bioinformatics and Biostatistics, Cancer Research UK London Research Institute, London, United Kingdom
| | - Daan W. Hommes
- Department of Gastroenterology and Hepatology; Leiden University Medical Centre, Leiden, The Netherlands
| | - Maikel P. Peppelenbosch
- Department of Cell Biology, University Medical Centre Groningen, University of Groningen, Groningen, The Netherlands
| | - Bart Vanhaesebroeck
- Centre for Cell Signalling, Institute of Cancer, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| |
Collapse
|
18
|
Yang Y, Wang HC, Sun XH. Id1 induces apoptosis through inhibition of RORgammat expression. BMC Immunol 2008; 9:20. [PMID: 18489764 PMCID: PMC2408562 DOI: 10.1186/1471-2172-9-20] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Accepted: 05/19/2008] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Basic helix-loop-helix E proteins are transcription factors that play crucial roles in T cell development by controlling thymocyte proliferation, differentiation and survival. E protein functions can be repressed by their naturally occurring inhibitors, Id proteins (Id1-4). Transgenic expression of Id1 blocks T cell development and causes massive apoptosis of developing thymocytes. However, the underlying mechanisms are not entirely understood due to relatively little knowledge of the target genes regulated by E proteins. RESULTS We designed a unique strategy to search for genes directly controlled by E proteins and found RORgammat to be a top candidate. Using microarray analyses and reverse-transcriptase PCR assays, we showed that Id1 expression diminished RORgammat mRNA levels in T cell lines and primary thymocytes while induction of E protein activity restored RORgammat expression. E proteins were found to specifically bind to the promoter region of RORgammat, suggesting their role in activating transcription of the gene. Functional significance of E protein-controlled RORgammat expression was established based on the finding that RORgammat rescued apoptosis caused by Id1 overexpression. Furthermore, expression of RORgammat prevented Id1-induced p38 MAP kinase hyper-activation. CONCLUSION These results suggest that E protein-dependent RORgammat gene expression aids the survival of developing thymocytes, which provides a possible explanation for the massive apoptosis found in Id1 transgenic mice.
Collapse
Affiliation(s)
- Yuanzheng Yang
- Immunobiology and Cancer Program, Oklahoma Medical Research Foundation, 825 NE 13th St, Oklahoma City, OK 73104, USA.
| | | | | |
Collapse
|
19
|
Abstract
CD8(+) cytotoxic and CD4(+) helper/inducer T cells develop from common thymocyte precursors that express both CD4 and CD8 molecules. Upon T cell receptor signaling, these cells initiate a differentiation program that includes complex changes in CD4 and CD8 expression, allowing identification of transitional intermediates in this developmental pathway. Little is known about regulation of these early transitions or their specific importance to CD4 and CD8 T cell development. Here, we show a severe block at the CD4(lo)CD8(lo) transitional stage of positive selection caused by loss of the nuclear HMG box protein TOX. As a result, CD4 lineage T cells, including regulatory T and CD1d-dependent natural killer T cells, fail to develop. In contrast, functional CD8(+) T cells develop in TOX-deficient mice. Our data suggest that TOX-dependent transition to the CD4(+)CD8(lo) stage is required for continued development of class II major histocompatibility complex-specific T cells, regardless of ultimate lineage fate.
Collapse
Affiliation(s)
- Parinaz Aliahmad
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
20
|
Naito T, Gómez-Del Arco P, Williams CJ, Georgopoulos K. Antagonistic interactions between Ikaros and the chromatin remodeler Mi-2beta determine silencer activity and Cd4 gene expression. Immunity 2007; 27:723-34. [PMID: 17980631 DOI: 10.1016/j.immuni.2007.09.008] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2007] [Revised: 07/24/2007] [Accepted: 09/17/2007] [Indexed: 01/28/2023]
Abstract
Lineage commitment is induced by changes in gene expression dictated by the intimate interaction between transcription factors and chromatin regulators. Here, we revealed the antagonistic interplay between Ikaros and its associate the chromatin remodeler Mi-2beta during T cell development, as exemplified by the regulation of Cd4 expression. Loss of Ikaros or Mi-2beta led to activation or repression, respectively, of the Cd4 locus at inappropriate stages of development. Their combined mutation reverted to normal CD4 expression. In double-negative thymocytes, Ikaros binding to the Cd4 silencer contributed to its repressive activity. In double-positive thymocytes, concomitant binding of Mi-2beta with Ikaros to the Cd4 silencer caused silencer inactivation, thereby allowing for CD4 expression. Mi-2beta facilitated recruitment of histone acetyl transferases to the silencer. This recruitment possibly antagonized Ikaros and associated repressive activities. Thus, concomitant interactions between functionally opposing chromatin-regulating machineries are an important mode of gene regulation during lineage determination.
Collapse
Affiliation(s)
- Taku Naito
- Cutaneous Biology Research Center, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | | | | | | |
Collapse
|
21
|
Kolly C, Zakher A, Strauss C, Suter MM, Müller EJ. Keratinocyte transcriptional regulation of the human c-Myc promoter occurs via a novel Lef/Tcf binding element distinct from neoplastic cells. FEBS Lett 2007; 581:1969-76. [PMID: 17466981 DOI: 10.1016/j.febslet.2007.04.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2007] [Revised: 04/07/2007] [Accepted: 04/10/2007] [Indexed: 11/24/2022]
Abstract
The proto-oncogene c-Myc is involved in early neoplastic transformations. Two consensus Lef/Tcf binding elements (TBE) were found to be prerequisite for transcriptional transactivation by the armadillo proteins beta-catenin and plakoglobin (PG) together with Tcf4 in human neoplastic cells. In epidermal keratinocytes, c-Myc was reported to be repressed by Lef-1 and PG. Using reporter gene assays, here we demonstrate that deletion of the two consensus TBE fails to abrogate transcriptional regulation by Lef-1/PG in wildtype and beta-catenin-/- keratinocytes, while it reduces transcription in pre-neoplastic PG-/- keratinocytes. We identified a TBE sequence variant downstream of the major transcriptional initiation site that binds Lef-1 in vitro and in vivo, and its mutation compromised transcriptional regulation by Lef-1/PG. Collectively, this study demonstrates that the two consensus TBE's reported in neoplastic cells are dispensable for c-Myc regulation in normal keratinocytes, which instead use a novel TBE sequence variant. This unprecedented finding may have important implications for armadillo target genes involved in carcinogenesis.
Collapse
Affiliation(s)
- Carine Kolly
- Molecular Dermatology, Institute of Animal Pathology, Vetsuisse Faculty, 3012 Bern, Switzerland
| | | | | | | | | |
Collapse
|
22
|
Huang Z, Xie H, Ioannidis V, Held W, Clevers H, Sadim MS, Sun Z. Transcriptional regulation of CD4 gene expression by T cell factor-1/beta-catenin pathway. THE JOURNAL OF IMMUNOLOGY 2006; 176:4880-7. [PMID: 16585583 DOI: 10.4049/jimmunol.176.8.4880] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
By interacting with MHC class II molecules, CD4 facilitates lineage development as well as activation of Th cells. Expression of physiological levels of CD4 requires a proximal CD4 enhancer to stimulate basic CD4 promoter activity. T cell factor (TCF)-1/beta-catenin pathway has previously been shown to regulate thymocyte survival via up-regulating antiapoptotic molecule Bcl-xL. By both loss and gain of function studies, in this study we show additional function of TCF-1/beta-catenin pathway in the regulation of CD4 expression in vivo. Mice deficient in TCF-1 displayed significantly reduced protein and mRNA levels of CD4 in CD4+ CD8+ double-positive (DP) thymocytes. A transgene encoding Bcl-2 restored survival but not CD4 levels of TCF-1(-/-) DP cells. Thus, TCF-1-regulated survival and CD4 expression are two separate events. In contrast, CD4 levels were restored on DP TCF-1(-/-) cells by transgenic expression of a wild-type TCF-1, but not a truncated TCF-1 that lacks a domain required for interacting with beta-catenin. Furthermore, forced expression of a stabilized beta-catenin, a coactivator of TCF-1, resulted in up-regulation of CD4. TCF-1 or stabilized beta-catenin greatly stimulated activity of a CD4 reporter gene driven by a basic CD4 promoter and the CD4 enhancer. However, mutation of a potential TCF binding site located within the enhancer abrogated TCF-1 and beta-catenin-mediated activation of CD4 reporter. Finally, recruitment of TCF-1 to CD4 enhancer was detected in wild-type but not TCF-1 null mice by chromatin-immunoprecipitation analysis. Thus, our results demonstrated that TCF/beta-catenin pathway enhances CD4 expression in vivo by recruiting TCF-1 to stimulate CD4 enhancer activity.
Collapse
Affiliation(s)
- Zhaofeng Huang
- Department of Microbiology & Immunology, College of Medicine, University of Illinois, Chicago, IL 60612, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Bilic I, Koesters C, Unger B, Sekimata M, Hertweck A, Maschek R, Wilson CB, Ellmeier W. Negative regulation of CD8 expression via Cd8 enhancer-mediated recruitment of the zinc finger protein MAZR. Nat Immunol 2006; 7:392-400. [PMID: 16491076 PMCID: PMC3001192 DOI: 10.1038/ni1311] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2005] [Accepted: 01/17/2006] [Indexed: 01/22/2023]
Abstract
Coreceptor expression is tightly regulated during thymocyte development. Deletion of specific Cd8 enhancers leads to variegated expression of CD8alphabeta heterodimers in double-positive thymocytes. Here we show CD8 variegation is correlated with an epigenetic 'off' state, linking Cd8 enhancer function with chromatin remodeling of the adjacent genes Cd8a and Cd8b1 (Cd8). The zinc finger protein MAZR bound the Cd8 enhancer and interacted with the nuclear receptor corepressor N-CoR complex in double-negative thymocytes. MAZR was downregulated in double-positive and CD8 single-positive thymocytes. 'Enforced' expression of MAZR led to impaired Cd8 activation and variegated CD8 expression. Our results demonstrate epigenetic control of the Cd8 loci and identify MAZR as an important regulator of Cd8 expression.
Collapse
Affiliation(s)
- Ivan Bilic
- Institute of Immunology, Medical University of Vienna, Vienna 1090, Austria
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Xie H, Huang Z, Sadim MS, Sun Z. Stabilized β-Catenin Extends Thymocyte Survival by Up-Regulating Bcl-xL. THE JOURNAL OF IMMUNOLOGY 2005; 175:7981-8. [PMID: 16339534 DOI: 10.4049/jimmunol.175.12.7981] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD4+CD8+ double-positive (DP) thymocytes, which are extremely sensitive to apoptosis, specifically up-regulate Bcl-xL to extend their lifespan. Deletion of the Bcl-xL gene leads to premature apoptosis of the thymocytes. In this study, we show that stabilization of beta-catenin, a critical coactivator for T cell factor (TCF), enhances DP thymocyte survival via up-regulating Bcl-xL. Spontaneous or glucocorticoid-induced thymocyte apoptosis was associated with reduced levels of beta-catenin and Bcl-xL. Transgenic expression of a stabilized beta-catenin protected DP thymocytes from both spontaneous and glucocorticoid-induced apoptosis, resulting in significantly increased thymic cellularity. Compared with the wild-type mice, both protein and transcript levels of Bcl-xL were significantly increased in thymocytes of beta-catenin transgenic mice. In addition, TCF-1 as well as beta-catenin were able to stimulate transcriptional activity of the reporter driven by a Bcl-xL promoter. beta-Catenin/TCF is thus able to act as a signal to up-regulate Bcl-xL levels in DP thymocytes, resulting in their enhanced survival.
Collapse
Affiliation(s)
- Huimin Xie
- Department of Microbiology and Immunology, College of Medicine, University of Illinois, Chicago, IL 60612, USA
| | | | | | | |
Collapse
|
25
|
Abstract
Development is the process whereby a multipotent cell gives rise, through series of divisions, to progeny with successively restricted potentials. During T cell development, the process begins with a multipotent hematopoietic stem cell (HSC) in the bone marrow, moves to the thymus where early T cells or thymocytes pass through signal-initiated developmental checkpoints, and ends in the periphery where mature T cells reside. At each step along this developmental pathway, T lymphocyte progenitors must be able to turn genes on and off, creating a specialized program of gene expression, to allow further development. How is gene expression coordinated? This review will summarize what has been learned about the function of chromatin structure in generating a "blueprint" of gene expression during T cell development. This will include discussion of mechanisms of chromatin remodeling, histone modification, and heritable gene silencing. In many cases, these processes are carried out by multi-protein complexes whose components are largely ubiquitously expressed. The spatial and temporal specificity of these complexes is contributed by sequence specific DNA binding factors, some of which are cell type restricted in their expression. This review will summarize research underway to identify these key genetic "targeters." Taken together, the research reviewed here provides a glimpse into the importance of regulation of chromatin structure in T cell development and the "players" involved.
Collapse
Affiliation(s)
- Susan Winandy
- Department of Microbiology-Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.
| |
Collapse
|
26
|
Bosselut R. CD4/CD8-lineage differentiation in the thymus: from nuclear effectors to membrane signals. Nat Rev Immunol 2004; 4:529-40. [PMID: 15229472 DOI: 10.1038/nri1392] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Affiliation(s)
- Rémy Bosselut
- Laboratory of Immune Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA.
| |
Collapse
|
27
|
Williams CJ, Naito T, Arco PGD, Seavitt JR, Cashman SM, De Souza B, Qi X, Keables P, Von Andrian UH, Georgopoulos K. The chromatin remodeler Mi-2beta is required for CD4 expression and T cell development. Immunity 2004; 20:719-33. [PMID: 15189737 DOI: 10.1016/j.immuni.2004.05.005] [Citation(s) in RCA: 203] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2003] [Revised: 03/09/2004] [Accepted: 04/14/2004] [Indexed: 11/20/2022]
Abstract
Changes in chromatin structure underlie the activation or silencing of genes during development. The chromatin remodeler Mi-2beta is highly expressed in thymocytes and is presumed to be a transcriptional repressor because of its presence in the nucleosome remodeling deacetylase (NuRD) complex. Using conditional inactivation, we show that Mi-2beta is required at several steps during T cell development: for differentiation of beta selected immature thymocytes, for developmental expression of CD4, and for cell divisions in mature T cells. We further show that Mi-2beta plays a direct role in promoting CD4 gene expression. Mi-2beta associates with the CD4 enhancer as well as the E box binding protein HEB and the histone acetyltransferase (HAT) p300, enabling their recruitment to the CD4 enhancer and causing histone H3-hyperacetylation to this regulatory region. These findings provide important insights into the regulation of CD4 expression during T cell development and define a role for Mi-2beta in gene activation.
Collapse
Affiliation(s)
- Christine J Williams
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Arman M, Calvo J, Trojanowska ME, Cockerill PN, Santana M, López-Cabrera M, Vives J, Lozano F. Transcriptional Regulation of Human CD5: Important Role of Ets Transcription Factors in CD5 Expression in T Cells. THE JOURNAL OF IMMUNOLOGY 2004; 172:7519-29. [PMID: 15187131 DOI: 10.4049/jimmunol.172.12.7519] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CD5 is a surface receptor constitutively expressed on thymocytes and mature T and B-1a cells. CD5 expression is tightly regulated during T and B cell development and activation processes. In this study we shown that the constitutive expression of CD5 on human T cells correlates with the presence of a DNase I-hypersensitive (DH) site at the 5'-flanking region of CD5. Human CD5 is a TATA-less gene for which 5'-RACE analysis shows multiple transcriptional start sites, the most frequent of which locates within an initiator sequence. Luciferase reporter assays indicate that a 282-bp region upstream of the initiation ATG displays full promoter activity in human T cells. Two conserved Ets-binding sites (at positions -239 and -185) were identified as functionally relevant to CD5 expression by site-directed mutagenesis, EMSAs, and cotransfection experiments. A possible contribution of Sp1 (-115 and -95), c-Myb (-177), and AP-1-like (-151) motifs was also detected. Further DH site analyses revealed an inducible DH site 10 kb upstream of the human CD5 gene in both T and B CD5(+) cells. Interestingly, a 140-bp sequence showing high homology with a murine inducible enhancer is found within that site. The data presented indicate that the 5'-flanking region of human CD5 is transcriptionally active in T cells, and that Ets transcription factors in conjunction with other regulatory elements are responsible for constitutive and tissue-specific CD5 expression.
Collapse
Affiliation(s)
- Mònica Arman
- Servei d'Immunologia, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Hospital Clínic i Provincial de Barcelona, Villaroel 170, Barcelona 08036, Spain
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Taniuchi I, Ellmeier W, Littman DR. The CD4/CD8 lineage choice: new insights into epigenetic regulation during T cell development. Adv Immunol 2004; 83:55-89. [PMID: 15135628 DOI: 10.1016/s0065-2776(04)83002-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
von Boehmer H. Selection of the T-Cell Repertoire: Receptor-Controlled Checkpoints in T-Cell Development. Adv Immunol 2004; 84:201-38. [PMID: 15246254 DOI: 10.1016/s0065-2776(04)84006-9] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- Harald von Boehmer
- Harvard Medical School, Dana-Farber Cancer Institute, Boston, Massachusetts USA
| |
Collapse
|
31
|
Pierce M, Wang C, Stump M, Kamb A. Overexpression of the beta-catenin binding domain of cadherin selectively kills colorectal cancer cells. Int J Cancer 2003; 107:229-37. [PMID: 12949799 DOI: 10.1002/ijc.11372] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The beta-catenin pathway is involved in growth, differentiation and tumor formation. Suppression of pathway activity by expressed inhibitors can cause growth arrest or apoptosis in certain colon carcinoma lines. We compare the effects of 2 pathway inhibitors, a VE-cadherin cytoplasmic domain fragment (Cad5CD) and a truncated, dominant-negative Tcf4 (TcfDN), using a microplate assay for cell death and microarray gene expression analysis. The cell-lethal assay shows that Cad5CD, when expressed in HT29 human colon tumor cells and 3 non-colon lines, selectively kills the HT29 cells. Cad5CD overexpression inhibits beta-catenin/Tcf4 transcriptional activity, as determined by results from microarray experiments. Our results support the view that beta-catenin is an attractive anti-cancer target, especially if the Cad5CD binding site or Cad5CD itself can be exploited for drug development. In addition, therapeutically relevant phenotypes such as drug selectivity may be difficult to predict from gene expression analysis alone. Other more specialized phenotypic tests such as cell-lethal assays may be required.
Collapse
|
32
|
Dillon TJ, Karpitski V, Wetzel SA, Parker DC, Shaw AS, Stork PJS. Ectopic B-Raf expression enhances extracellular signal-regulated kinase (ERK) signaling in T cells and prevents antigen-presenting cell-induced anergy. J Biol Chem 2003; 278:35940-9. [PMID: 12855697 DOI: 10.1074/jbc.m301506200] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
T cells that receive stimulation through the T cell receptor (TCR) in the absence of costimulation become anergic and are refractory to subsequent costimulation. This unresponsiveness is associated with the constitutive activation of the small G protein, Rap1, and the lack of Ras-dependent activation of ERK. Recent studies suggest that Rap1 can activate the MAP kinase kinase kinase B-Raf that is either endogenously or ectopically expressed. Peripheral T cells generally do not express B-Raf; therefore, to test the hypothesis that ectopic expression of B-Raf could permit Rap1 to activate ERK signaling, we generated transgenic mice expressing B-Raf within peripheral T cells. This converted Rap1 into an activator of ERK, to enhance ERK activation and proliferation following TCR engagement in the absence of costimulation. When T cells were incubated with engineered APCs presenting antigen on I-Ek and expressing low levels of B7, they became anergic, displayed constitutive activation of Rap1, and were deficient in Ras and ERK activation. However, when incubated with the same APCs, T cells expressing the B-Raf transgene proliferated upon restimulation and displayed elevated ERK activation. Thus B-Raf expression and enhanced ERK activation is sufficient to prevent anergy in a model of APC-induced T cell anergy. However, studies using anti-TCR antibody-induced anergy showed that the ability of ERKs to reverse T cell anergy is dependent on the anergic model utilized.
Collapse
MESH Headings
- Animals
- Antigen-Presenting Cells/metabolism
- Antigens, CD/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- Blotting, Western
- Cell Division
- Cell Nucleus/metabolism
- DNA-Binding Proteins
- Dose-Response Relationship, Drug
- Enzyme Activation
- Fibroblasts/metabolism
- Flow Cytometry
- Humans
- Jurkat Cells
- Lectins, C-Type
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Mitogen-Activated Protein Kinases/metabolism
- Models, Biological
- Peptides/chemistry
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins B-raf
- Proto-Oncogene Proteins c-raf/biosynthesis
- Proto-Oncogene Proteins c-raf/metabolism
- Receptors, Antigen, T-Cell/metabolism
- Signal Transduction
- T-Lymphocytes/cytology
- T-Lymphocytes/enzymology
- T-Lymphocytes/metabolism
- Tissue Distribution
- Transcription Factors
- ets-Domain Protein Elk-1
- rap1 GTP-Binding Proteins/metabolism
Collapse
Affiliation(s)
- Tara J Dillon
- Vollum Institute, Oregon Health Sciences University, Portland, Oregon 97239, USA
| | | | | | | | | | | |
Collapse
|
33
|
Kioussis D, Ellmeier W. Chromatin and CD4, CD8A and CD8B gene expression during thymic differentiation. Nat Rev Immunol 2002; 2:909-19. [PMID: 12461564 DOI: 10.1038/nri952] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The regulation of gene expression during thymocyte development provides an ideal experimental system to study lineage-commitment processes. In particular, expression of the CD4, CD8A and CD8B genes seems to correlate well with the cell-fate decisions that are taken by thymocytes, and elucidating the molecular mechanisms that underlie the differential expression of these genes could reveal key events in differentiation processes. Here, we review examples of how gene cis elements (such as promoters, enhancers and locus control regions) and trans elements (such as transcription factors, chromatin-remodelling complexes and histone-modification enzymes) come together to orchestrate a finely tuned sequence of events that results in the complex pattern of CD4, CD8A and CD8B gene expression that is observed during thymocyte development.
Collapse
Affiliation(s)
- Dimitris Kioussis
- Division of Molecular Immunology, National Institute for Medical Research, The Ridgeway, London NW7 1AA, UK.
| | | |
Collapse
|
34
|
Abstract
T cell lineage commitment as the double-positive (DP) thymocyte matures into the single-positive (SP) T cell requires the irreversible repression or maintenance of CD4 gene expression. Signals transmitted from the T cell antigen receptor (TCR) during thymic selection are believed to be linked to the transcriptional regulation of the CD4 gene; thus, a study of the factors that control CD4 gene expression may lead to further insight into the molecular mechanisms that drive T cell development. This review discusses the work conducted to date to identify and characterize the transcriptional control elements in the CD4 locus and the factors that mediate their function. From these studies, it is clear that the molecular mechanisms controlling CD4 gene expression are very complex and are controlled by many different signals as the thymocyte develops.
Collapse
Affiliation(s)
- Gerald Siu
- Department of Microbiology, College of Physicians and Surgeons, Columbia University, 701 West 168th Street, New York, NY 10032, USA.
| |
Collapse
|
35
|
Taniuchi I, Osato M, Egawa T, Sunshine MJ, Bae SC, Komori T, Ito Y, Littman DR. Differential requirements for Runx proteins in CD4 repression and epigenetic silencing during T lymphocyte development. Cell 2002; 111:621-33. [PMID: 12464175 DOI: 10.1016/s0092-8674(02)01111-x] [Citation(s) in RCA: 610] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
T lymphocytes differentiate in discrete stages within the thymus. Immature thymocytes lacking CD4 and CD8 coreceptors differentiate into double-positive cells (CD4(+)CD8(+)), which are selected to become either CD4(+)CD8(-)helper cells or CD4(-)CD8(+) cytotoxic cells. A stage-specific transcriptional silencer regulates expression of CD4 in both immature and CD4(-)CD8(+) thymocytes. We show here that binding sites for Runt domain transcription factors are essential for CD4 silencer function at both stages, and that different Runx family members are required to fulfill unique functions at each stage. Runx1 is required for active repression in CD4(-)CD8(-) thymocytes whereas Runx3 is required for establishing epigenetic silencing in cytotoxic lineage thymocytes. Runx3-deficient cytotoxic T cells, but not helper cells, have defective responses to antigen, suggesting that Runx proteins have critical functions in lineage specification and homeostasis of CD8-lineage T lymphocytes.
Collapse
Affiliation(s)
- Ichiro Taniuchi
- Howard Hughes Medical Institute, Molecular Pathogenesis Program, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Taniuchi I, Sunshine MJ, Festenstein R, Littman DR. Evidence for distinct CD4 silencer functions at different stages of thymocyte differentiation. Mol Cell 2002; 10:1083-96. [PMID: 12453416 DOI: 10.1016/s1097-2765(02)00735-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
An intronic silencer within the CD4 gene is the critical cis regulatory element for T cell subset-specific expression of CD4. We have combined transfection studies with gene targeting in mice to identify several key sequences within the silencer core that are required for gene silencing during thymocyte development. In mice, mutations in individual sites resulted in variegated, but heritable, derepression of CD4 in mature CD8(+) T lymphocytes, whereas compound mutations resulted in full derepression. These results indicate that there is partial redundancy in recruiting a chromatin remodeling machinery that results in epigenetic silencing. Mutations in single sites also resulted in partial derepression of CD4 in immature double-negative thymocytes, but there was no apparent variegation. These findings suggest two distinct modes of CD4 silencer function at different developmental stages: active repression in CD4(-)CD8(-) thymocytes, in which silencing must be reversible, and epigenetic gene silencing upon differentiation to the CD8(+) cytotoxic T cell lineage.
Collapse
Affiliation(s)
- Ichiro Taniuchi
- Howard Hughes Medical Institute, Molecular Pathogenesis Program, Skirball Institute of Biomolecular Medicine, New York University School of Medicine, 540 First Avenue, New York, NY 10016, USA.
| | | | | | | |
Collapse
|
37
|
Jin ZX, Kishi H, Wei XC, Matsuda T, Saito S, Muraguchi A. Lymphoid Enhancer-Binding Factor-1 Binds and Activates the Recombination-Activating Gene-2 Promoter Together with c-Myb and Pax-5 in Immature B Cells. THE JOURNAL OF IMMUNOLOGY 2002; 169:3783-92. [PMID: 12244173 DOI: 10.4049/jimmunol.169.7.3783] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The recombination-activating gene (RAG)-1 and RAG-2 are expressed specifically in immature lymphoid cells undergoing the recombination of Ag receptor genes. We studied the regulation of murine RAG-2 promoter and revealed that -41/-17 RAG-2 promoter region, which was indispensable for the RAG-2 promoter activity in B cell lines, contained binding sites for lymphoid enhancer-binding factor-1 (LEF-1), c-Myb, and Pax-5. We showed that these three transcription factors bound the promoter region in vitro and in vivo. Cotransfection assays using a human embryonic kidney cell line (293T) showed that LEF-1, c-Myb, and Pax-5 cooperatively activated the RAG-2 promoter, via their synergistic DNA binding. We also showed that LEF-1, c-Myb, and Pax-5 physically interact in the cells. Finally, we demonstrated that a dominant-negative LEF-1 protein, which lacks the binding site for beta-catenin, suppressed the RAG-2 promoter activity as well as the endogenous RAG-2 expression in a pre-B cell line (18.81). These results suggest that LEF-1/beta-catenin complex regulates the RAG-2 promoter activation in concert with c-Myb and Pax-5 in immature B cells. The link between LEF-1/beta-catenin and Wnt signaling in B lineage cells will be discussed.
Collapse
Affiliation(s)
- Zhe-Xiong Jin
- Department of Immunology, Faculty of Medicine, Toyama Medical and Pharmaceutical University, Sugitani, Toyama, Japan
| | | | | | | | | | | |
Collapse
|
38
|
Abstract
Antigen-specific responses by CD8+ T cells require direct cell-cell interactions between T cells and antigen-presenting cells (APC). Initially, naïve T cells must communicate with APC in lymphoid organs. Once stimulated, the resulting effector cells interact with APC in peripheral tissues. To this end, T cells must migrate to discrete sites throughout the body where antigen may be found. Recent progress in the field has revealed that the migratory abilities of T cells are critically dependent on their differentiation state, which is shaped by a multitude of factors. Thus, naïve T cells are normally restricted to recirculate between the blood and secondary lymphoid tissues, although in some autoimmune diseases they may also accumulate in chronically inflamed tissues. When CD8+ T cells encounter antigen and differentiate into short-lived effector CTL, they lose the ability to home to lymph nodes but gain access to peripheral tissues and sites of inflammation. Long-lived memory cells exist in (at least) two flavors: central memory cells that migrate to both lymphoid organs and peripheral sites of inflammation, and effector memory cells that are preferentially localized in non-lymphoid tissues. Our current understanding of the interplay of T cell differentiation and migration has been boosted by the development of T-GFP mice, in which transgenic green fluorescent protein is expressed selectively in naïve and central memory T cells, but not in effector cytotoxic T cells (CTL). This review will focus on recent studies in which T-GFP mice were used to dissect the traffic signals for naïve T cell homing to secondary lymphoid organs, the factors that influence the differentiation of naïve CD8+ T cells into cytotoxic and memory cells, as well as the in vivo trafficking routes of antigen-experienced subsets.
Collapse
Affiliation(s)
- Wolfgang Weninger
- The Center for Blood Research and Department of Pathology, Harvard Medical School, 200 Longwood Avenue, Boston, MA 02115, USA
| | | | | |
Collapse
|
39
|
Abstract
An important but laborious approach to understanding the concepts underlying T cell lineage commitment is to characterize the cis-acting control elements governing the expression of CD4 and CD8. Previous studies on the CD4 gene have shown that lineage commitment information is directed by the intronic silencer; however, a similarly simple mechanism for controlling CD8 gene expression has not been uncovered. In this issue of Immunity, two groups have investigated the role of putative enhancers in the CD8 locus. The deletion of three different elements in the intergeneic region between CD8 beta and CD8 alpha provides evidence for control at the level of chromatin accessibility.
Collapse
Affiliation(s)
- Stephen M Hedrick
- Department of Biology and The Cancer Center, University of California, San Diego, La Jolla, CA 92093, USA.
| |
Collapse
|
40
|
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
|
41
|
Ellmeier W, Sunshine MJ, Maschek R, Littman DR. Combined deletion of CD8 locus cis-regulatory elements affects initiation but not maintenance of CD8 expression. Immunity 2002; 16:623-34. [PMID: 12049715 DOI: 10.1016/s1074-7613(02)00309-6] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Developmental stage-, subset-, and lineage-specific CD8 enhancers have been identified recently by transgenic reporter analyses. Enhancer E8(II) (CIV-4,5) is active in both immature double-positive thymocytes (DP) and mature CD8 single-positive (SP) thymocytes and T cells, whereas E8(I) (CIII-1,2) directs expression only in mature cells. In mice lacking either E8(I) (CIII-1,2) or E8(II) (CIV-4,5), there was no effect on CD8 expression in DP thymocytes. However, deletion of both enhancers resulted in variegated expression of CD8, with appearance of CD4(+)CD8(-) SP thymocytes expressing surface markers characteristic of DP thymocytes. Consequently, fewer mature CD8(+) T cells developed from the reduced pool of DP cells. These results suggest that the initiation of CD8 expression is mediated by cis-regulatory elements that are distinct from any that may be involved in maintenance of expression.
Collapse
|
42
|
Hanna Z, Rebai N, Poudrier J, Jolicoeur P. Distinct regulatory elements are required for faithful expression of human CD4 in T cells, macrophages, and dendritic cells of transgenic mice. Blood 2001; 98:2275-8. [PMID: 11568018 DOI: 10.1182/blood.v98.7.2275] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To identify the regulatory elements controlling expression of the human CD4 (hCD4) gene in different cell types of the immune system, deletion and chimeric (human/murine) reporter genes were constructed and tested in transgenic (Tg) mice. Regulatory elements required for the proper hCD4 expression in the immature double-positive thymic T cells were identified in the enhancer and in the 3' end of intron 1. Expression of hCD4 in macrophages is controlled by at least 2 sets of regulatory elements: one present in front of exon 1 and the second at the 5' end of intron 1. The hCD4 elements required for expression on both myeloid and lymphoid CD8alpha(+) dendritic cells (DCs) from lymph node and thymus were found to be different from those required for macrophage expression. The results indicate that expression of hCD4 in T cells, macrophages, and DCs is controlled by distinct regulatory elements.
Collapse
Affiliation(s)
- Z Hanna
- Laboratory of Molecular Biology, Clinical Research Institute of Montreal, Montreal, Quebec, Canada.
| | | | | | | |
Collapse
|
43
|
Abstract
The Complement Receptor Type 2 (Cr2-145,CR2, CD21) is an important receptor in the innate and acquired immune response. CD21 is produced by B cells and follicular dendritic cells, where it binds cleavage products of the C3 complement protein. CD21 facilitates internalization of immune complexes by B cells to enhance antigen presentation. CD21, in association with CD19/CD81, also serves as a coaccessory activation complex with the B-cell antigen receptor, permitting a lower antigen concentration to achieve maximal B-cell activation. CD21 traps immune complexes on the surface of follicular dendritic cells and displays them to activated B cells in germinal centers. Much work has been conducted to determine the transcriptional control mechanisms dictating CD21 expression. Appropriate transcriptional control of the CD21 gene evidently requires the CD21 promoter, as well as intronic sequences with enhancer and suppressor functions. Chromatin structure has been implicated in regulating the coordination of CD21 promoter and intronic control sequences by regulating access to them by putative transcription factors. This review assesses the past and current research into CD21 transcriptional regulation and offers insight into future experimental directions.
Collapse
Affiliation(s)
- M D Zabel
- Division of Cell Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA.
| | | |
Collapse
|
44
|
Matsunaga S, Mukai R, Inoue-Murayama M, Yoshikawa Y, Murayama Y. Sequence and functional properties of African green monkey CD4 silencer. Immunol Lett 2000; 75:47-53. [PMID: 11163866 DOI: 10.1016/s0165-2478(00)00273-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We reported previously that in African green monkey (AGM) CD4 lymphocytes, CD4 mRNA expression undergoes a decrease following in vitro activation, and CD4 cells are therefore subject to loss of CD4 expression on the cell surface. To examine the transcriptional regulation of the CD4 gene in this species. we analyzed the CD4 silencer, which has been identified as a regulatory element responsible for the down regulation of CD4 transcription in CD8 cell lineage cells. Sequence analysis indicated that the CD4 silencer of the AGM was highly homologous to that of humans. However, two nucleotide substitutions were present in one of the nuclear protein binding sites, which was characterized as the FP II site having a strong enhancing effect on transgene expression in CD4 cells. By performing transient transfection assays. we found that the enhancing activities of the CD4 silencer or FP II-containing fragment of the AGM were greatly reduced in a human CD4 cell line as compared to those of human materials. The CD4 mRNA level was significantly decreased in the human CD4 cell line when synthetic oligonucleotide corresponding to the human FP II sequence was added to the culture. These observations imply that FP II-protein interaction might be required for the maintenance of sufficient expression of the CD4 gene, and the enhancing activity mediated by the above interaction might be decreased in the AGM CD4 silencer, due probably to the nucleotide changes occurring at the FP II site.
Collapse
Affiliation(s)
- S Matsunaga
- Institute of Basic Medical Sciences, University of Tsukuba, Ibaraki, Japan
| | | | | | | | | |
Collapse
|
45
|
Zhao-Emonet JC, Marodon G, Pioche-Durieu C, Cosset FL, Klatzmann D. T cell-specific expression from Mo-MLV retroviral vectors containing a CD4 mini-promoter/enhancer. J Gene Med 2000; 2:416-25. [PMID: 11199262 DOI: 10.1002/1521-2254(200011/12)2:6<416::aid-jgm142>3.0.co;2-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
BACKGROUND Gene therapy of various immunological disorders will greatly benefit from improved retroviral vectors (RVs) with T cell specificity. Such vectors can be designed by placing a gene of therapeutic interest under the control of tissue-specific transcriptional elements. However, low titers and loss of specificity are frequently encountered with tissue-specific vectors. The aim of the present study was to develop a T cell-specific RV. METHODS We constructed a series of Moloney murine leukemia virus (Mo-MLV)-based RVs expressing enhanced green fluorescent protein (EGFP) under the control of a mini-promoter/enhancer cassette derived from the CD4 gene (CD4pmE) and tested them in cell lines and peripheral blood lymphocytes. Expression of EGFP was monitored by fluorescence microscopy and analyzed by flow cytometry. RESULTS The CD4pmE cassette was inserted between the viral long terminal repeats (LTRs) in self-inactivating vectors (SIN vectors) or was substituted to the 3' U3 viral promoter/enhancer (hybrid vectors). High vector titers but poor specific expression of EGFP were achieved when CD4pmE was inserted in sense orientation in SIN vectors. Low titers but high specificity were observed when the CD4pmE cassette was in anti-sense orientation. In contrast, high titers and good T cell specificity were obtained with hybrid vectors. CONCLUSION An efficient T cell-specific retroviral vector was obtained.
Collapse
Affiliation(s)
- J C Zhao-Emonet
- Laboratoire de Biologie et Thérapeutique des Pathologies Immunitaires UPMC-CNRS ESA 7087-CERVI-H pital de la Pitié, Paris, France
| | | | | | | | | |
Collapse
|
46
|
Zabel MD, Byrne BL, Weis JJ, Weis JH. Cell-specific expression of the murine CD21 gene depends on accessibility of promoter and intronic elements. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:4437-45. [PMID: 11035082 DOI: 10.4049/jimmunol.165.8.4437] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The murine complement receptor type 2 gene (Cr2/CD21) is transcriptionally active in murine B and follicular dendritic cells, but not in murine T cells. We have previously shown that altering chromatin structure via histone deacetylase inhibitors results in CD21 expression in murine T cells, and that the minimal CD21 promoter provided appropriate cell-specific expression of luciferase reporter constructs only in the presence of the first third of intron 1, fragment A. We extend this work by showing that replacing the CD21 gene promoter with the SV40 promoter resulted in the loss of this cell-specific control. Further delineation of intronic regulatory elements by fragmentation also resulted in the loss of cell-specific gene expression, suggesting that multiple CD21 promoter and intronic elements interact for appropriate CD21 gene expression. To assess this model, we performed EMSAs to define protein binding sites within promoter and intronic regions and DNase I hypersensitivity assays to determine chromatin accessibility. Multiple DNA binding factors were shown to be present in B and T cell extracts; a minority demonstrated B cell specificity. However, the DNase I sensitivity of T cell CD21 regulatory elements was not comparable to that of B cells until the histone acetylation status of the gene was altered. Taken together, these data suggest that chromatin remodeling facilitates cell-specific CD21 gene expression by modulating access of transcription factors to regulatory elements in the promoter and intron.
Collapse
Affiliation(s)
- M D Zabel
- Division of Cell Biology and Immunology, Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
| | | | | | | |
Collapse
|
47
|
Indraccolo S, Minuzzo S, Habeler W, Zamarchi R, Fregonese A, Günzburg WH, Salmons B, Uckert W, Chieco-Bianchi L, Amadori A. Modulation of Moloney leukemia virus long terminal repeat transcriptional activity by the murine CD4 silencer in retroviral vectors. Virology 2000; 276:83-92. [PMID: 11021997 DOI: 10.1006/viro.2000.0519] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We investigated whether CD4 gene regulatory sequences might be useful for developing transcriptionally targeted Moloney murine leukemia virus (Mo-MLV)-based retroviral vectors for gene expression specifically in CD4(+) cells. We could modulate Mo-MLV long terminal repeat (LTR) activity by inserting a 438-bp-long fragment containing the murine CD4 silencer in the LTR of the vector; both beta-galactosidase and green fluorescent protein reporter gene activities were strongly down-regulated in both murine and human CD8(+) cells, but not in CD4(+) lymphoid cell lines and freshly isolated lymphocytes transduced with this vector, compared with the findings using a control vector carrying wild-type LTRs. Titration experiments on NIH-3T3 cells revealed that inclusion of the CD4 silencer in the LTRs did not reduce the titer of the vectors. These findings indicate that a cellular silencer can be successfully included in retroviral vectors, where it maintains its transcription-regulatory function, thus suggesting a novel approach to transcriptional targeting.
Collapse
|
48
|
Leenders H, Whiffield S, Benoist C, Mathis D. Role of the forkhead transcription family member, FKHR, in thymocyte differentiation. Eur J Immunol 2000; 30:2980-90. [PMID: 11069081 DOI: 10.1002/1521-4141(200010)30:10<2980::aid-immu2980>3.0.co;2-9] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
While performing a large-scale analysis of mRNA transcripts in the murine thymus, our attention was drawn to the forkhead family transcription factor FKHR. Here we demonstrate that FKHR is expressed in thymocytes, most prominently in those that are undergoing positive selection. Interestingly, FKHR transcripts show a highly regionalized pattern of expression, concentrated in the innermost areas of the medulla. We define the FKHR binding site as (G/C)(A/C)N(G/a)T(A/c)AA(T/c) A(T/g)(T/g)(G/c), a sequence found in the regulatory elements of many genes, including certain that encode molecules crucial for thymocyte differentiation. To study the function of FKHR, we engineered mice expressing a dominant-negative mutant specifically in T cells in a tetracycline-regulatable fashion. In these animals, T cell differentiation appeared quite normal; however, total thymocyte numbers were decreased, owing to reductions in all four of the CD4/CD8 subsets, and incorporation of the thymidine analogue bromo-deoxyuridine was increased, again in all four subsets. These data suggest that, in thymocytes, FKHR may be involved in cell survival and/or cycling.
Collapse
Affiliation(s)
- H Leenders
- Institut de Genetique et de Biologie Moleculaire et Cellulaire (CNRS/INSERM/ULP), Ilkirch, France
| | | | | | | |
Collapse
|
49
|
Thomas-Vaslin V, Bellier B, Cohen JL, Boyer O, Raynal-Raschilas N, Glotz D, Klatzmann D. Prolonged allograft survival through conditional and specific ablation of alloreactive T cells expressing a suicide gene. Transplantation 2000; 69:2154-61. [PMID: 10852616 DOI: 10.1097/00007890-200005270-00034] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
BACKGROUND Control of antidonor activated T cells involved in allograft rejection while preserving immunocompetence is a challenging goal in transplantation. Engineered T cells expressing a viral thymidine kinase (TK) suicide gene metabolize the nontoxic prodrug ganciclovir (GCV) into a metabolite toxic only to dividing cells. We evaluated this suicide gene strategy for inducing transplantation tolerance in mice. METHODS Transgenic mice expressing TK in mature T cells were analyzed for (i) specific T-cell depletion under GCV treatment upon various stimulations; (ii) outcome of allogeneic nonvascularized skin or heart allografts under a short 14-day GCV treatment initiated at the time of transplantation; and (iii) the capacities of T cells from such allotransplanted mice to proliferate in mixed lymphocyte reactions and to induce graft-versus-host disease in irradiated recipients with the genetic background of the donor allograft. RESULTS Upon in vitro or in vivo GCV treatment, only activated dividing TK T cells but not B cells were efficiently depleted. Acute rejection of allogeneic grafts was prevented and a significant prolongation of graft survival was obtained, although associated with signs of chronic rejection. Prolonged skin graft survival correlated with decreased in vitro and in vivo T-cell reactivities against donor alloantigens, whereas overall immunocompetence was preserved. CONCLUSIONS Efficient and specific depletion of alloreactive TK T cells can be achieved by administrating GCV. These results open new perspectives for the control of allogeneic graft rejection using suicide gene therapy.
Collapse
Affiliation(s)
- V Thomas-Vaslin
- Biologie et Thérapeutique des Pathologies Immunitaires, UPMC/CNRS ESA 7087, Groupe Hospitalier Pitié-Salpêtrière, Paris, France
| | | | | | | | | | | | | |
Collapse
|
50
|
Diehl NL, Enslen H, Fortner KA, Merritt C, Stetson N, Charland C, Flavell RA, Davis RJ, Rincón M. Activation of the p38 mitogen-activated protein kinase pathway arrests cell cycle progression and differentiation of immature thymocytes in vivo. J Exp Med 2000; 191:321-34. [PMID: 10637276 PMCID: PMC2195760 DOI: 10.1084/jem.191.2.321] [Citation(s) in RCA: 78] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/1999] [Accepted: 11/02/1999] [Indexed: 11/25/2022] Open
Abstract
The development of T cells in the thymus is coordinated by cell-specific gene expression programs that involve multiple transcription factors and signaling pathways. Here, we show that the p38 mitogen-activated protein (MAP) kinase signaling pathway is strictly regulated during the differentiation of CD4(-)CD8(-) thymocytes. Persistent activation of p38 MAP kinase blocks fetal thymocyte development at the CD25(+)CD44(-) stage in vivo, and results in the lack of T cells in the peripheral immune system of adult mice. Inactivation of p38 MAP kinase is required for further differentiation of these cells into CD4(+)CD8(+) thymocytes. The arrest of cell cycle in mitosis is partially responsible for the blockade of differentiation. Therefore, the p38 MAP kinase pathway is a critical regulatory element of differentiation and proliferation during the early stages of in vivo thymocyte development.
Collapse
Affiliation(s)
- Nicole L. Diehl
- Immunobiology Program, Department of Medicine, University of Vermont, Burlington, Vermont 05405
| | - Hervé Enslen
- Program in Molecular Medicine, Department of Biochemistry and Molecular Biology, University of Massachusetts Medical School
- Howard Hughes Medical Institute, Worcester, Massachusetts 01605
| | - Karen A. Fortner
- Immunobiology Program, Department of Medicine, University of Vermont, Burlington, Vermont 05405
| | - Chris Merritt
- Immunobiology Program, Department of Medicine, University of Vermont, Burlington, Vermont 05405
| | - Nate Stetson
- Immunobiology Program, Department of Medicine, University of Vermont, Burlington, Vermont 05405
| | - Colette Charland
- Immunobiology Program, Department of Medicine, University of Vermont, Burlington, Vermont 05405
| | - Richard A. Flavell
- Section of Immunobiology, Yale University School of Medicine
- Howard Hughes Medical Institute, New Haven Connecticut 06520
| | - Roger J. Davis
- Program in Molecular Medicine, Department of Biochemistry and Molecular Biology, University of Massachusetts Medical School
- Howard Hughes Medical Institute, Worcester, Massachusetts 01605
| | - Mercedes Rincón
- Immunobiology Program, Department of Medicine, University of Vermont, Burlington, Vermont 05405
| |
Collapse
|